US20110076273A1 - Highly Concentrated Pharmaceutical Formulations - Google Patents

Highly Concentrated Pharmaceutical Formulations Download PDF

Info

Publication number
US20110076273A1
US20110076273A1 US12/879,486 US87948610A US2011076273A1 US 20110076273 A1 US20110076273 A1 US 20110076273A1 US 87948610 A US87948610 A US 87948610A US 2011076273 A1 US2011076273 A1 US 2011076273A1
Authority
US
United States
Prior art keywords
antibody
weeks
highly concentrated
stable pharmaceutical
formulation
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
US12/879,486
Other languages
English (en)
Inventor
Michael Adler
Hanns-Christian Mahler
Oliver Boris Stauch
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.)
Genentech Inc
Original Assignee
Genentech 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=43732866&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110076273(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Genentech Inc filed Critical Genentech Inc
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ADLER, MICHAEL, MAHLER, HANNS-CHRISTIAN, STAUCH, OLIVER BORIS
Assigned to GENENTECH, INC. reassignment GENENTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Publication of US20110076273A1 publication Critical patent/US20110076273A1/en
Priority to US14/260,558 priority Critical patent/US10280227B2/en
Priority to US14/944,508 priority patent/US10377831B2/en
Priority to US16/450,906 priority patent/US10752696B2/en
Priority to US16/943,378 priority patent/US20210054092A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • A61K9/0021Intradermal administration, e.g. through microneedle arrays, needleless injectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to highly concentrated, stable pharmaceutical formulations of a pharmaceutically active anti-CD20 antibody or a mixture of such antibody molecules for subcutaneous injection.
  • Such formulations comprise, in addition to the high amounts of anti-CD20 antibody or mixture thereof, a buffering agent, a stabilizer or a mixture of two ore more stabilizing agents, a nonionic surfactant and an effective amount of at least one hyaluronidase enzyme.
  • the invention also relates to a process for the preparation of the formulation and to the uses of such formulation.
  • glycosaminoglycanase enzyme(s) in order to increase the interstitial space into which the antibody formulation can be injected (WO2006/091871).
  • RITUXAN®/MABTHERA® is a chimeric antibody which binds to the CD20 antigen on B-cells.
  • the commercial formulation is a sterile, clear, colorless, preservative-free liquid concentrate for intravenous (IV) administration.
  • Rituximab is supplied at a concentration of 10 mg/mL (10 mL) in either 100 mg or 500 mg (50 mL) single-use vials.
  • the product is formulated in 9 mg/mL sodium chloride, 7.35 mg/mL sodium citrate dehydrate, 0.7 mg/mL polysorbate 80, and Water for Injection. The pH is adjusted to 6.5.
  • An alternative liquid formulation for Rituximab suitable for IV administration is disclosed in U.S. Pat. No. 6,991,790.
  • HERCEPTIN® (Trastuzumab) is a monoclonal antibody directed against the HER2 receptor (anti-HER2) which is currently marketed in Europe in form of a 150 mg lyophilized powder (containing the antibody, ⁇ , ⁇ -trehalose dihydrate, L-histidine and L-histidine hydrochloride and polysorbate 20) which should be reconstituted for infusions with water for injection to yield injection dose of approximately 21 mg/ml. In the USA and many other countries a multiple dosage vial containing 440 mg Trastuzumab is marketed.
  • AVASTIN® (Bevacizumab) is a monoclonal antibody directed against the vascular endothelial growth factor (VEGF) which is currently marketed in Europe as a liquid formulation in two types of vials: a) 100 mg Bevacizumab in 4 ml and b) 400 mg Bevacizumab in 16 ml, respectively, providing a final concentration of 25 mg/ml in water for injection containing the following excipients: trehalose dihydrate, sodium phosphate and polysorbate 20.
  • VEGF vascular endothelial growth factor
  • HUMIRA® (Adalimumab) is a monoclonal antibody directed against tumor necrosis factor alpha (TNF alpha) which is currently marketed in Europe in form of a 40 mg dose in 0.8 ml injection volume for subcutaneous application (concentration: 50 mg antibody/ml injection volume).
  • XOLAIR® (Omalizumab) a monoclonal antibody directed against immunoglobulin E (anti-IgE) which is currently marketed in form of a 150 mg lyophilized powder (containing the antibody, sucrose, histidine and histidine hydrochloride monohydrate and polysorbate 20) which should be reconstituted with water for subcutaneous injection to yield a 125 mg/ml injection dose.
  • parenteral drugs into the hypodermis is generally limited to volumes of less than 2 ml due to this viscoelastic resistance to hydraulic conductance in the subcutaneous (SC) tissue and generated backpressure upon injection (Aukland K. and Reed R., “Interstitial-Lymphatic Mechanisms in the control of Extracellular Fluid Volume”, Physiology Reviews”, 73:1-78 (1993)) as well as due to the perceptions of pain.
  • SC subcutaneous
  • antibodies have a very similar overall structure, such antibodies differ in the amino acid composition (in particular in the CDR regions responsible for the binding to the antigen) and the glycosylation pattern. Moreover there may additionally be post-translational modifications such as charge and glycosylation variants.
  • the present invention provides a highly concentrated, stable pharmaceutical formulation of a pharmaceutically active anti-CD20 antibody or a mixture of such antibody molecules, preferably for subcutaneous injection.
  • a pharmaceutically active anti-CD20 antibody formulation of the present invention comprises:
  • antibody herein is used in the broadest sense and specifically covers full length antibodies, genetically engineered antibodies like monoclonal antibodies, or recombinant antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two full length antibodies, chimeric antibodies, humanized antibodies, fully human antibodies, and as well as fragments of such antibodies as long as they exhibit the desired biological activity.
  • multispecific antibodies e.g. bispecific antibodies
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they are uncontaminated by other immunoglobulins.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Köhler et al, Nature, 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clarkson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol.
  • the terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of a single amino acid composition. Accordingly, the term “human monoclonal antibody” refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobulin sequences. In one embodiment, the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g. a transgenic mouse, having a genome comprising a human heavy chain transgene and a light human chain transgene fused to an immortalized cell.
  • a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g. a transgenic mouse, having a genome comprising a human heavy chain transgene and a light human chain transgene fused to an immortalized cell.
  • Antibody fragments comprise a portion of a full length antibody, generally at least the antigen binding portion or the variable region thereof.
  • antibody fragments include Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies, single-chain antibody molecules, immunotoxins, and multispecific antibodies formed from antibody fragments.
  • antibody fragments comprise single chain polypeptides having the characteristics of a VH chain, namely being able to assemble together with a VL chain binding to the CD20 antigen.
  • “Antibody fragments” also comprise such fragments which per se are not able to provide effector functions (ADCC/CDC) but provide this function in a manner according to the invention after being combined with appropriate antibody constant domain(s).
  • amino acid sequence variant antibody herein is an antibody with an amino acid sequence which differs from a main species antibody.
  • amino acid sequence variants will possess at least about 70% homology with the main species antibody, and preferably, they will be at least about 80%, more preferably at least about 90% homologous with the main species antibody.
  • the amino acid sequence variants possess substitutions, deletions, and/or additions at certain positions within or adjacent to the amino acid sequence of the main species antibody.
  • amino acid sequence variants herein include acidic variant (e.g. deamidated antibody variant), basic variant, the antibody with an amino-terminal leader extension (e.g.
  • glycosylation variant antibody herein is an antibody with one or more carbohydrate moieties attached thereto which differ from one or more carbohydrate moieties attached to a main species antibody.
  • glycosylation variants herein include antibody with a G1 or G2 oligosaccharide structure, instead a G0 oligosaccharide structure, attached to an Fc region thereof, antibody with one or two carbohydrate moieties attached to one or two light chains thereof, antibody with no carbohydrate attached to one or two heavy chains of the antibody, etc, and combinations of glycosylation alterations.
  • glycosylation variant includes also glycoengineered antibodies such as those described in WO 1,331,266 and U.S. Pat. No. 7,517,670.
  • full length antibodies can be assigned to different “classes”. There are five major classes of full length antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called ⁇ (alpha), ⁇ (delta), ⁇ (epsilon), ⁇ (gamma), and ⁇ (mu), respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. See, e.g., Riechmann, L., et al., Nature 332 (1988) 323-327; and Neuberger, M. S., et al., Nature 314 (1985) 268-270.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NS0 or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences in a rearranged form.
  • the recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation.
  • the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • binding affinity is of Kd value of 10 ⁇ 9 mol/l or lower (e.g. 10 ⁇ 10 mol/l), preferably with a Kd value of 10 ⁇ 10 mol/l or lower (e.g. 10 ⁇ 12 mol/l).
  • the binding affinity is determined with a standard binding assay, such as surface plasmon resonance technique (BIACORE®).
  • the “constant domains” are not involved directly in binding the antibody to an antigen but are involved in the effector functions (ADCC, complement binding, and CDC).
  • variable region denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
  • the domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three “hypervariable regions” (or complementarity determining regions, CDRs).
  • the framework regions adopt a ⁇ -sheet conformation and the CDRs may form loops connecting the b-sheet structure.
  • the CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site.
  • the antibody heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
  • hypervariable region or “antigen-binding portion of an antibody” when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from the “complementarity determining regions” or “CDRs”.
  • “Framework” or “FR” regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • CDR3 of the heavy chain is the region which contributes most to antigen binding.
  • CDR and FR regions are determined according to the standard definition of Kabat, et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)) and/or those residues from a “hypervariable loop”.
  • CD20 and CD20 antigen are used interchangeably herein, and include any variants, isoforms and species homologs of human CD20 which are naturally expressed by cells or are expressed on cells transfected with the CD20 gene. Binding of an antibody of the invention to the CD20 antigen mediate the killing of cells expressing CD20 (e.g., a tumor cell) by inactivating CD20. The killing of the cells expressing CD20 may occur by one or more of the following mechanisms: antibody dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxity (CDC), inducing cell death and/or apoptosis, homotypic aggregation etc.
  • ADCC antibody dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxity
  • inducing cell death and/or apoptosis homotypic aggregation etc.
  • CD20 Synonyms of CD20, as recognized in the art, include B-lymphocyte antigen CD20, B-lymphocyte surface antigen B1, Leu-16, and Bp35.
  • anti-CD20 antibody is an antibody that binds specifically to CD20 antigen.
  • two types of anti-CD20 antibodies can be distinguished according to Cragg, M. S., et al, Blood 103 (2004) 2738-2743; and Cragg, M. S., et al Blood 101 (2003) 1045-1051, see Table 1:
  • Type II anti-CD20 antibodies Type II anti-CD20 antibodies
  • Type I CD20 epitope Localize CD20 to lipid rafts Do not localize CD20 to lipid rafts Increased CDC (if IgG1 isotype) Decreased CDC (if IgG1 isotype) ADCC activity (if IgG1 isotype) ADCC activity (if IgG1 isotype) Full binding capacity Reduced binding capacity Homotypic aggregation Stronger homotypic aggregation Apoptosis induction upon cross-linking Strong cell death induction without cross-linking
  • type I and type II anti-CD20 antibody can be classified by the ratio of the binding capacities to CD20 on Raji cells (ATCC-No. CCL-86) of said anti-CD20 antibody compared to rituximab.
  • anti-CD20 antibody can be either a type I or type II antibody. Preferably, it is a type I antibody, most preferred it is rituximab.
  • the type I anti-CD20 antibodies have a ratio of the binding capacities to CD20 on Raji cells (ATCC No. CCL-86) of said anti-CD20 antibody compared to rituximab of 0.8 to 1.2, preferably of 0.9 to 1.1.
  • type I anti-CD20 antibodies include e.g. Rituximab, in U.S. Pat. No. 7,381,560 (Anderson et. al., see e.g. FIGS.
  • 1F5 IgG2a (ECACC, hybridoma; Press et al., Blood 69/2:584-591 (1987)), H147 IgG3 (ECACC, hybridoma), 2C6 IgG1 (as disclosed in WO2005/103081), 2F2 IgG1 or ofatumumab (as disclosed and WO 2004/035607 and WO2005/103081) and 2H7 IgG1 (as disclosed in WO 2004/056312) and WO 2006/084264 (e.g. the variants disclosed in tables 1 and 2).
  • said type I anti-CD20 antibody is a monoclonal antibody that binds to the same epitope as rituximab.
  • the type II anti-CD20 antibodies have a ratio of the binding capacities to CD20 on Raji cells (ATCC No. CCL-86) of said anti-CD20 antibody compared to Rituximab of 0.3 to 0.6, preferably of 0.35 to 0.55, more preferably 0.4 to 0.5.
  • type II anti-CD20 antibodies include e.g. tositumomab (B1 IgG2a), humanized B-Ly1 antibody IgG1 (a chimeric humanized IgG1 antibody as disclosed in WO2005/044859), 11B8 IgG1 (as disclosed in WO 2004/035607), and AT80 IgG1.
  • said type II anti-CD20 antibody is a monoclonal antibody that binds to the same epitope as humanized B-Ly1 antibody (as disclosed in WO2005/044859).
  • the “ratio of the binding capacities to CD20 on Raji cells (ATCC-No. CCL-86) of an anti-CD20 antibodies compared to rituximab” is determined by direct immunofluorescence measurement (the mean fluorescent intensities (MFI) is measured) using said anti-CD20 antibody conjugated with Cy5 and rituximab conjugated with Cy5 in a FACSArray (Becton Dickinson) with Raji cells (ATCC-No. CCL-86), and calculated as follows:
  • Cy5-labeling ratio means number of Cy5-label molecules per molecule antibody.
  • said type I anti-CD20 antibody has a ratio of the binding capacities to CD20 on Raji cells (ATCC-No. CCL-86) of said first anti-CD20 antibody compared to rituximab of 0.8 to 1.2, preferably 0.9 to 1.1.
  • said type II anti-CD20 antibody has a ratio of the binding capacities to CD20 on Raji cells (ATCC-No. CCL-86) of said second anti-CD20 antibody compared to rituximab of 0.3 to 0.6, preferably 0.35 to 0.55, more preferably 0.4 to 0.5.
  • said type II anti-CD20 antibody preferably a humanized B-Ly1 antibody, has increased antibody dependent cellular cytotoxicity (ADCC).
  • ADCC antibody dependent cellular cytotoxicity
  • ADCC antibody having increased antibody dependent cellular cytotoxicity
  • the assay uses target cells that are known to express the target antigen recognized by the antigen-binding region of the antibody;
  • PBMCs peripheral blood mononuclear cells
  • the PBMCs are isolated using standard density centrifugation procedures and are suspended at 5 ⁇ 10 6 cells/ml in RPMI cell culture medium;
  • the target cells are grown by standard tissue culture methods, harvested from the exponential growth phase with a viability higher than 90%, washed in RPMI cell culture medium, labeled with 100 micro-Curies of 51 CI washed twice with cell culture medium, and resuspended in cell culture medium at a density of 10 5 cells/ml;
  • the antibody is serially-diluted from 4000 ng/ml to 0.04 ng/ml in cell culture medium and 50 microliters of the resulting antibody solutions are added to the target cells in the 96-well microtiter plate, testing in triplicate various antibody concentrations covering the whole concentration range above;
  • PBMC suspension 50 microliters of the PBMC suspension (point i above) are added to each well to yield an effector:target cell ratio of 25:1 and the plates are placed in an incubator under 5% CO2 atmosphere at 37 C for 4 hours;
  • “increased ADCC” is defined as either an increase in the maximum percentage of specific lysis observed within the antibody concentration range tested above, and/or a reduction in the concentration of antibody required to achieve one half of the maximum percentage of specific lysis observed within the antibody concentration range tested above.
  • the increase in ADCC is relative to the ADCC, measured with the above assay, mediated by the same antibody, produced by the same type of host cells, using the same standard production, purification, formulation and storage methods, which are known to those skilled in the art, but that has not been produced by host cells engineered to overexpress GnTIII.
  • Said “increased ADCC” can be obtained by glycoengineering of said antibodies, that means enhance said natural, cell-mediated effector functions of monoclonal antibodies by engineering their oligosaccharide component as described in Umana, P. et al., Nature Biotechnol. 17:176-180 (1999) and U.S. Pat. No. 6,602,684.
  • CDC complement-dependent cytotoxicity
  • CDC refers to lysis of human tumor target cells by the antibody according to the invention in the presence of complement.
  • CDC is measured preferably by the treatment of a preparation of CD20 expressing cells with an anti-CD20 antibody according to the invention in the presence of complement.
  • CDC is found if the antibody induces at a concentration of 100 nM the lysis (cell death) of 20% or more of the tumor cells after 4 hours.
  • the assay is performed preferably with 51 Cr or Eu labeled tumor cells and measurement of released 51 Cr or Eu. Controls include the incubation of the tumor target cells with complement but without the antibody.
  • type I and type II anti-CD20 antibodies of the IgG1 isotype show characteristic CDC properties.
  • Type I anti-CD20 antibodies have and increased CDC (if IgG1 isotype) and type II anti-CD20 antibodies have a decreased CDC (if IgG1 isotype) compared to each other.
  • Preferably both type I and type II anti-CD20 antibodies are IgG1 isotype antibodies.
  • the “rituximab” antibody is a genetically engineered chimeric human gamma 1 murine constant domain containing monoclonal antibody directed against the human CD20 antigen.
  • This chimeric antibody contains human gamma 1 constant domains and is identified by the name “C2B8” in EP2000149B1 and U.S. Pat. No. 7,381,560 (Anderson et. al., see e.g. FIGS. 4 and 5).
  • Rituximab is approved for the treatment of patients with relapsed or refracting low-grade or follicular, CD20 positive, B cell non-Hodgkin's lymphoma.
  • rituximab exhibits human complement—dependent cytotoxicity (CDC) (Reff et. al, Blood 83(2): 435-445 (1994)). Additionally, it exhibits significant activity in assays that measure antibody-dependent cellular cytotoxicity (ADCC).
  • humanized B-Ly1 antibody refers to humanized B-Ly1 antibody as disclosed in WO2005/044859, which were obtained from the murine monoclonal anti-CD20 antibody B-Ly1 (variable region of the murine heavy chain (VH): SEQ ID NO: 1; variable region of the murine light chain (VL): SEQ ID NO: 2; see Poppema, S, and Visser, L., Biotest Bulletin 3: 131-139 (1987)) by chimerization with a human constant domain from IgG1 and following humanization (see WO2005/044859).
  • VH murine heavy chain
  • VL variable region of the murine light chain
  • VL variable region of the murine light chain
  • the “humanized B-Ly1 antibody” has variable region of the heavy chain (VH) selected from group of SEQ ID No: 3 to SEQ ID No: 20 (B-HH2 to B-HH9 and B-HL8 to B-HL17 of WO2005/044859). Especially preferred are Seq. ID Nos: 3, 4, 7, 9, 11, 13 and 15 (B-HH2, BHH-3, B-HH6, B-HH8, B-HL8, B-HL11 and B-HL13 of WO2005/044859). Most preferably, said VH is BHH6.
  • the “humanized B-Ly1 antibody” has variable region of the light chain (VL) of SEQ ID No: 20 (B-KV1) of WO2005/044859.
  • humanized B-Ly1 antibody is preferably an IgG1 antibody.
  • humanized B-Ly1 antibodies are glycoengineered (GE) in the Fc region according to the procedures described in WO2005/044859, WO 2004/065540, Umana, P. et al., Nature Biotechnol. 17:176-180 (1999) and WO 99/154342.
  • Most glycoengineered humanized B-Ly1 antibodies have an altered pattern of glycosylation in the Fc region, preferably having a reduced level of fucose residues.
  • the oligosaccharides of the Fc region are non-fucosylated.
  • the oligosaccharides of the Fc region are preferably bisected.
  • the “humanized B-Ly1 antibody” comprises VH B-HH6 and VL B-KV1 of WO2005/044859. As used herein, said antibody is also referred to as “HuMab ⁇ CD20>”. Said antibody was designated with the INN Afutuzumab.
  • said antibody has a reduced level of fucose residues as defined above and/or the oligosaccharides of the Fc region are most preferably bisected. In yet another most preferable embodiment, said antibody displays increased ADCC as defined herein.
  • the oligosaccharide component can significantly affect properties relevant to the efficacy of a therapeutic glycoprotein, including physical stability, resistance to protease attack, interactions with the immune system, pharmacokinetics, and specific biological activity. Such properties may depend not only on the presence or absence, but also on the specific structures, of oligosaccharides. Some generalizations between oligosaccharide structure and glycoprotein function can be made. For example, certain oligosaccharide structures mediate rapid clearance of the glycoprotein from the bloodstream through interactions with specific carbohydrate binding proteins, while others can be bound by antibodies and trigger undesired immune reactions. (Jenkins et al., Nature Biotechnol. 14:975-81 (1996)).
  • Mammalian cells are the preferred hosts for production of therapeutic glycoproteins, due to their capability to glycosylate proteins in the most compatible form for human application. (Cumming et al., Glycobiology 1:115-30 (1991); Jenkins et al., Nature Biotechnol. 14:975-81 (1996)). Bacteria very rarely glycosylate proteins, and like other types of common hosts, such as yeasts, filamentous fungi, insect and plant cells, yield glycosylation patterns associated with rapid clearance from the blood stream, undesirable immune interactions, and in some specific cases, reduced biological activity. Among mammalian cells, Chinese hamster ovary (CHO) cells have been most commonly used during the last two decades.
  • these cells allow consistent generation of genetically stable, highly productive clonal cell lines. They can be cultured to high densities in simple bioreactors using serumfree media, and permit the development of safe and reproducible bioprocesses.
  • Other commonly used animal cells include baby hamster kidney (BHK) cells, NS0— and SP2/0-mouse myeloma cells. More recently, production from transgenic animals has also been tested. (Jenkins et al., Nature Biotechnol. 14: 975-981 (1996)).
  • All antibodies contain carbohydrate structures at conserved positions in the heavy chain constant regions, with each isotype possessing a distinct array of N-linked carbohydrate structures, which variably affect protein assembly, secretion or functional activity.
  • N-linked carbohydrate structures which variably affect protein assembly, secretion or functional activity.
  • the structure of the attached N-linked carbohydrate varies considerably, depending on the degree of processing, and can include highmannose, multiply-branched as well as biantennary complex oligosaccharides. (Wright, A., and Morrison, S. L., Trends Biotech. 15: 26-32 (1997)).
  • ADCC antibody dependent cellular cytotoxicity
  • GnTIII ⁇ (1,4)-N-acetylglucosaminyltransferase III
  • the antibody chCE7 belongs to a large class of unconjugated monoclonal antibodies which have high tumor affinity and specificity, but have too little potency to be clinically useful when produced in standard industrial cell lines lacking the GnTIII enzyme (Umana, P., et al., Nature Biotechnol. 17: 176-180 (1999)). That study was the first to show that large increases of ADCC activity could be obtained by engineering the antibody producing cells to express GnTIII, which also led to an increase in the proportion of constant region (Fc)-associated, bisected oligosaccharides, including bisected, non-fucosylated oligosaccharides, above the levels found in naturally-occurring antibodies.
  • Fc constant region
  • lymphomas preferably B-Cell Non-Hodgkin's lymphomas (NHL)
  • lymphocytic leukemias include e.g.: (a) follicular lymphomas, (b) Small Non-Cleaved Cell Lymphomas/Burkitt's lymphoma (including endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma and Non-Burkitt's lymphoma), (c) marginal zone lymphomas (including extranodal marginal zone B cell lymphoma (Mucosa-associated lymphatic tissue lymphomas, MALT), nodal marginal zone B cell lymphoma and splenic marginal zone lymphoma), (d) Mantle cell lymphoma (MCL), (e) Large Cell Lymphoma (including B-cell diffuse large cell lymphoma
  • the CD20 expressing cancer is a B-Cell Non-Hodgkin's lymphomas (NHL).
  • CD20 expressing cancers include: Mantle cell lymphoma (MCL), acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), B-cell diffuse large cell lymphoma (DLCL), Burkitt's lymphoma, hairy cell leukemia, follicular lymphoma, multiple myeloma, marginal zone lymphoma, post transplant lymphoproliferative disorder (PTLD), HIV associated lymphoma, Waldenstrom's macroglobulinemia, or primary CNS lymphoma.
  • MCL Mantle cell lymphoma
  • ALL acute lymphocytic leukemia
  • CLL chronic lymphocytic leukemia
  • DLCL B-cell diffuse large cell lymphoma
  • Burkitt's lymphoma hairy cell leukemia
  • follicular lymphoma multiple myelo
  • autoimmune disease relates to a disease or disorder arising from and directed against an individual's own tissues.
  • autoimmune diseases or disorders include, but are not limited to arthritis (rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), psoriasis, dermatitis, polymyositis/dermatomyositis, toxic epidermal necrolysis, systemic scleroderma and sclerosis, responses associated with inflammatory bowel disease, Crohn's disease, ulcerative colitis, respiratory distress syndrome, adult respiratory distress syndrome (ARDS), meningitis, encephalitis, uveitis, colitis, glomerulonephritis, allergic conditions, eczema, asthma, conditions involving infiltration of T cells and chronic inflammatory responses, atherosclerosis, autoimmune myocarditis, leukocyte adhesion deficiency, systemic lupus erythematosus (SLE)
  • a “growth inhibitory agent” when used herein refers to a compound or composition which inhibits growth of a cell, especially a CD20 expressing cancer cell either in vitro or in vivo.
  • the growth inhibitory agent may be one which significantly reduces the percentage of CD20 expressing cells in S phase.
  • growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest.
  • Classical M-phase blockers include the vincas (vincristine and vinblastine), taxanes, and topo II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin.
  • DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C.
  • DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disease as well as those in which the disease is to be prevented. Hence, the patient to be treated herein may have been diagnosed as having the disease or may be predisposed or susceptible to the disease.
  • cytotoxic agent refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • the term is intended to include radioactive isotopes (e.g. At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu), chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof.
  • radioactive isotopes e.g. At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu
  • chemotherapeutic agents e.g. At 211 , I 131 , I 125 , Y 90 , Re
  • chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXANTM); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelarnine; acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARENOLTM); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topot
  • dynemicin including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCINTM, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin, doxorubicin HCl liposome injection (DOXILTM), liposomal
  • celecoxib or etoricoxib proteosome inhibitor
  • proteosome inhibitor e.g. PS341
  • bortezomib VELCADETM
  • CCI-779 tipifarnib (R11577); orafenib, ABT510
  • Bc1-2 inhibitor such as oblimersen sodium (GENASENSETM)
  • pixantrone EGFR inhibitors (see definition below); tyrosine kinase inhibitors (see definition below); and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone (optionally further comprising interferon- ⁇ (CHVP/interferon- ⁇ ), FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATINTM) combined with 5-FU and leu
  • an “anti-angiogenic agent” refers to a compound which blocks, or interferes with to some degree, the development of blood vessels.
  • the anti-angiogenic factor may, for instance, be a small molecule or antibody that binds to a growth factor or growth factor receptor involved in promoting angiogenesis.
  • the preferred anti-angiogenic factor herein is an antibody that binds to Vascular Endothelial Growth Factor (VEGF), such as Bevacizumab (AVASTINTM).
  • VEGF Vascular Endothelial Growth Factor
  • cytokine is a generic term for proteins released by one cell population which act on another cell as intercellular mediators.
  • cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone, parathyroid hormone, thyroxine, insulin, proinsulin, relaxin; prorelaxin, glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH), hepatic growth factor; fibroblast growth factor, prolactin, placental lactogen, tumor necrosis factor ⁇ and ⁇ , mullerian-inhibiting substance, mouse gonadotropin-associated peptide, inhibin; activin, vascular endothelial growth factor, integrin, thrombopoietin (TPO), nerve growth factors such as NGF-13, platelet-growth factor; transforming growth hormone such
  • an effective amount refers to an amount which provides the desired effect.
  • an effective amount is the amount necessary to increase the dispersion and absorption of the co-administered anti-CD20 antibody in such a way that the anti-CD20 antibody can act in a therapeutically effective way as outline above.
  • a pharmaceutical drug substance it is the amount of active ingredient effective to treat a disease in the patient.
  • the effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • the effective amount may extend progression free survival, result in an objective response (including a partial response, PR, or complete response, CR), increase overall survival time, and/or improve one or more symptoms of cancer.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of the active ingredient to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile.
  • a “sterile” formulation is aseptic or free from all living microorganisms and their spores.
  • a “stable” formulation is one in which all the protein therein essentially retain their physical stability and/or chemical stability and/or biological activity upon storage at the intended storage temperature, e.g. 2-8° C.
  • the formulation essentially retains its physical and chemical stability, as well as its biological activity upon storage.
  • the storage period is generally selected based on the intended shelf-life of the formulation.
  • the formulation is preferably stable following freezing (to, e.g., ⁇ 20° C.) and thawing of the formulation, for example following 1 or more cycles of freezing and thawing.
  • Stability can be measured at a selected temperature for a selected time period.
  • Stability can be evaluated qualitatively and/or quantitatively in a variety of different ways, including evaluation of aggregate formation (for example using size exclusion chromatography, by measuring turbidity, and/or by visual inspection); by assessing charge heterogeneity using cation exchange chromatography or capillary zone electrophoresis; SDS-PAGE analysis to compare reduced and intact antibody; evaluating biological activity or antigen binding function of the antibody; etc.
  • Instability may involve any one or more of: aggregation, deamidation (e.g. Asn deamidation), oxidation (e.g. Met oxidation), isomerization (e.g. Asp isomeriation), clipping/hydrolysis/fragmentation (e.g. hinge region fragmentation), succinimide formation, unpaired cysteine(s), etc.
  • Therapeutic formulations of the antibodies used in accordance with the present invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed.
  • surfactant denotes a pharmaceutically acceptable surface-active agent.
  • the amount of surfactant is described a percentage expressed in weight/volume. The most commonly used weight/volume unit is mg/mL.
  • suitable examples of pharmaceutically acceptable surfactants include polyoxyethylen-sorbitan fatty acid esters (Tween), polyethylene-polypropylene glycols, polyoxyethylene-stearates, polyoxyethylene alkyl ethers, e.g.
  • Most suitable polyoxyethylenesorbitan-fatty acid esters are polysorbate 20, (sold under the trademark Tween 20TM) and polysorbate 80 (sold under the trademark Tween 80TM).
  • Most suitable polyethylene-polypropylene copolymers are those sold under the names Pluronic® F68 or Poloxamer 188TM.
  • Preferred polyoxyethylene-stearates are those sold under the trademark MyrjTM.
  • Most suitable polyoxy-ethylene alkyl ethers are those sold under the trademark BrijTM
  • Most suitable alkylphenolpoly-oxyethylene ethers are sold under the trade name Triton-X.
  • buffer denotes a pharmaceutically acceptable buffer.
  • buffering agent providing a pH of 5.5 ⁇ 2.0 refers to an agent which provides that the solution comprising it resists changes in pH by the action of its acid/base conjugate components.
  • Suitable pharmaceutically acceptable buffers according to the invention comprise but are not limited to histidine-buffers, citrate-buffers, gluconate-buffers, succinate-buffers, acetate-buffers glycylglycine and other organic acid buffers, and phosphate-buffers.
  • Preferred buffers comprise L-histidine or mixtures of L-histidine with L-histidine hydrochloride with isotonicity agents and potentially pH adjustment with an acid or a base known in the art. Most preferred is L-histidine.
  • a “histidine buffer” is a buffer comprising the amino acid histidine.
  • histidine buffers include histidine chloride, histidine acetate, histidine phosphate, histidine sulfate.
  • the preferred histidine buffer identified in the examples herein was found to be histidine chloride.
  • the histidine chloride buffer is prepared by titrating L-histidine (free base, solid) with diluted hydrochloric acid or by dissolving L-histidine and L-histidine hydrochloride (e.g. as monohydrate) in a defined amount and ratio.
  • isotonic is meant that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmolality of ⁇ 300 mOsm/kg. Isotonicity can be measured using a vapor pressure or freezing-point depression type osmometer.
  • isotonicity agents denotes pharmaceutically acceptable isotonicity agents. Isotonicity agents are used to provide an isotonic formulation.
  • An isotonic formulation is liquid or liquid reconstituted from a solid form, e.g. a lyophilized form and denotes a solution having the same tonicity as some other solution with which it is compared, such as physiologic salt solution and the blood serum.
  • Suitable isotonicity agents comprise but are not limited to salts, including but not limited to sodium chloride (NaCl) or potassium chloride, sugars and sugar alcohols including but not limited to glucose, sucrose, trehalose or glycerol and any component from the group of amino acids, sugars, salts and combinations thereof.
  • Isotonicity agents are generally used in a total amount of about 5 mM to about 350 mM.
  • liquid as used herein in connection with the formulation according to the invention denotes a formulation which is liquid at a temperature of at least about 2 to about 8° C.
  • lyophilized as used herein in connection with the formulation according to the invention denotes a formulation which is dried by freezing the formulation and subsequently subliming the ice from the frozen content by any freeze-drying methods known in the art, for example commercially available freeze-drying devices.
  • salts denotes a salt in an amount of about 1 mM to about 500 mM.
  • Non-limiting examples of salts include salts of any combinations of the cations sodium potassium, calcium or magnesium with anions chloride, phosphate, citrate, succinate, sulphate or mixtures thereof.
  • amino acid denotes an amino acid in an amount of about 1 to about 100 mg/mL comprising but not limited to arginine, glycine, ornithine, glutamine, asparagine, lysine, histidine, glutamic acid, asparagic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophane, methionine, serine, proline.
  • a “saccharide” herein comprises the general composition (CH 2 O) n and derivatives thereof, including monosaccharides, disaccharides, trisaccharides, polysaccharides, sugar alcohols, reducing sugars, nonreducing sugars, etc.
  • saccharides herein include glucose, sucrose, trehalose, lactose, fructose, maltose, dextran, glycerin, dextran, erythritol, glycerol, arabitol, sylitol, sorbitol, mannitol, mellibiose, melezitose, raffinose, mannotriose, stachyose, maltose, lactulose, maltulose, glucitol, maltitol, lactitol, iso-maltulose, etc.
  • glucosamine N-Methylglucosamine (so-called “Meglumine”), galactosamine and neuraminic acid and combinations of the saccharides according to the invention.
  • the preferred saccharide herein is a non-reducing disaccharide, such as trehalose or sucrose.
  • the most preferred saccharide in accordance with the present invention is trehalose.
  • stabilizer refers to pharmaceutically acceptable stabilizers, like for example but not limited to amino acids and sugars as described in the above sections as well as commercially available dextrans of any kind and molecular weight as known in the art.
  • antioxidant denotes a pharmaceutically acceptable antioxidant. This may include excipients such as methionine, benzylalcohol or any other excipient used to minimize oxidation.
  • a method of treating when applied to, for example, cancer refers to a procedure or course of action that is designed to reduce or eliminate the number of cancer cells in a patient, or to alleviate the symptoms of a cancer.
  • a method of treating does not necessarily mean that the cancer cells or other disorder will, in fact, be eliminated, that the number of cells or disorder will, in fact, be reduced, or that the symptoms of a cancer or other disorder will, in fact, be alleviated.
  • a method of treating cancer will be performed even with a low likelihood of success, but which, given the medical history and estimated survival expectancy of a patient, is nevertheless deemed to induce an overall beneficial course of action.
  • the problem to be solved by the present invention is therefore to provide novel highly concentrated, stable pharmaceutical formulations of a pharmaceutically active anti-CD20 antibody or a mixture of such antibody molecules for subcutaneous injection.
  • Such formulations comprise, in addition to the high amounts of anti-CD20 antibody or mixture thereof, a buffering agent, a stabilizer or a mixture of two or more stabilizers, a nonionic surfactant and preferably an effective amount of at least one hyaluronidase enzyme.
  • the preparation of highly-concentrated antibody formulations is challenging because of a potential increase in viscosity at higher protein concentration and a potential increase in protein aggregation, a phenomenon that is per se concentration-dependent. High viscosities negatively impact the process ability (e.g.
  • the present invention provides a highly concentrated, stable pharmaceutical formulation of a pharmaceutically active anti-CD20 antibody or a mixture of such antibody molecules for parenteral application.
  • the route of application is intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, or intrathecal routes.
  • Intravenous or subcutaneous administration of the antibodies is preferred; subcutaneous injection is preferred most.
  • a pharmaceutically active anti-CD20 antibody formulation of the present invention comprises:
  • the highly concentrated, stable pharmaceutical formulation of a pharmaceutically active anti-CD20 antibody formulation of the present invention may be provided in liquid form or may be provided in lyophilized form.
  • the antibody concentration in the reconstituted formulation can be increased by reconstitution of a lyophilized formulation to provide a protein concentration in the reconstituted formulation which is about 2-40 times greater than the protein concentration in the mixture before the lyophilization step.
  • the preferred anti-CD20 antibody concentration is 50 to 150 mg/ml, more preferred is 75 to 150 mg/ml, even more preferred is 120 ⁇ 20 mg/ml, most preferred is about 120 mg/ml.
  • the preferred concentration of the buffering agent is 1 to 50 mM, more preferably 10 to 30 mM; the most preferred concentration is about 20 mM.
  • Various buffering agents are known to the person skilled in the art as outlined above.
  • the preferred buffering agent is selected from the group consisting of a histidine buffer, acetic acid buffer, and citric acid buffer, most preferably a L-histidine/HCl buffer.
  • the histidine-buffer according to the invention is used in an amount of about 1 mM to about 50 mM, preferably of about 10 mM to about 30 mM and still more preferably of about 20 mM.
  • the acetic acid buffer according to the invention is preferably of about 10 mM to about 30 mM and most preferably of about 20 mM.
  • the citric acid buffer according to the invention is preferably of about 10 mM to about 30 mM and most preferably of about 20 mM.
  • the pH will be adjusted at a value comprising about 4.5 to about 7.0 and preferably about 5.5 to about 6.5, also preferably preferably selected from the group consisting of 5.5, 6.0, 6.1 and 6.5.
  • This pH can be obtained by adjustment with an acid or base known in the art or by using adequate mixtures of buffer components or both.
  • the stabilizer(s) (used synonymously with the term “stabilizing agent” in the present patent description) is/are preferably selected from the group consisting of a salt, a carbohydrate, saccharide and amino acid(s), more preferably a carbohydrate or saccharide, more preferably a sugar admitted by the authorities as a suitable additive or excipient in pharmaceutical formulations, most preferably selected from the group consisting of ⁇ , ⁇ -trehalose dihydrate, NaCl and methionine.
  • the preferred concentration of the stabilizer is 15 to 250 mM, or more preferably 150 to 250 mM. Most preferred is a concentration of about 210 mM.
  • the formulation may contain a secondary stabilizer, whereby this secondary stabilizer is preferably methionine, preferably in a concentration of 5 to 25 mM, more preferably in a concentration of 5 to 15 mM.
  • This most preferred methionine concentration is about 10 mM.
  • the nonionic surfactant is preferably a polysorbate, more preferably is selected from the group of polysorbate 20, polysorbate 80 and polyethylene-polypropylene copolymer.
  • the concentration of the nonionic surfactant is 0.01 to 0.1% (w/v), or 0.02 to 0.08% (w/v) and preferably 0.02 to 0.06% (w/v), most preferably about 0.06% (w/v).
  • sugar denotes a pharmaceutically acceptable sugar used in an amount of about 25 mM to about 500 mM. Preferred is 100 to 300 mM. More preferred is 180 to 240 mM. Most preferred is 210 mM.
  • the concentration of the hyaluronidase enzyme depends on the actual hyaluronidase enzyme used in the preparation of the formulation in accordance with the invention.
  • An effective amount of the hyaluronidase enzyme can easily be determined by the person skilled in the art based on the disclosure further below. It should be provided in sufficient amount so that an increase in the dispersion and absorption of the co-administered anti-CD20 antibody is possible.
  • the effective amount of the hyaluronidase enzyme is preferably about 1,000 to 16,000 U/ml, whereby the said amount corresponds to about 0.01 mg to 0.15 mg protein based on an assumed specific activity of 100,000 U/mg.
  • the preferred concentration of the hyaluronidase enzyme is about 1,500 to 12,000 U/ml. Most preferred is a concentration of about 2,000 U/ml or about 12,000 U/ml.
  • the amounts specified herein before correspond to the amount of hyaluronidase enzyme initially added to the formulation.
  • the hyaluronidase enzyme is present either as a combined final formulation or for use for co-administration, e.g. as a co-formulation as further outlined below.
  • the important issue for the claimed formulation is that at the time it is ready for use and/or is injected it has the claimed composition.
  • the hyaluronidase enzyme may be derived from animals, human samples or manufactured based on the recombinant DNA technology as described further below.
  • compositions More particularly the highly concentrated, stable pharmaceutical formulations in accordance with the present invention have one of the following preferred compositions:
  • a) 100 to 150 mg/ml anti-CD20 antibody whereby this antibody is preferably Rituximab, Ocrelizumab or HuMab ⁇ CD20>; 1 to 50 mM of a histidine buffer, preferably L-histidine/HCl at a pH of about 5.5; 15 to 250 mM of a stabilizer which is preferably ⁇ , ⁇ -trehalose dihydrate and optionally methionine as a second stabilizer at a concentration of 5 to 25 mM; a non-ionic surfactant selected from the group of polysorbate 20 and polysorbate 80, preferably 0.02 to 0.06% (w/v), and optionally 1,000 to 16,000 U/ml of a hyaluronidase enzyme, preferably rHuPH20, most preferably at a concentration of 2,000 U/ml or 12,000 U/ml.
  • a histidine buffer preferably L-histidine/HCl at a pH of about 5.5
  • a stabilizer which is preferably
  • b) 120 ⁇ 20 mg/ml anti-CD20 antibody whereby this antibody is preferably Rituximab, Ocrelizumab or HuMab ⁇ CD20>; 10 to 30 mM, preferably 20 mM of a histidine buffer, preferably L-histidine/HCl at a pH of about 5.5; 150 to 250 mM, preferably 210 mM of a stabilizer which is preferably ⁇ , ⁇ -trehalose dihydrate and optionally methionine as a second stabilizer at a concentration of 5 to 25 mM, preferably 5 to 15 mM, most preferably 10 mM; a non-ionic surfactant selected from the group of polysorbate 20 and polysorbate 80, preferably 0.02 to 0.06% (w/v), and optionally 1,000 to 16,000 U/ml, preferably 1,500 to 12.000 U/ml, most preferably 2,000 U/ml or 12,000 U/ml of a hyaluronidase enzyme, preferably rHu
  • c) 120 mg/ml anti-CD20 antibody whereby this antibody is preferably Rituximab, Ocrelizumab or HuMab ⁇ CD20>; 10 to 30 mM, preferably 20 mM of a histidine buffer, preferably L-histidine/HCl at a pH of about 5.5; 150 to 250 mM, preferably 210 mM of a stabilizer which is preferably ⁇ , ⁇ -trehalose dihydrate and optionally methionine as a second stabilizer at a concentration of 5 to 25 mM, preferably 5 to 15 mM, most preferably 10 mM; a non-ionic surfactant selected from the group of polysorbate 20 and polysorbate 80, preferably 0.02 to 0.06% (w/v), and optionally 1,000 to 16,000 U/ml, preferably 1,500 to 12,000 U/ml, most preferably 2,000 U/ml or 12,000 U/ml of a hyaluronidase enzyme, preferably rHuPH20
  • a histidine buffer preferably L-histidine/HCl at a pH of about 5.5
  • 210 mM ⁇ , ⁇ -trehalose dihydrate optionally 10 mM methionine as a second stabilizer
  • a non-ionic surfactant selected from the group of polysorbate 20 and polysorbate 80, preferably 0.02 to 0.06% (w
  • a lyophilized formulation comprising 120 mg/ml anti-CD20 antibody, preferably Rituximab; 20 mM of a histidine buffer, preferably L-histidine/HC1 at a pH of about 5.5; 210 mM of ⁇ , ⁇ -trehalose dihydrate and optionally 10 mM methionine as a second stabilizer; a non-ionic surfactant selected from the group of polysorbate 20 and polysorbate 80, preferably 0.02 to 0.06% (w/v), and optionally 2,000 U/ml or 12,000 U/ml of a hyaluronidase enzyme, preferably rHuPH20.
  • a stable pharmaceutical formulation of a pharmaceutically active anti-CD20 antibody comprising about 30 mg/ml to 350 mg/ml, for example about 30 mg/ml to 100 mg/ml (including about 30 mg/ml, about 50 mg/ml or about 100 mg/ml) Ocrelizumab (e.g. humanized 2H7.v16); about 1 to 100 mM of a buffering agent (e.g. sodium acetate) providing a pH of 5.5 ⁇ 2.0 (e.g. pH 5.3); about 15 to 250 mM of a stabilizer or a mixture of two or more stabilizers (including trehalose, e.g.
  • a buffering agent e.g. sodium acetate
  • a pH of 5.5 ⁇ 2.0 e.g. pH 5.3
  • trehalose e.g.
  • trehalose dihydrate about 8% trehalose dihydrate); about 0.01 to 0.1% (w/v) of a nonionic surfactant; and optionally an effective amount of at least one hyaluronidase enzyme (e.g. rhHUPH20), preferably in an amount from about 1,500 U/ml to about 12,000 U/ml.
  • hyaluronidase enzyme e.g. rhHUPH20
  • soluble hyaluronidase glycoproteins see WO2006/091871. It has been shown that the addition of such soluble hyaluronidase glycoproteins (either as a combined formulation or by co-administration) facilitates the administration of therapeutic drug into the hypodermis.
  • sHASEGPs soluble hyaluronidase glycoproteins
  • By rapidly depolymerizing hyaluronan HA in the extracellular space sHASEGP reduces the viscosity of the interstitium, thereby increasing hydraulic conductance and allowing for larger volumes to be administered safely and comfortably into the subcutaneous tissue.
  • the increased hydraulic conductance induced by sHASEGP through reduced interstitial viscosity allows for greater dispersion, potentially increasing the systemic bioavailability of SC administered therapeutic drug.
  • the highly concentrated, stable pharmaceutical formulations of the present invention comprising a soluble hyaluronidase glycoprotein are therefore particularly suited for subcutaneous injection. It is clearly understood by the person skilled in the art that such a formulation comprising an anti-CD20 antibody and a soluble hyaluronidase glycoprotein can be provided for administration in form of one single combined formulation or alternatively in form of two separate formulations which can be mixed just prior to the subcutaneous injection. Alternatively the anti-CD20 antibody and the soluble hyaluronidase glycoprotein can be administered as separate injections at different sites of the body, preferably at sites which are immediately adjacent to each other. It is also possible to inject the therapeutic agents present in the formulation in accordance with the present invention as consecutive injections, e.g.
  • the soluble hyaluronidase glycoprotein followed by the injection of the anti-CD20 antibody formulation.
  • injections can also be performed in the reversed order, viz. by first injecting the anti-CD20 antibody formulation followed by injecting the soluble hyaluronidase glycoprotein.
  • the anti-CD20 antibody and the soluble hyaluronidase glycoprotein are administered as separate injections, one or both of the proteins have to be provided with the buffering agent, the stabilizer(s) and the nonionic surfactant in the concentrations as specified in the appended claims but excluding the hyaluronidase enzyme.
  • the hyaluronidase enzyme can then be provided e.g.
  • the anti-CD20 antibody is provided with the buffering agent, the stabilizer(s) and the nonionic surfactant in the concentrations as specified in the appended claims.
  • the soluble hyaluronidase glycoprotein may be considered to be a further excipient in the anti-CD20 formulation.
  • the soluble hyaluronidase glycoprotein may be added to the anti-CD20 formulation at the time of manufacturing the anti-CD20 formulation or may be added shortly before the injection.
  • the soluble hyaluronidase glycoprotein may be provided as a separate injection.
  • the soluble hyaluronidase glycoprotein may be provided in a separate vial either in lyophilized form which must be reconstituted with suitable diluents before the subcutaneous injection takes place, or my be provided as a liquid formulation by the manufacturer.
  • the anti-CD20 formulation and the soluble hyaluronidase glycoprotein may be procured as separate entities or may also be provided as kits comprising both injection components and suitable instructions for their subcutaneous administration. Suitable instructions for the reconstitution and/or administration of one or both of the formulations may also be provided.
  • the present invention also provides pharmaceutical compositions consisting of an a highly concentrated, stable pharmaceutical formulation of a pharmaceutically active anti-CD20 antibody or a mixture of such antibody and a suitable amount of at least one hyaluronidase enzyme in the form of a kit comprising both injection components and suitable instructions for their subcutaneous administration.
  • a further aspect of the present invention relates to injection devices comprising a highly concentrated, stable pharmaceutical formulation in accordance with the present invention.
  • Such formulation may consist of a pharmaceutically active anti-CD20 antibody or a mixture of such antibody molecules and suitable excipients as outlined below and may additionally comprise a soluble hyaluronidase glycoprotein either as a combined formulation or as a separate formulation for co-administration.
  • anti-CD20 antibodies are known in the prior art. Such antibodies are preferably monoclonal antibodies. The may either be so-called chimaeric antibodies, humanized antibodies or fully human antibodies. They may either be full length anti-CD20 antibodies; anti-CD20 antibody fragments having the same biological activity; including amino acid sequence variants and/or glycosylation variants of such antibodies or fragments.
  • humanized anti-CD20 antibodies are known under the INN names Rituximab, Ocrelizumab and Afutuzumab (HuMab ⁇ CD20>). The most successful therapeutic anti-CD20 antibody is Rituximab sold by Genentech Inc. and F. Hoffmann-La Roche Ltd under the trade name MABTHERA® or RITUXAN®.
  • the anti-CD20 antibody as defined herein is preferably selected from the group of Rituximab (see e.g. U.S. Pat. No. 7,381,560, and EP2000149B1 Anderson et. al., see e.g. FIGS. 4 and 5), Ocrelizumab (as disclosed in WO 2004/056312) and WO 2006/084264 (e.g. the variants disclosed in tables 1 and 2), preferably the variant v.16 or v.114 or v.511 and Afutuzumab (HuMab ⁇ CD20>; see WO2005/044859).
  • the most preferred anti-CD20 antibody is Rituximab.
  • Rituximab encompass all corresponding anti-CD20 antibodies that fullfill the requirements necessary for obtaining a marketing authorization as an identical or biosimilar product in a country or territory selected from the group of countries consisting of the USA, Europe and Japan.
  • Rituximab has the CDR regions defined in U.S. Pat. No. 7,381,560 and EP2000149B1.
  • a number of a soluble hyaluronidase glycoproteins are known in the prior art. In order to further define the function, the mechanism of action and the properties of such soluble hyaluronidase glycoproteins the following background information is provided.
  • the SC (hypodermal) interstitial matrix is comprised of a network of fibrous proteins embedded within a viscoelastic gel of glycosaminoglycans.
  • Hyaluronan a non-sulfated repeating linear disaccharide, is the prominent glycosaminoglycan of the SC tissue.
  • HA is secreted into the interstitium by fibroblasts as a high molecular weight, megadalton viscous polymer that is subsequently degraded locally, in the lymph, and in the liver, through the action of lysosomal hyaluronidases and exoglycosidases.
  • HA As a major constituent of the gel-like component of the hypodermal matrix, HA contributes significantly to its viscosity.
  • Glycosaminoglycans are complex linear polysaccharides of the extracellular matrix (ECM). GAGs are characterized by repeating disaccharide structures of an N-substituted hexosamine and an uronic acid (in the case of hyaluronan (HA), chondroitin sulfate (CS), chondroitin (C), dermatan sulfate (DS), heparan sulfate (HS), and heparin (H)), or a galactose (in the case of keratan sulfate (KS)). Except for HA, all exist covalently bound to core proteins.
  • the GAGs with their core proteins are structurally referred to as proteoglycans (PGs).
  • Hyaluronan (HA) is found in mammals predominantly in connective tissues, skin, cartilage, and in synovial fluid. Hyaluronan is also the main constituent of the vitreous of the eye. In connective tissue, the water of hydration associated with hyaluronan creates hydrated matrices between tissues. Hyaluronan plays a key role in biological phenomena associated with cell motility including rapid development, regeneration, repair, embryogenesis, embryological development, wound healing, angiogenesis, and tumorigenesis (Toole, Cell Biol. Extracell. Matrix, Hay (ed), Plenum Press, New York, 1991; pp. 1384-1386; Bertrand et al., Int. J.
  • HA is found in the extracellular matrix of many cells, especially in soft connective tissues. HA has been assigned various physiological functions, such as in water and plasma protein homeostasis (Laurent T. C. et al., FASEB J., 1992; 6: 2397-2404). HA production increases in proliferating cells and may play a role in mitosis. It has also been implicated in locomotion and cell migration. HA seems to play important roles in cell regulation, development, and differentiation (Laurent et al., supra).
  • HA has widely been used in clinical medicine. Its tissue protective and rheological properties have proved useful in ophthalmic surgery (e.g. to protect the corneal endothelium during cataract surgery). Serum HA is diagnostic of liver disease and various inflammatory conditions, such as rheumatoid arthritis. Interstitial edema caused by accumulation of HA may cause dysfunction in various organs (Laurent et al., supra).
  • Hyaluronan protein interactions also are involved in the structure of the extracellular matrix or “ground substance”.
  • Hyaluronidases are a group of generally neutral- or acid-active enzymes found throughout the animal kingdom. Hyaluronidases vary with respect to substrate specificity, and mechanism of action (WO 2004/078140). There are three general classes of hyaluronidases:
  • Mammalian-type hyaluronidases (EC 3.2.1.35) which are endo-beta-N-acetylhexosaminidases with tetrasaccharides and hexasaccharides as the major end products. They have both hydrolytic and transglycosidase activities, and can degrade hyaluronan and chondroitin sulfates (CS), generally C4-S and C6-S.
  • Bacterial hyaluronidases (EC 4.2.99.1) degrade hyaluronan and, and to various extents, CS and DS.
  • Hyaluronidases (EC 3.2.1.36) from leeches, other parasites, and crustaceans are endo-beta-glucuronidases that generate tetrasaccharide and hexasaccharide end products through hydrolysis of the beta 1-3 linkage.
  • Mammalian hyaluronidases can be further divided into two groups: neutral-active and acid-active enzymes. There are six hyaluronidase-like genes in the human genome, HYAL1, HYAL2, HYAL3, HYAL4, HYALP1 and PH20/SPAM1. HYALP1 is a pseudogene, and HYAL3 has not been shown to possess enzyme activity toward any known substrates. HYAL4 is a chondroitinase and exhibits little activity towards hyaluronan. HYAL1 is the prototypical acid-active enzyme and PH20 is the prototypical neutral-active enzyme.
  • Acid-active hyaluronidases such as HYAL1 and HYAL2 generally lack catalytic activity at neutral pH (i.e. pH 7).
  • HYAL1 has little catalytic activity in vitro over pH 4.5 (Frost I. G. and Stern, R., “A microtiter-based assay for hyaluronidase activity not requiring specialized reagents”, Anal. Biochemistry, 1997; 251:263-269).
  • HYAL2 is an acid-active enzyme with a very low specific activity in vitro.
  • bovine PH20 for example is very loosely attached to the plasma membrane and is not anchored via a phospholipase sensitive anchor (Lalancette et al, Biol Reprod., 2001; 65(2):628-36). This unique feature of bovine hyaluronidase has permitted the use of the soluble bovine testes hyaluronidase enzyme as an extract for clinical use (WydaseTM, HyalaseTM)
  • Other PH20 species are lipid anchored enzymes that are generally not soluble without the use of detergents or lipases.
  • human PH20 is anchored to the plasma membrane via a GPI anchor.
  • sHASEGP When injected in the hypodermis, the depolymerization of HA by sHASEGP is localized to the injection site in the SC tissue.
  • Experimental evidence shows that the sHASEGP is inactivated locally in the interstitial space with a half life of 13 to 20 minutes in mice, without detectable systemic absorption in blood following single intravenous dose in CD-1 mice.
  • sHASEGP Within the vascular compartment sHASEGP demonstrates a half life of 2.3 and 5 minutes in mice and Cynomolgus monkeys, respectively, with doses up to 0.5 mg/kg.
  • Hyaluronidase products of animal origin have been used clinically for over 60 years, primarily to increase the dispersion and absorption of other co-administered drugs and for hypodermoclysis (SC injection/infusion of fluid in large volume) (Frost G. I., “Recombinant human hyaluronidase (rHuPH20): an enabling platform for subcutaneous drug and fluid administration”, Expert Opinion on Drug Delivery, 2007; 4: 427-440).
  • the usual dose used for this purpose is 150 units.
  • hypodermoclysis hyaluronidase is used to aid the subcutaneous administration of relatively large volumes of fluids.
  • 1500 units of hyaluronidase are generally given with each 500 to 1000 ml of fluid for subcutaneous use.
  • 150 units are considered adequate for each liter of hypodermoclysis solution.
  • 150 to 300 units are considered adequate for this purpose.
  • the diffusion of local anesthetics is accelerated by the addition of 1500 units.
  • Germany and the US 150 units are considered adequate for this purpose.
  • the dosage differences notwithstanding (the dosage in the UK is ten times higher than in the US), no apparent differences in the safety profiles of animal-derived hyaluronidase products marketed in the US and UK, respectively, have been reported.
  • Halozyme Therapeutics Inc. received approval from the FDA for an injectable formulation of the recombinant human hyaluronidase, rHuPH20(HYLENEXTM).
  • the FDA approved HYLENEXTM at a dose of 150 units for SC administration of the following indications:
  • rHuPH20 possesses the same properties of enhancing the dispersion and absorption of other injected drugs as the previously approved animal-derived hyaluronidase preparations, but with an improved safety profile.
  • the use of recombinant human hyaluronidase (rHuPH20) compared with animal-derived hyaluronidases minimizes the potential risk of contamination with animal pathogens and transmissible spongiform encephalopathies.
  • Soluble Hyaloronidase glycoproteins (sHASEGP), a process for preparing the same and their use in pharmaceutical compositions have been described in WO 2004/078140.
  • the hyaluronidase enzyme in the formulation in accordance with the present invention is believed to enhance the delivery of the anti-CD20 antibody to the systemic circulation, e.g. by increasing the absorption of the active substance (it acts as a permeation enhancer).
  • the hyaluronidase enzyme is also believed to increases the delivery of the therapeutic anti-CD20 antibody into the systemic circulation via the subcutaneous application route by the reversible hydrolyzation of hyaluronan, an extracellular component of the SC interstitial tissue.
  • the hydrolysis of hyaluronan in the hypodermis temporarily opens channels in the interstitial space of the SC tissue and thereby improves the delivery of the therapeutic anti-CD20 antibody into the systemic circulation.
  • the administration shows reduced pain in humans and less volume-derived swelling of the SC tissue.
  • Hyaluronidase when administered locally has its entire effect locally.
  • hyaluronidase is inactivated and metabolized locally in minutes and has not been noted to have systemic or long term effects.
  • the rapid inactivation of hyaluronidase within minutes when it enters the blood stream precludes a realistic ability to perform comparable biodistribution studies between different hyaluronidase products. This property also minimizes any potential systemic safety concerns because the hyaluronidase product cannot act at distant sites.
  • the unifying feature of all hyaluronidase enzymes in accordance with the present invention is their ability to depolymerize hyaluronan, regardless of differences in chemical structure, in species source, in tissue sources, or in the batches of drug product sourced from the same species and tissue. They are unusual in the fact that their activity is the same (except for potency) in spite of having different structures.
  • the rHuPH20 protein is synthesized with a 35 amino acid signal peptide that is removed from the N-terminus during the process of secretion.
  • the mature rHuPH20 protein contains an authentic N-terminal amino acid sequence orthologous to that found in some bovine hyaluronidase preparations.
  • the PH20 hyaluronidases including the animal derived PH20 and recombinant human rHuPH20, depolymerize hyaluronan by the hydrolysis of the ⁇ -1,4 linkage between the C 1 position of N-acetyl glucosamine and the C 4 position of glucuronic acid.
  • the tetrasaccharide is the smallest digestion product (Weissmann, B., “The transglycosylative action of testicular hyaluronidase”, J. Biol. Chem., 1955; 216: 783-94).
  • rHuPH20(HYLENEXTM) The amino acid sequence of rHuPH20(HYLENEXTM) is well known and available under CAS Registry No. 757971-58-7. The approximate molecular weight is 61 kDa. See, also, U.S. Pat. No. 7,767,429.
  • PH-20 gene is the gene used for the recombinant product rHuPH20; however the recombinant drug product is a 447 amino acid truncated version of the full protein encoded by the PH-20 gene. Structural similarities with respect to amino acid sequences rarely exceed 60% in any comparison. Functional comparisons show that the activity of rHuPH20 is very similar to that of previously approved hyaluronidase products. This information is consistent with the clinical findings during the past 50 years that regardless of the source of the hyaluronidase, the clinical safety and efficacy of units of hyaluronidase are equivalent.
  • the stable pharmaceutical formulation in accordance with the invention is essentially free from visible (human eye inspection) particles.
  • the sub-visible particles should preferably fulfill the following criteria:
  • the present invention provides a method of treating a disease or disorder which is amenable to treatment with an anti-CD20 antibody (e.g. cancer (preferred) or a non-malignant disease) in a subject comprising administering the formulation described herein to a subject in an amount effective to treat the said disease or disorder.
  • an anti-CD20 antibody e.g. cancer (preferred) or a non-malignant disease
  • the cancer or a non-malignant disease will generally involve CD20-expressing cells, such that the CD20 antibody in the therapeutic pharmaceutical SC formulation in accordance with the present invention is able to bind to the affected cells.
  • the cancer is preferably a CD20 expressing cancer.
  • the non-malignant disease that can be treated with the composition in accordance with the present invention is preferably an autoimmune disease as defined herein.
  • the anti-CD20 antibody is co-administered concomitantly or sequentially with a chemotherapeutic agent.
  • the addition of the hyaluronidase to the formulation allows increasing the injection volume which can be safely and comfortably administered subcutaneously.
  • the preferred injection volume is 1 to 15 ml. It has been observed that the administration of the formulation in accordance with the present invention increases the dispersion, absorption and the bioavailability of the therapeutic antibody. Large molecules (i.e. >16 kDa) that are administered via the SC route are preferentially absorbed into the vascular compartment through the draining lymphatic fluids (Supersaxo, A., et al., “Effect of Molecular Weight on the Lymphatic Absorption of Water-Soluble Compounds Following Subcutaneous Administration”, 1990; 2:167-169; Swartz, M.
  • the subcutaneous CD20 antibody (preferably Rituximab) formulation in accordance with the invention requires high antibody concentrations (approx. 120 mg/ml) in the final step of purification of the manufacturing process. Therefore an additional process step (ultrafiltration/diafiltration) is added to the conventional manufacturing process of the CD20 antibody, preferably Rituximab.
  • the highly concentrated, stable pharmaceutical anti-CD20 antibody formulation in accordance with the present invention can also be provided as stabilized protein formulation which can reconstituted with a suitable diluent to generate a high anti-CD20 antibody concentration reconstituted formulation.
  • various in vivo assays are available to the skilled practitioner.
  • a detectable label e.g. a radioactive isotope
  • the CD20 antibody SC formulation in accordance with this invention may also be used to treat various non-malignant diseases or disorders, such a include autoimmune disease as defined herein; endometriosis; scleroderma; restenosis; polyps such as colon polyps, nasal polyps or gastrointestinal polyps; fibroadenoma; respiratory disease; cholecystitis; neurofibromatosis; polycystic kidney disease; inflammatory diseases; skin disorders including psoriasis and dermatitis; vascular disease; conditions involving abnormal proliferation of vascular epithelial cells; gastrointestinal ulcers; Menetrier's disease, secreting adenomas or protein loss syndrome; renal disorders; angiogenic disorders; ocular disease such as age related macular degeneration, presumed ocular histoplasmosis syndrome, retinal neovascularization from proliferative diabetic retinopathy, retinal vascularization, diabetic retinopathy, or age related macular degeneration
  • autoimmune disease
  • microbial infections including microbial pathogens selected from adenovirus, hantaviruses, Borrelia burgdorferi, Yersinia spp. and Bordetella pertussis; thrombus caused by platelet aggregation; reproductive conditions such as endometriosis, ovarian hyperstimulation syndrome, preeclampsia, dysfunctional uterine bleeding, or menometrorrhagia; synovitis; atheroma; acute and chronic nephropathies (including proliferative glomerulonephritis and diabetes-induced renal disease); eczema; hypertrophic scar formation; endotoxic shock and fungal infection; familial adenomatosis polyposis; neurodedenerative diseases (e.g.
  • Treatment with the said antibody formulation will result in an improvement in the signs or symptoms of cancer or disease.
  • such therapy may result in an improvement in survival (overall survival and/or progression free survival) and/or may result in an objective clinical response (partial or complete).
  • treatment with the combination of the chemotherapeutic agent and the antibody formulation may result in a synergistic or greater than additive, therapeutic benefit to the patient.
  • the antibody in the formulation administered is a naked antibody.
  • the antibody administered may be conjugated with a cytotoxic agent.
  • the immunoconjugate and/or antigen to which it is bound is/are internalized by the cell, resulting in increased therapeutic efficacy of the immunoconjugate in killing the cancer cell to which it binds.
  • the cytotoxic agent targets or interferes with nucleic acid in the cancer cell. Examples of such cytotoxic agents include maytansinoids, calioheamicins, ribonucleases and DNA endonucleases.
  • the preferred immunoconjugates are Rituximab-maytansinoid immunoconjugates similarly to Trastuzumab-DM1 (T-DM1) as they are described in WO 2003/037992, more preferably the immunoconjugate T-MCC-DM1.
  • fixed dosages for patients with autoimmune disease such as rheumatoid arthritis, multiple sclerosis, lupus nephritis, diabetes, ITP, and vasculitis are as follows.
  • Preferred are about 1200 mg to about 2200 mg of said anti-CD20 antibody per dose, for example about 1500 mg of said anti-CD20 antibody (e.g. Rituximab) per dose.
  • the stable pharmaceutical formulation of the pharmaceutically active anti-CD20 antibody in accordance with the invention is preferably administered as subcutaneous injection, whereby the administration is preferably repeated several times with time intervals of 3 weeks (q3w). Most preferably the full volume of the injection fluid is administered within a time period of 1 to 10 minutes, preferably 2 to 6 minutes, most preferably 3 ⁇ 1 minutes. Most preferably, 2 ml/minute are administered, i.e. for example approx. 240 mg/min. For many patients where no other intravenous (IV) chemotherapeutic agents are given, such subcutaneous administration leads to increased patient convenience with the potential for self-administration at home.
  • IV intravenous
  • Vepesid® 6-mercaptopurine (6 MP), 6-thiocguanine (6TG), cytarabine (Ara-C), 5-fluorouracil (5-FU), capecitabine (e.g. Xeloda®), dacarbazine (DTIC), and the like; antibiotics, such as actinomycin D, doxorubicin (DXR; e.g. Adriamycin®), daunorubicin (daunomycin), bleomycin, mithramycin and the like; alkaloids, such as vinca alkaloids such as vincristine (VCR), vinblastine, and the like; and other antitumor agents, such as paclitaxel (e.g.
  • Ethyol® dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, lomustine (CCNU), doxorubicin lipo (e.g. Doxil®), gemcitabine (e.g. Gemzar®), daunorubicin lipo (e.g. Daunoxome®), procarbazine, mitomycin, docetaxel (e.g.
  • cytotoxic and anticancer agents described above as well as antiproliferative target-specific anticancer drug like protein kinase inhibitors in chemotherapeutic regimens is generally well characterized in the cancer therapy arts, and their use herein falls under the same considerations for monitoring tolerance and effectiveness and for controlling administration routes and dosages, with some adjustments.
  • the actual dosages of the cytotoxic agents may vary depending upon the patient's cultured cell response determined by using histoculture methods. Generally, the dosage will be reduced compared to the amount used in the absence of additional other agents.
  • Typical dosages of an effective cytotoxic agent can be in the ranges recommended by the manufacturer, and where indicated by in vitro responses or responses in animal models, can be reduced by up to about one order of magnitude concentration or amount.
  • the actual dosage will depend upon the judgment of the physician, the condition of the patient, and the effectiveness of the therapeutic method based on the in vitro responsiveness of the primary cultured malignant cells or histocultured tissue sample, or the responses observed in the appropriate animal models.
  • Radioactive atoms for use in the context of this invention can be selected from the group including, but not limited to, radium, cesium-137, iridium-192, americium-241, gold-198, cobalt-57, copper-67, technetium-99, iodine-123, iodine-131, and indium-111.
  • the antibody Preferably the anti-CD20 antibody treatment is used without such ionizing radiation.
  • the patient may be subjected to surgical removal of cancer cells and/or radiation therapy.
  • an article of manufacture which contains the pharmaceutical formulation of the present invention and provides instructions for its use.
  • This article of manufacture comprises a container.
  • Suitable containers include, for example, bottles, vials (e.g. multiple or dual chamber vials), syringes (such as multiple or dual chamber syringes) and test tubes.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the container holds the formulation and the label on, or associated with, the container may indicate directions for use.
  • the container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g. from 2 to 6 administrations) of the reconstituted formulation.
  • the article of manufacture may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • the antibody which is formulated in accordance with the present invention is preferably essentially pure and desirably essentially homogeneous (i.e. free from contaminating proteins etc, whereby the hyaluronidase enzyme in the formulation in accordance of this invention is not to be considered to be a contaminating protein of the anti-CD20 monoclonal antibody in accordance of the present invention).
  • anti-CD20 formulations for subcutaneous administration were developed based on the experimental results as provided below using the general preparatory and analytical methods and assays as outlined below.
  • Rituximab is manufactured by techniques generally known from the production of recombinant proteins.
  • a genetically engineered Chinese hamster ovary cell line (CHO) prepared as described in U.S. Pat. No. 7,381,560 is expanded in cell culture from a master cell bank.
  • the Rituximab monoclonal antibody is harvested from the cell culture fluid and purified using immobilized Protein A affinity chromatography, cation exchange chromatography, a filtration step to remove viral contaminations, followed by anion exchange chromatography and an ultrafiltration/diafiltration step.
  • rHuPH20 is manufactured by techniques generally known from the production of recombinant proteins. The process begins with thawing of cells from the working cell bank (WCB) or master cell bank (MCB) and expansion through cell culture in a series of spinner flasks. The cell culture up to 6 liters is used to provide a continuous source of cells maintained under selective pressure with methotrexate. When expanded to approximately 36 liters the culture is transferred to a 400 liters bioreactor for a final batch volume of approximately 300 liters. The production bioreactor is operated in the fed-batch mode, with no selection pressure, and the duration of the production phase is approximately two weeks. rHuPH20 is secreted into the culture fluid.
  • WB working cell bank
  • MBB master cell bank
  • a 1000 liters bioreactor can also be used for a final batch volume of 500 liters.
  • the harvest is clarified by filtration, and is then treated with solvent/detergent to inactivate viruses.
  • the protein is then purified by a series of four column chromatography processes to remove process and product related impurities.
  • a viral filtration step is performed, and the filtered bulk is then concentrated, formulated into the final buffer: 10 mg/mL rHuPH20 in 20 mM L-histidine/HCl buffer, pH 6.5, 130 mM NaCl, 0.05% (w/v) polysorbate 80.
  • the rHuPH20 bulk is stored below ⁇ 70° C.
  • Rituximab was buffer exchanged against a diafiltration buffer containing the anticipated buffer composition and where required, concentrated by diafiltration to an antibody concentration of approx. 200 mg/ml.
  • the excipients e.g. trehalose, rHuPH20, surfactant
  • the protein concentration was adjusted with a buffer to the final Rituximab concentration of approx. 120 mg/ml.
  • UV spectroscopy used for determination of protein content, was performed on a Perkin Elmer ⁇ 35 UV spectrophotometer in a wavelength range from 240 nm to 400 nm. Neat protein samples were diluted to approx. 0.5 mg/ml with the corresponding formulation buffer. The protein concentration was calculated according to Equation 1.
  • the UV light absorption at 280 nm was corrected for light scattering at 320 nm and multiplied with the dilution factor, which was determined from the weighed masses and densities of the neat sample and the dilution buffer.
  • the numerator was divided by the product of the cuvette's path length d and the extinction coefficient E.
  • IEC Ion Exchange Chromatography
  • turbidity opalescence was measured in FTU (turbidity units) using a HACH 2100AN turbidimeter at room temperature.
  • Samples were analyzed for visible particles by using a Seidenader V90-T visual inspection instrument.
  • An in vitro enzyme assay of rHuPH20 as hyaluronidase was used as activity assay.
  • the assay is based on the formation of an insoluble precipitate when hyaluronan (sodium hyaluronate) binds to a cationic precipitant.
  • Enzyme activity was measured by incubating rHuPH20 with hyaluronan substrate and then precipitating the undigested hyaluronan with acidified serum albumin (horse serum).
  • the turbidity was measured at a wavelength of 640 nm and the decrease in turbidity resulting from enzyme activity on the hyaluronan substrate is a measure of the enzyme activity.
  • the procedure is run using a standard curve generated with dilutions of rHuPH20 assay reference standard, and sample activity is read from the curve.
  • Formulation A is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM L-histidine, 210 mM trehalose dihydrate, 10 mM methionine, 0.06% polysorbate 80, 2,000 U/ml rHuPH20, at pH 5.5.
  • Formulation B is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM L-histidine, 210 mM trehalose dihydrate, 10 mM methionine, 0.06% polysorbate 80, 2,000 U/ml rHuPH20, at pH 6.1.
  • Formulation C is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM L-histidine, 210 mM trehalose dihydrate, 10 mM methionine, 0.06% polysorbate 80, 12,000 U/ml rHuPH20, at pH 5.5.
  • n.d. 4.2 Essentially free 11869 Thawing from particles 5° C. 7 weeks 124 1.5 98.5 0.0 26 62 5.0 Free from 12206 particles 19 weeks 120 1.5 98.5 0.1 26 62 4.1 Free from 11945 particles 26 weeks n.d. n.d. n.d. n.d. n.d. n.d. n.d. Free from n.d. particles 25° C. 7 weeks n.d. 1.5 97.8 0.7 25 63 5.8 Free from 12259 particles 19 weeks n.d. 1.5 97.3 1.2 24 61 4.8 Free from 13137 particles 26 weeks n.d. 1.8 96.6 1.6 24 60 4.4 Free from 12948 particles 40° C. 7 weeks n.d.
  • Formulation D is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM acetic acid, 210 mM trehalose dihydrate, 10 mM methionine, 0.06% polysorbate 20, 12,000 U/ml rHuPH20, at pH 5.5.
  • n.d. 4.4 Essentially free 12394 Thawing from particles 5° C. 7 weeks 125 1.5 98.4 0.0 26 63 5.0 Essentially free 10030 from particles 19 weeks 123 1.5 98.5 0.1 26 62 4.7 Free from 15324 particles 26 weeks n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. Free from n.d. particles 25° C. 7 weeks n.d. 1.6 97.7 0.7 25 62 4.9 Free from 13099 particles 19 weeks n.d. 1.6 97.2 1.2 24 61 5.1 Free from 13031 particles 26 weeks n.d. n.d. n.d. n.d. n.d. n.d.
  • Formulation E is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM L-histidine, 210 mM trehalose dihydrate, 10 mM methionine, 0.06% polysorbate 20, 12,000 U/ml rHuPH20, at pH 5.5.
  • Formulation F is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM L-histidine, 120 mM sodium chloride, 10 mM methionine, 0.02% polysorbate 80, 12,000 U/ml rHuPH20, at pH 5.5.
  • Formulation G is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM citric acid, 120 mM sodium chloride, 10 mM methionine, 0.02% polysorbate 80, 12,000 U/ml rHuPH20, at pH 6.5.
  • Formulation H is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM citric acid, 210 mM trehalose dihydrate, 10 mM methionine, 0.06% polysorbate 80, 12,000 U/ml rHuPH20, at pH 6.5.
  • Formulation I is a liquid formulation with the composition 120 mg/ml Rituximab, 20 mM L-histidine, 120 mM sodium chloride, 10 mM methionine, 0.04% polysorbate 80, 12,000 U/ml rHuPH20, at pH 6.0.
  • Formulation J is a liquid formulation with the composition 25 mg/ml GA101 (huMAb ⁇ CD20>), 20 mM L-histidine, 240 mM trehalose dihydrate, 0.02% poloxamer 188, 2,000 U/ml rHuPH20, at pH 6.0.
  • recombinant humanized 2H7 anti-CD20 antibody (2H7.v16 as disclosed in WO 2006/084264) was buffer exchanged against a diafiltration buffer containing the anticipated buffer composition and where required, concentrated to an antibody concentration of approx. 60 and 120 mg/ml.
  • the excipients e.g. trehalose, rHuPH20, polysorbate 20
  • the protein concentration was adjusted with the final formulation buffer to a humanized 2H7 concentration of approx. 30, 50, and 100 mg/ml.
  • Protein concentration was determined by ultraviolet absorption spectroscopy using an Agilent 8453 spectrophotometer in a wavelength range from 240 nm to 400 nm. Samples were gravimetrically diluted to approx. 0.5 mg/ml with the corresponding formulation buffer. The protein concentrations were calculated using Equation 1:
  • DF is the dilution factor
  • d is the cuvette path length
  • is the extinction coefficient, which is 1.75 (cm 2 /mg ⁇ 1 ) for 2H7 at A max .
  • the UV light absorption at A max (typically 278 to 280 nm) was corrected for light scattering at 320 nm and multiplied with the dilution factor, which was determined from the weighed masses and densities of the neat sample and the dilution buffer.
  • the numerator was divided by the product of the cuvette's path length d and the extinction coefficient ⁇ .
  • SEC Size Exclusion Chromatography
  • IEC Ion Exchange Chromatography
  • Acidic and basic variants were separated using a linear gradient of 25 mM potassium phosphate at pH 6.9 (mobile phase A) and 120 mM potassium chloride dissolved in 25 mM potassium phosphate (mobile phase B) with a flow rate of 0.5 mL/min.
  • the complement-dependent cytotoxicity assay (CDC) assay was performed to determine the in vitro activity of the anti-CD20 antibody.
  • the complement dependent cytotoxicity (CDC) potency assay is used to measure the ability of the antibody to lyse human B lymphoblastoid (WIL2-S) cells in the presence of human complement.
  • the assay is performed in 96 well tissue culture microtiter plates. In this assay, varying concentrations of the anti-CD20 antibody reference material, control, or sample(s) diluted in assay diluent are incubated with WIL2-S cells (50,000 cells/well) in the presence of a fixed amount of human complement. The plate is incubated at 37° C./5% CO2 in a humidified incubator for 1 to 2 hours.
  • ALAMARBLUETM is a redox dye that fluoresces at an excitation wavelength of 530 nm and an emission wavelength of 590 nm when reduced by live cells. Therefore, the changes in color and fluorescence are proportional to the number of viable cells.
  • the results, expressed in relative fluorescence units (RFU), are plotted against the anti-CD20 antibody concentrations and a parallel line program is used to estimate the activity of anti-CD20 antibody samples relative to the reference material.
  • a turbidimetric assay was used to determine hyaluronidase activity and enzyme concentration. This method is based on the formation of an insoluble precipitate when hyaluronic acid binds with acidified serum albumin. Briefly, a dilution series of the rhuPH20 hyaluronidase (Halozyme, Inc.) working reference standard ranging from 2.5 U/ml to 0.25 U/ml is prepared in enzyme diluent (70 mM NaCl, 25 mM PIPES, pH 5.5, 0.66 mg/ml gelatin hydrolysate, 0.1% human serum albumin). The test samples are diluted to a final concentration of 1.5 U/ml in enzyme diluent.
  • the turbidity of the reaction is measured at a wavelength 640 nm on a microplate reader.
  • the decrease in turbidity resulting from enzyme activity on the hyaluronic acid substrate is a measure of the hyaluronidase activity.
  • the sample activity is determined relative to the calibration curve generated with the dilutions of the rhuPH20 working reference standard.
  • Formulation K is a liquid formulation with the composition 30 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 0 U/ml of rhuPH20 at pH 5.3.
  • Formulation L is a liquid formulation with the composition 30 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 1500 U/ml of rhuPH20 at pH 5.3.
  • Formulation M is a liquid formulation with the composition 30 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 12,000 U/ml of rhuPH20 at pH 5.3.
  • Formulation N is a liquid formulation with the composition 50 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 0 U/ml of rhuPH20 at pH 5.3.
  • Formulation O is a liquid formulation with the composition 50 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 1500 U/ml of rhuPH20 at pH 5.3.
  • Formulation P is a liquid formulation with the composition 50 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 12,000 U/ml of rhuPH20 at pH 5.3.
  • Formulation Q is a liquid formulation with the composition 100 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 0 U/ml of rhuPH20 at pH 5.3.
  • Formulation R is a liquid formulation with the composition 100 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 1500 U/ml of rhuPH20 at pH 5.3.
  • Formulation S is a liquid formulation with the composition 100 mg/ml humanized 2H7, 30 mM sodium acetate, 8% trehalose dihydrate, 0.02% polysorbate 20, 12,000 U/ml of rhuPH20 at pH 5.3.
  • Rituximab-containing regimens have become the standard of care for patients suffering from various CD20-positive B-cell malignancies.
  • rituximab is administered as an intravenous (IV) infusion over several hours. These long infusion times and the side effects related to the infusion were cited by some patients as uncomfortable consequences of the current therapeutic treatment.
  • IV intravenous
  • SC Subcutaneous
  • rHuPH20 Recombinant human hyaluronidase
  • This example provides stage 1 data from a randomized, open-label, multi-centre adaptive Phase Ib study.
  • 124 patients were randomized to one of four rituximab maintenance treatment groups: 16 patients IV control, 34 patients SC dose 1 (375 mg/m 2 ), 34 patients SC dose 2 (625 mg/m 2 ) and 40 patients SC dose 3 (800 mg/m 2 ).
  • eligible patients Prior to randomization, eligible patients were treated with at least one IV rituximab dose at 375 mg/m 2 in the maintenance setting.
  • a single IV dose was replaced by a SC dose.
  • Patients received rituximab either on an every 2 month (q2m) or every 3 month (q3m) regimen, as per local practice.
  • the total volume administered SC in each patient ranged between 4.4-15.0 mL.
  • the average injection duration was 2 mL/min.
  • Rituximab maximum serum concentrations in the SC cohorts occurred between Day 2 and Day 8 (48 h and 168 h).
  • Pharmacokinetic parameters were linear with respect to dose over the range of SC doses administered (375, 625 and 800 mg/m 2 ).
  • Rituximab concentrations on Day 28 (C28) and the extent of serum exposure (AUC 0-57 ) in patients administered 625 mg/m 2 rituximab SC were comparable to those in patients administered the standard rituximab IV dose of 375 mg/m 2 SC.
  • subcutaneous rituximab can be delivered quickly, comfortably and safely while achieving serum exposure comparable to the approved intravenous formulation in FL patients during maintenance treatment.
  • the patient experience was favourable.
  • rituximab SC formulation prepared according to Example 1, Formulation A
  • rituximab IV in combination with CHOP or CVP
  • Patients will be randomized to receive 375 mg/m 2 Rituximab as intravenous infusion or 1400 mg Rituximab given subcutaneously.
  • patients will receive standard chemotherapy (CVP or CHOP).
  • CVP or CHOP standard chemotherapy
  • Patients who achieved a complete or partial response after 8 treatment cycles, will receive maintenance treatment for a further maximum number of 12 cycles. Maintenance treatment cycles will be repeated every 8 weeks. The anticipated time on study treatment is 96 weeks.
  • Treatment with 1400 mg SQ Rituximab anti-CD20 antibody as a maintenance treatment every 8 weeks for up to 12 cycles is expected to be safe and efficacious in treating follicular lymphoma, optionally in combination with chemotherapy (including CHOP or CVP).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Dermatology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Endocrinology (AREA)
  • Inorganic Chemistry (AREA)
  • Hematology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US12/879,486 2009-09-11 2010-09-10 Highly Concentrated Pharmaceutical Formulations Abandoned US20110076273A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/260,558 US10280227B2 (en) 2009-09-11 2014-04-24 Highly concentrated pharmaceutical formulations
US14/944,508 US10377831B2 (en) 2009-09-11 2015-11-18 Highly concentrated pharmaceutical formulations
US16/450,906 US10752696B2 (en) 2009-09-11 2019-06-24 Highly concentrated pharmaceutical formulations
US16/943,378 US20210054092A1 (en) 2009-09-11 2020-07-30 Highly concentrated pharmaceutical formulations

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP09170110.2 2009-09-11
EP09170110 2009-09-11

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/260,558 Continuation US10280227B2 (en) 2009-09-11 2014-04-24 Highly concentrated pharmaceutical formulations

Publications (1)

Publication Number Publication Date
US20110076273A1 true US20110076273A1 (en) 2011-03-31

Family

ID=43732866

Family Applications (5)

Application Number Title Priority Date Filing Date
US12/879,486 Abandoned US20110076273A1 (en) 2009-09-11 2010-09-10 Highly Concentrated Pharmaceutical Formulations
US14/260,558 Active US10280227B2 (en) 2009-09-11 2014-04-24 Highly concentrated pharmaceutical formulations
US14/944,508 Active US10377831B2 (en) 2009-09-11 2015-11-18 Highly concentrated pharmaceutical formulations
US16/450,906 Active US10752696B2 (en) 2009-09-11 2019-06-24 Highly concentrated pharmaceutical formulations
US16/943,378 Abandoned US20210054092A1 (en) 2009-09-11 2020-07-30 Highly concentrated pharmaceutical formulations

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/260,558 Active US10280227B2 (en) 2009-09-11 2014-04-24 Highly concentrated pharmaceutical formulations
US14/944,508 Active US10377831B2 (en) 2009-09-11 2015-11-18 Highly concentrated pharmaceutical formulations
US16/450,906 Active US10752696B2 (en) 2009-09-11 2019-06-24 Highly concentrated pharmaceutical formulations
US16/943,378 Abandoned US20210054092A1 (en) 2009-09-11 2020-07-30 Highly concentrated pharmaceutical formulations

Country Status (42)

Country Link
US (5) US20110076273A1 (zh)
EP (5) EP4218716A1 (zh)
JP (4) JP5674788B2 (zh)
KR (3) KR101782195B1 (zh)
CN (2) CN104399075B (zh)
AR (1) AR078161A1 (zh)
AU (1) AU2010294186B2 (zh)
BR (1) BR112012005017B8 (zh)
CA (1) CA2771571C (zh)
CL (1) CL2012000620A1 (zh)
CO (1) CO6440527A2 (zh)
CR (1) CR20120090A (zh)
CY (1) CY1117941T1 (zh)
DK (2) DK3061445T3 (zh)
DO (2) DOP2012000064A (zh)
EA (2) EA201792552A1 (zh)
EC (1) ECSP12011722A (zh)
ES (2) ES2953478T3 (zh)
FI (1) FI3061445T3 (zh)
GT (1) GT201200069A (zh)
HK (2) HK1174553A1 (zh)
HR (2) HRP20231019T1 (zh)
HU (2) HUE062860T2 (zh)
IL (2) IL218165A (zh)
LT (1) LT3061445T (zh)
MA (1) MA33592B1 (zh)
MX (3) MX360432B (zh)
MY (2) MY174009A (zh)
NZ (2) NZ621448A (zh)
PE (2) PE20121178A1 (zh)
PH (1) PH12016501002A1 (zh)
PL (2) PL3061445T3 (zh)
PT (1) PT3061445T (zh)
RS (1) RS54769B1 (zh)
SG (1) SG179030A1 (zh)
SI (2) SI3061445T1 (zh)
SM (1) SMT201600176B (zh)
TN (1) TN2012000101A1 (zh)
TW (2) TWI515020B (zh)
UA (1) UA104909C2 (zh)
WO (1) WO2011029892A2 (zh)
ZA (1) ZA201201605B (zh)

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090010921A1 (en) * 2003-11-05 2009-01-08 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2013036297A1 (en) * 2011-09-06 2013-03-14 Selecta Biosciences, Inc. Therapeutic protein-specific induced tolerogenic dendritic cells and methods of use
WO2013173687A1 (en) 2012-05-18 2013-11-21 Genentech, Inc. High-concentration monoclonal antibody formulations
US8652487B2 (en) 2011-04-29 2014-02-18 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
WO2014165280A1 (en) * 2013-03-12 2014-10-09 Teva Pharmaceutical Industries Ltd. Rituximab induction therapy followed by glatiramer acetate therapy
WO2015057910A1 (en) * 2013-10-16 2015-04-23 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US20150239977A1 (en) * 2012-09-27 2015-08-27 Massachusetts Institute Of Technology Cd20- and egfr-binding proteins with enhanced stability
WO2015191969A1 (en) * 2014-06-13 2015-12-17 Mayo Foundation For Medical Education And Research Treating lymphomas
CN105189559A (zh) * 2013-03-15 2015-12-23 塔科达有限责任公司 抗体制剂及其用途
US9757453B2 (en) 2012-10-01 2017-09-12 Mayo Foundation For Medical Education And Research Nanoparticle complexes of anti-CD20 antibodies, albumin and paclitaxel
US9777067B2 (en) 2012-09-27 2017-10-03 Massachusetts Institute Of Technology HER2- and VEGF-A-binding proteins with enhanced stability
US9968676B2 (en) 2009-07-31 2018-05-15 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
US9981021B1 (en) 2015-04-09 2018-05-29 Kinetiq, Inc. Subcutaneous therapeutic enzyme formulations, uses, and methods for generating thereof
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
US10213513B2 (en) 2014-06-16 2019-02-26 Mayo Foundation For Medical Education And Research Treating myelomas
US10280227B2 (en) 2009-09-11 2019-05-07 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10287344B2 (en) 2008-07-03 2019-05-14 Mayo Foundation For Medical Education And Research Methods for reducing global chronic inflammation and the presence of melanoma
US10300016B2 (en) 2014-10-06 2019-05-28 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US20190282674A1 (en) * 2014-07-16 2019-09-19 New York University Use of hyaluronidase for treatment of muscle stiffness
CN110559435A (zh) * 2013-09-11 2019-12-13 伊戈尔生物药品股份有限公司 包含离子性液体的液体蛋白质制剂
US10561726B2 (en) 2015-10-06 2020-02-18 Vavotar Life Sciences LLC Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US10568951B2 (en) 2013-11-18 2020-02-25 Formycon Ag Pharmaceutical composition of an anti-VEGF antibody
US10618969B2 (en) 2016-04-06 2020-04-14 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US10646569B2 (en) 2017-05-16 2020-05-12 Bhami's Research Laboratory, Pvt. Ltd. High concentration protein formulations with reduced viscosity
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
US10702606B2 (en) 2015-12-10 2020-07-07 Menicon Co., Ltd. Peptide composition
US10765741B2 (en) 2011-05-09 2020-09-08 Mayo Foundation For Medical Education And Research Methods for treating VEGF-expressing cancer using preformed nanoparticle complexes comprising albumin-bound paclitaxel and bevacizumab
US10781261B2 (en) 2015-11-03 2020-09-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US10793630B2 (en) 2014-12-04 2020-10-06 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute myeloid leukemia
US10800851B2 (en) 2014-02-28 2020-10-13 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
EP3370770B1 (en) 2015-11-03 2021-01-20 Janssen Biotech, Inc. Subcutaneous formulations of anti-cd38 antibodies and their uses
CN112533947A (zh) * 2018-07-03 2021-03-19 加拉帕戈斯股份有限公司 高浓度液体抗体制剂
US11021543B2 (en) 2015-06-24 2021-06-01 Janssen Biotech, Inc. Immune modulation and treatment of solid tumors with antibodies that specifically bind CD38
US11160876B2 (en) 2016-09-01 2021-11-02 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
US11241387B2 (en) 2015-08-18 2022-02-08 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US11305020B2 (en) 2016-03-21 2022-04-19 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US11311631B2 (en) 2016-09-06 2022-04-26 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
US11351254B2 (en) 2016-02-12 2022-06-07 Mayo Foundation For Medical Education And Research Hematologic cancer treatments
US11400109B2 (en) * 2016-11-10 2022-08-02 Translate Bio, Inc. Subcutaneous delivery of messenger RNA
US11427637B2 (en) 2016-09-06 2022-08-30 Mayo Foundation For Medical Education And Research Methods of treating PD-L1 expressing cancer
US11426451B2 (en) 2017-03-11 2022-08-30 Selecta Biosciences, Inc. Methods and compositions related to combined treatment with antiinflammatories and synthetic nanocarriers comprising an immunosuppressant
US11548946B2 (en) 2016-09-01 2023-01-10 Mayo Foundation For Medical Education And Research Carrier-PD-L1 binding agent compositions for treating cancers
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
US11590098B2 (en) 2016-09-06 2023-02-28 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
US11713355B2 (en) 2014-02-28 2023-08-01 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
US11878061B2 (en) 2016-03-21 2024-01-23 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RS59913B1 (sr) 2008-10-17 2020-03-31 Sanofi Aventis Deutschland Kombinacija insulina i glp-1-agonista
BR112012011403B8 (pt) 2009-11-13 2021-05-25 Sanofi Aventis Deutschland composição farmacêutica líquida compreendendo um agonista glp-1 e metionina e uso da mesma
KR101972301B1 (ko) 2009-11-13 2019-04-25 사노피-아벤티스 도이칠란트 게엠베하 Glp-1 작용제, 인슐린 및 메티오닌을 포함하는 약제학적 조성물
CA2809321C (en) 2010-08-30 2018-08-14 Sanofi-Aventis Deutschland Gmbh Use of ave0010 for the manufacture of a medicament for the treatment of diabetes mellitus type 2
US9821032B2 (en) 2011-05-13 2017-11-21 Sanofi-Aventis Deutschland Gmbh Pharmaceutical combination for improving glycemic control as add-on therapy to basal insulin
HUE027989T2 (en) 2011-08-29 2016-11-28 Sanofi Aventis Deutschland A pharmaceutical composition for use in glycemic control in patients with type 2 diabetes
AR087744A1 (es) 2011-09-01 2014-04-16 Sanofi Aventis Deutschland Composicion farmaceutica para uso en el tratamiento de una enfermedad neurodegenerativa
PT2771031T (pt) 2011-10-28 2018-05-24 Prothena Biosciences Ltd Anticorpos humanizados que reconhecem a alfa-sinucleína
BR112014018561A8 (pt) 2012-01-27 2017-07-11 Neotope Biosciences Ltd Anticorpo; ácido nucleico; célula hospedeira; composição farmacêutica; uso de um anticorpo; método para produção de um anticorpo; e método para produção de uma linhagem celular que produz um anticorpo
UA118441C2 (uk) 2012-10-08 2019-01-25 Протена Біосаєнсиз Лімітед Антитіло, що розпізнає альфа-синуклеїн
EP2727602A1 (en) * 2012-10-31 2014-05-07 Takeda GmbH Method for preparation of a high concentration liquid formulation of an antibody
AU2013359550A1 (en) * 2012-12-12 2015-07-23 Teva Pharmaceutical Industries Ltd. Fusion of human growth hormone and albumin, formulation and uses thereof
EP2945703A4 (en) 2013-01-15 2016-08-31 Teva Pharma ALBU-BCHE FORMULATIONS, PREPARATION THEREOF AND USES THEREOF
TWI780236B (zh) 2013-02-04 2022-10-11 法商賽諾菲公司 胰島素類似物及/或胰島素衍生物之穩定化醫藥調配物
SG10201913932VA (en) * 2013-03-13 2020-03-30 Genentech Inc Antibody formulations
US10513555B2 (en) * 2013-07-04 2019-12-24 Prothena Biosciences Limited Antibody formulations and methods
WO2015104314A1 (en) 2014-01-09 2015-07-16 Sanofi Stabilized pharmaceutical formulations of insulin analogues and/or insulin derivatives
AU2015205620A1 (en) 2014-01-09 2016-07-14 Sanofi Stabilized pharmaceutical formulations of insulin aspart
AU2015205621A1 (en) 2014-01-09 2016-07-14 Sanofi Stabilized glycerol free pharmaceutical formulations of insulin analogues and/or insulin derivatives
BR112017004393A2 (pt) * 2014-09-15 2018-02-27 Genentech Inc formulações de anticorpo
SI3229828T1 (sl) 2014-12-12 2023-06-30 Sanofi-Aventis Deutschland Gmbh Formulacija s fiksnim razmerjem inzulin glargin/liksisenatid
TWI748945B (zh) 2015-03-13 2021-12-11 德商賽諾菲阿凡提斯德意志有限公司 第2型糖尿病病患治療
TW201705975A (zh) 2015-03-18 2017-02-16 賽諾菲阿凡提斯德意志有限公司 第2型糖尿病病患之治療
CN104761645A (zh) * 2015-04-30 2015-07-08 山东禹王制药有限公司 α干扰素融合蛋白制剂及应用
DE102015107307B3 (de) * 2015-05-11 2016-09-08 Labor L + S Aktiengesellschaft Optisches Verfahren zur Gehaltsbestimmung von Hyaluronsäure
WO2016187546A1 (en) * 2015-05-20 2016-11-24 Janssen Biotech, Inc. Anti-cd38 antibodies for treatment of light chain amyloidosis and other cd38-positive hematological malignancies
EP3124976B1 (en) * 2015-07-28 2018-09-12 F. Hoffmann-La Roche AG Improved bacterial endotoxin test for the determination of endotoxins
US10159660B2 (en) 2016-07-29 2018-12-25 Oncternal Therapeutics, Inc. Uses of indolinone compounds
IL301948B1 (en) * 2016-08-29 2024-05-01 Tiziana Life Sciences Plc Anti-CD3 antibody formulations
CN110869050A (zh) * 2017-07-28 2020-03-06 豪夫迈·罗氏有限公司 双特异性抗体配制剂
CN107596349A (zh) * 2017-10-26 2018-01-19 刘冬连 一种稳定的红细胞生成素制剂配方
CN107898756B (zh) * 2017-12-22 2020-11-20 东曜药业有限公司 一种用于皮下或肌肉注射的高浓度尼妥珠单抗制剂及其制备方法和应用
AU2019339740A1 (en) * 2018-09-13 2021-04-01 F. Hoffmann-La Roche Ag Csf-1r antibody formulation
CN111202845A (zh) * 2018-11-22 2020-05-29 上海医药集团股份有限公司 包含抗cd20抗体的药物配制剂及其制备方法和应用
JP2023507053A (ja) * 2019-10-12 2023-02-21 バイオ-セラ ソリューションズ リミテッド 抗cd20抗体製剤及びcd20陽性疾患の治療のための抗cd20抗体の使用
AU2020399619A1 (en) * 2019-12-09 2022-06-09 Genentech, Inc. Anti-PD-L1 antibody formulations
CU20190104A7 (es) 2019-12-17 2021-08-06 Ct Inmunologia Molecular Formulación estable del anticuerpo nimotuzumab
WO2022141232A1 (zh) * 2020-12-30 2022-07-07 上海宝济药业有限公司 一种重组人透明质酸酶制剂及其应用
EP4326288A1 (en) 2021-04-21 2024-02-28 Indapta Therapeutics, Inc. Methods of treatment and dosing of natural killer cell compositions
CN113855798A (zh) * 2021-09-30 2021-12-31 佛山汉腾生物科技有限公司 抗cd20的抗体制剂
WO2023204554A1 (ko) * 2022-04-20 2023-10-26 주식회사 알토스바이오로직스 오크렐리주맙을 포함하는 약학적 조성물과 그의 용도
WO2024007020A1 (en) 2022-06-30 2024-01-04 Indapta Therapeutics, Inc. Combination of engineered natural killer (nk) cells and antibody therapy and related methods
WO2024025440A1 (en) * 2022-07-26 2024-02-01 Joint Stock Company "Biocad" Pharmaceutical composition of anti-cd20 antibody and use thereof

Citations (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5776456A (en) * 1992-11-13 1998-07-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US6127526A (en) * 1996-11-27 2000-10-03 Genentech, Inc. Protein purification by Protein A chromatography
US6171586B1 (en) * 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US20010018041A1 (en) * 1999-11-08 2001-08-30 Idec Pharmaceuticals Corporation Treatment of B cell malignancies using anti-CD40L antibodies in combination with anti-CD20 antibodies and/or chemotherapeutics and radiotherapy
US20020006404A1 (en) * 1999-11-08 2002-01-17 Idec Pharmaceuticals Corporation Treatment of cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
US20020009444A1 (en) * 2000-04-25 2002-01-24 Idec Pharmaceuticals Corporation Intrathecal administration of rituximab for treatment of central nervous system lymphomas
US20020012665A1 (en) * 2000-03-31 2002-01-31 Nabil Hanna Combined use of anti-cytokine antibodies or antagonists and anti-CD20 for treatment of B cell lymphoma
US20020028178A1 (en) * 2000-07-12 2002-03-07 Nabil Hanna Treatment of B cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
US20020039557A1 (en) * 2000-06-20 2002-04-04 Christine White Treatment of B-cell associated diseases such as malignances and autoimmune diseases using a cold anti-CD20 antibody/radiolabeled anti-CD22 antibody combination
US20020058029A1 (en) * 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
US6417335B1 (en) * 1998-05-06 2002-07-09 Genentech, Inc. Protein purification
US20020128448A1 (en) * 2000-10-20 2002-09-12 Idec Pharmaceuticals Corporation Variant IgG3 Rituxan and therapeutic use thereof
US6455043B1 (en) * 1998-08-11 2002-09-24 Idec Pharmaceuticals Corporation Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibody
US20020159996A1 (en) * 2001-01-31 2002-10-31 Kandasamy Hariharan Use of CD23 antagonists for the treatment of neoplastic disorders
US20030103971A1 (en) * 2001-11-09 2003-06-05 Kandasamy Hariharan Immunoregulatory antibodies and uses thereof
US20030157108A1 (en) * 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20030180290A1 (en) * 1995-06-07 2003-09-25 Idec Pharmaceuticals Corporation Anti-CD80 antibody having ADCC activity for ADCC mediated killing of B cell lymphoma cells alone or in combination with other therapies
US20030180292A1 (en) * 2002-03-14 2003-09-25 Idec Pharmaceuticals Treatment of B cell malignancies using anti-CD40L antibodies in combination with anti-CD20 antibodies and/or chemotherapeutics and radiotherapy
US20040191256A1 (en) * 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US20040202658A1 (en) * 2003-04-09 2004-10-14 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to TNF-alpha inhibitor
US20040213784A1 (en) * 1998-11-09 2004-10-28 Idec Pharmaceuticals Corporation Use of chimeric anti-CD20 antibody as in vitro or in vivo purging agent in patients receiving BMT or PBSC transplant
US20050032130A1 (en) * 2003-07-29 2005-02-10 Genentech, Inc. Neutralizing antibody assay and uses therefor
US20050053602A1 (en) * 2003-08-29 2005-03-10 Genentech, Inc. Therapy of ocular disorders
US20050095243A1 (en) * 2003-06-05 2005-05-05 Genentech, Inc. Combination therapy for B cell disorders
US6893625B1 (en) * 1986-10-27 2005-05-17 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
US20050123546A1 (en) * 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US20050158316A1 (en) * 1997-06-13 2005-07-21 Genentech, Inc. Antibody formulation
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
US20050186206A1 (en) * 2003-12-19 2005-08-25 Genentech, Inc. Detection of CD20 in therapy of autoimmune diseases
US20050191297A1 (en) * 2003-12-19 2005-09-01 Genentech, Inc. Detection of CD20 in transplant rejection
US20060002930A1 (en) * 2004-04-16 2006-01-05 Genentech, Inc. Treatment of disorders
US20060024300A1 (en) * 2002-12-16 2006-02-02 Genentech, Inc. Immunoglobulin variants and uses thereof
US20060024295A1 (en) * 2004-06-04 2006-02-02 Genentech, Inc. Method for treating lupus
US20060051345A1 (en) * 2004-06-04 2006-03-09 Genentech, Inc. Method for treating multiple sclerosis
US20060062787A1 (en) * 2004-07-22 2006-03-23 Genentech, Inc. Method for treating Sjogren's syndrome
US20060067930A1 (en) * 2004-08-19 2006-03-30 Genentech, Inc. Polypeptide variants with altered effector function
US20060088523A1 (en) * 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulations
US20060099662A1 (en) * 2004-04-16 2006-05-11 Genentech, Inc. Assay for antibodies
US20060110387A1 (en) * 2004-10-05 2006-05-25 Genentech, Inc. Method for treating vasculitis
US20060134111A1 (en) * 2004-12-17 2006-06-22 Genentech, Inc. Antiangiogenesis therapy of autoimmune disease in patients who have failed prior therapy
US20060172385A1 (en) * 2004-12-22 2006-08-03 Genentech, Inc. Methods for producing soluble membrane-spanning proteins
US20060171950A1 (en) * 2001-01-31 2006-08-03 Biogen Idec Inc. Use of cd23 antagonists for the treatment of neoplastic disorders
US20060179501A1 (en) * 2002-12-16 2006-08-10 Chan Andrew C Transgenic mice expressing human cd20
US20060188495A1 (en) * 2005-01-13 2006-08-24 Genentech, Inc. Treatment method
US20060233797A1 (en) * 2005-04-15 2006-10-19 Genentech, Inc. Treatment of inflammatory bowel disease (IBD)
US20060240007A1 (en) * 2005-04-22 2006-10-26 Genentech, Inc. Method for treating dementia or Alzheimer's disease
US20070009519A1 (en) * 2001-01-31 2007-01-11 Biogen Idec Inc. Immunoregulatory Antibodies and Uses Thereof
US20070009518A1 (en) * 2005-05-18 2007-01-11 Biogen Idec Inc. Methods for treating fibrotic conditions
US20070071675A1 (en) * 2005-08-19 2007-03-29 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20070212733A1 (en) * 2005-11-23 2007-09-13 Genentech, Inc. Methods and compositions related to B cell assays
US20080044421A1 (en) * 2004-09-08 2008-02-21 Genentech, Inc. Methods of using death receptor ligands and CD20 antibodies
US20080075719A1 (en) * 2004-04-16 2008-03-27 Genentech, Inc. Method for Augmenting B Cell Depletion
US20090004189A1 (en) * 2007-06-18 2009-01-01 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
US20090053786A1 (en) * 2007-07-09 2009-02-26 Yung-Hsiang Kao Prevention of disulfide bond reduction during recombinant production of polypeptides
US20090060913A1 (en) * 2007-09-05 2009-03-05 Thomas Friess Combination therapy with type i and type ii anti-cd20 antibodies
US7517670B2 (en) * 1998-04-20 2009-04-14 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US20090098118A1 (en) * 2007-10-15 2009-04-16 Thomas Friess Combination therapy of a type ii anti-cd20 antibody with an anti-bcl-2 active agent
US20090110688A1 (en) * 2007-10-24 2009-04-30 Georg Fertig Combination therapy of type ii anti-cd20 antibody with a proteasome inhibitor
US20090148435A1 (en) * 2007-10-30 2009-06-11 Genentech, Inc. Antibody purification by cation exchange chromatography
US20090162352A1 (en) * 2007-12-21 2009-06-25 Michael Adler Antibody formulation
US20090169550A1 (en) * 2007-12-21 2009-07-02 Genentech, Inc. Therapy of rituximab-refractory rheumatoid arthritis patients
US20090175854A1 (en) * 2004-09-08 2009-07-09 Genentech, Inc. Methods of using death receptor ligands and CD20 antibodies
US20090214561A1 (en) * 2007-09-24 2009-08-27 David Robert Close Treatment method
US20100158903A1 (en) * 2008-09-16 2010-06-24 Craig Smith Methods for treating progressive multiple sclerosis
US7767429B2 (en) * 2003-03-05 2010-08-03 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof

Family Cites Families (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2795529A (en) 1954-06-17 1957-06-11 American Home Prod Stabilized hyaluronidase solution containing calcium chloride
CU22545A1 (es) 1994-11-18 1999-03-31 Centro Inmunologia Molecular Obtención de un anticuerpo quimérico y humanizado contra el receptor del factor de crecimiento epidérmico para uso diagnóstico y terapéutico
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4753894A (en) 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
JP2547556B2 (ja) * 1987-02-06 1996-10-23 株式会社 ミドリ十字 r−グロブリンの液状製剤
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
JP3040121B2 (ja) 1988-01-12 2000-05-08 ジェネンテク,インコーポレイテッド 増殖因子レセプターの機能を阻害することにより腫瘍細胞を処置する方法
US5721348A (en) 1990-12-14 1998-02-24 University Of Connecticut DNA encoding PH-20 proteins
JP3854306B2 (ja) 1991-03-06 2006-12-06 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング ヒト化及びキメラモノクローナル抗体
JPH04346934A (ja) * 1991-05-24 1992-12-02 Green Cross Corp:The γ−グロブリンの液状製剤
WO1992022653A1 (en) 1991-06-14 1992-12-23 Genentech, Inc. Method for making humanized antibodies
WO1993021319A1 (en) 1992-04-08 1993-10-28 Cetus Oncology Corporation HUMANIZED C-erbB-2 SPECIFIC ANTIBODIES
EP0655924A4 (en) 1992-06-30 1996-09-11 Oncologix Inc A COMBINATION OF MONOCLONAL ANTIBODIES AGAINST erbB-2 AND METHOD FOR THEIR USE.
US7744877B2 (en) 1992-11-13 2010-06-29 Biogen Idec Inc. Expression and use of anti-CD20 Antibodies
GB9401182D0 (en) 1994-01-21 1994-03-16 Inst Of Cancer The Research Antibodies to EGF receptor and their antitumour effect
CA2222231A1 (en) 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
ES2435462T3 (es) * 1995-07-27 2013-12-19 Genentech, Inc. Formulación de proteína liofilizada isotónica estable
US5783186A (en) 1995-12-05 1998-07-21 Amgen Inc. Antibody-induced apoptosis
EP0852951A1 (de) 1996-11-19 1998-07-15 Roche Diagnostics GmbH Stabile lyophilisierte pharmazeutische Zubereitungen von mono- oder polyklonalen Antikörpern
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
AU740284B2 (en) 1997-06-13 2001-11-01 Genentech Inc. Stabilized antibody formulation
KR20010071271A (ko) 1998-05-15 2001-07-28 존 비. 랜디스 방사선 및 성장 인자 수용체 티로신 키나아제 억제제에의한 사람 종양의 치료법
KR20010103655A (ko) 1998-11-09 2001-11-23 케네쓰 제이. 울코트 키메라 항-cd20항체를 이용한 순환성 종양세포와관련된 혈액학적 악성종양의 치료법
CN101121021B (zh) 1999-06-25 2011-12-07 杰南技术公司 人源化抗ErbB2抗体及用抗ErbB2抗体进行的治疗
NZ520622A (en) 2000-02-25 2004-08-27 Us Gov Health & Human Serv Anti-EGFRvIII scFvs with improved cytotoxicity and yield, immunotoxins based thereon, and methods of use thereof
US6632979B2 (en) 2000-03-16 2003-10-14 Genentech, Inc. Rodent HER2 tumor model
CN1239701C (zh) 2000-05-19 2006-02-01 梨树化学株式会社 抗表皮生长因子受体的人源化抗体
US8632778B2 (en) 2000-08-11 2014-01-21 Chugai Seiyaku Kabushiki Kaisha Stabilized anti-interleukin-6 antibody-containing preparations
DK2314686T4 (da) 2000-10-06 2023-08-21 Kyowa Kirin Co Ltd Celler, der danner antistofsammensætninger
PT1324776E (pt) 2000-10-12 2009-12-23 Genentech Inc Formulações de proteína concentradas de viscosidade reduzida
US8703126B2 (en) 2000-10-12 2014-04-22 Genentech, Inc. Reduced-viscosity concentrated protein formulations
WO2002078766A2 (en) 2001-04-02 2002-10-10 Genentech, Inc. Combination therapy
JP4298498B2 (ja) 2001-06-13 2009-07-22 ゲンマブ エー/エス 上皮成長因子受容体(egfr)に対するヒトモノクローナル抗体
DE10153197A1 (de) 2001-10-27 2003-05-08 Merck Patent Gmbh Pigment mit Metallglanz
DK1475101T3 (da) 2002-02-14 2011-01-10 Chugai Pharmaceutical Co Ltd Antistof-holdige farmaceutiske opløsninger
US20050276812A1 (en) 2004-06-01 2005-12-15 Genentech, Inc. Antibody-drug conjugates and methods
PT3284753T (pt) 2002-10-17 2020-05-04 Genmab As Anticorpos monoclonais humanos contra cd20 para utilização no tratamento da esclerose múltipla
JP5425365B2 (ja) 2003-01-22 2014-02-26 グリカート バイオテクノロジー アクチェンゲゼルシャフト 増加したFcレセプター結合親和性およびエフェクター機能を有する抗体を作製するための融合構築物およびその使用
US20060104968A1 (en) * 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US20090123367A1 (en) 2003-03-05 2009-05-14 Delfmems Soluble Glycosaminoglycanases and Methods of Preparing and Using Soluble Glycosaminoglycanases
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
ZA200507757B (en) 2003-04-04 2007-01-31 Genentech Inc High concentration antibody and protein formulations
US20050158303A1 (en) 2003-04-04 2005-07-21 Genentech, Inc. Methods of treating IgE-mediated disorders comprising the administration of high concentration anti-IgE antibody formulations
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
KR20060080535A (ko) 2003-05-14 2006-07-10 이뮤노젠 아이엔씨 약물 콘쥬게이트 조성물
BRPI0414014A (pt) 2003-08-26 2006-10-24 Becton Dickinson Co métodos para distribuição intradérmica de agentes terapêuticos
JP5848861B2 (ja) 2004-04-20 2016-01-27 ジェンマブ エー/エスGenmab A/S Cd20に対するヒトモノクローナル抗体
SG175659A1 (en) 2004-05-05 2011-11-28 Genentech Inc Preventing autoimmune disease by using an anti-cd20 antibody
CA2562800A1 (en) 2004-05-15 2005-12-01 Genentech, Inc. Cross-screening system and methods for detecting a molecule having binding affinity for a target molecule
UA90480C2 (uk) 2004-07-22 2010-05-11 Джэнэнтэк, Инк. Фармацевтична композиція, що має протипухлинну активність, яка включає антитіло до her2
AU2006207901A1 (en) 2005-01-28 2006-08-03 Wyeth Stabilized liquid polypeptide formulations
DOP2006000029A (es) 2005-02-07 2006-08-15 Genentech Inc Antibody variants and uses thereof. (variantes de un anticuerpo y usos de las mismas)
TW200714289A (en) 2005-02-28 2007-04-16 Genentech Inc Treatment of bone disorders
ATE460672T1 (de) 2005-05-20 2010-03-15 Genentech Inc Vorbehandlung einer biologischen probe aus einem individuum mit autoimmunkrankheit
CN103145838A (zh) * 2005-06-30 2013-06-12 Abbvie公司 Il-12/p40结合蛋白
PE20070207A1 (es) 2005-07-22 2007-03-09 Genentech Inc Tratamiento combinado de los tumores que expresan el her
US7700299B2 (en) 2005-08-12 2010-04-20 Hoffmann-La Roche Inc. Method for predicting the response to a treatment
CN101370525B (zh) 2005-08-19 2013-09-18 Abbvie公司 双重可变结构域免疫球蛋白及其用途
TWI478940B (zh) 2005-08-26 2015-04-01 Roche Glycart Ag 具有經改變細胞傳訊活性之改質抗原結合分子
WO2007076062A2 (en) 2005-12-21 2007-07-05 Wyeth Protein formulations with reduced viscosity and uses thereof
WO2007074880A1 (ja) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha 抗体含有安定化製剤
TW200806317A (en) 2006-03-20 2008-02-01 Wyeth Corp Methods for reducing protein aggregation
KR20080104160A (ko) 2006-03-28 2008-12-01 에프. 호프만-라 로슈 아게 항-igf-1r 인간 단일클론 항체 제형
MX2009003774A (es) 2006-10-12 2009-04-22 Genentech Inc Anticuerpos para linfotoxina-alfa.
PE20090329A1 (es) 2007-05-30 2009-03-27 Abbott Lab Anticuerpos humanizados contra el globulomero ab(20-42) y sus usos
CA2702637A1 (en) 2007-10-22 2009-04-30 Schering Corporation Fully human anti-vegf antibodies and methods of using
PE20091174A1 (es) 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd Formulacion liquida con contenido de alta concentracion de anticuerpo
US7914785B2 (en) 2008-01-02 2011-03-29 Bergen Teknologieverforing As B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
ATE548052T1 (de) 2008-01-17 2012-03-15 Philogen Spa Kombination aus einem anti-edb-fibronectin- antikörper-il-2-fusionsprotein und einem b-zellen bindenden molekül, b-zellen-vorläufern und/oder deren krebserregendem gegenspieler
MY163544A (en) 2008-03-25 2017-09-29 Roche Glycart Ag Use of a type ii anti-cd20 antibody with increased antibody dependent cellular cytotoxicity (adcc) in combination with cyclophosphamide, vincristine and doxorubicine for treating non-hodgkin's lymphomas
US20090269339A1 (en) 2008-04-29 2009-10-29 Genentech, Inc. Responses to immunizations in rheumatoid arthritis patients treated with a cd20 antibody
CA2734035C (en) 2008-09-10 2014-05-06 F. Hoffmann-La Roche Ag Delivery device for use with a therapeutic drug
CA2736198A1 (en) 2008-09-19 2010-03-25 F. Hoffmann-La Roche Ag Novel antibody formulation
US9345661B2 (en) 2009-07-31 2016-05-24 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
CN102573459A (zh) 2009-08-04 2012-07-11 弗·哈夫曼-拉罗切有限公司 粘度降低的浓缩多肽制剂
AR078161A1 (es) 2009-09-11 2011-10-19 Hoffmann La Roche Formulaciones farmaceuticas muy concentradas de un anticuerpo anti cd20. uso de la formulacion. metodo de tratamiento.
KR20210021109A (ko) 2010-11-08 2021-02-24 제넨테크, 인크. 피하 투여용 항―il―6 수용체 항체
WO2013031266A1 (ja) 2011-08-26 2013-03-07 テルモ株式会社 シリンジ及びシリンジ収納容器

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6893625B1 (en) * 1986-10-27 2005-05-17 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
US20050163708A1 (en) * 1986-10-27 2005-07-28 Robinson Randy R. Chimeric antibody with specificity to human B cell surface antigen
US6120767A (en) * 1986-10-27 2000-09-19 Pharmaceutical Royalties, L.L.C. Chimeric antibody with specificity to human B cell surface antigen
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5677180A (en) * 1987-01-08 1997-10-14 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5721108A (en) * 1987-01-08 1998-02-24 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US6682734B1 (en) * 1992-11-13 2004-01-27 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US6399061B1 (en) * 1992-11-13 2002-06-04 Idec Pharmaceutical Corporation Chimeric and radiolabelled antibodies specific to human CD20 antigen and use thereof for treatment of B-cell lymphoma
US7422739B2 (en) * 1992-11-13 2008-09-09 Biogen Idec Inc. Anti-CD20 antibodies
US7381560B2 (en) * 1992-11-13 2008-06-03 Biogen Idec Inc. Expression and use of anti-CD20 antibodies
US5776456A (en) * 1992-11-13 1998-07-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US20030180290A1 (en) * 1995-06-07 2003-09-25 Idec Pharmaceuticals Corporation Anti-CD80 antibody having ADCC activity for ADCC mediated killing of B cell lymphoma cells alone or in combination with other therapies
US6797814B2 (en) * 1996-11-27 2004-09-28 Genentech, Inc. Protein purification
US6127526A (en) * 1996-11-27 2000-10-03 Genentech, Inc. Protein purification by Protein A chromatography
US20020032317A1 (en) * 1996-11-27 2002-03-14 Genentech, Inc. Protein purification
US6171586B1 (en) * 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6991790B1 (en) * 1997-06-13 2006-01-31 Genentech, Inc. Antibody formulation
US20050158316A1 (en) * 1997-06-13 2005-07-21 Genentech, Inc. Antibody formulation
US20040191256A1 (en) * 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US7517670B2 (en) * 1998-04-20 2009-04-14 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US20060182739A1 (en) * 1998-05-06 2006-08-17 Genentech, Inc. Protein purification
US6417335B1 (en) * 1998-05-06 2002-07-09 Genentech, Inc. Protein purification
US20080038261A1 (en) * 1998-08-11 2008-02-14 Biogen Idec Inc. Combination therapies for b-cell lymphomas comprising administration of anti-cd20 antibody
US20030026804A1 (en) * 1998-08-11 2003-02-06 Idec Pharmaceuticals Corporation Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibody
US6455043B1 (en) * 1998-08-11 2002-09-24 Idec Pharmaceuticals Corporation Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibody
US20040213784A1 (en) * 1998-11-09 2004-10-28 Idec Pharmaceuticals Corporation Use of chimeric anti-CD20 antibody as in vitro or in vivo purging agent in patients receiving BMT or PBSC transplant
US20020006404A1 (en) * 1999-11-08 2002-01-17 Idec Pharmaceuticals Corporation Treatment of cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
US20010018041A1 (en) * 1999-11-08 2001-08-30 Idec Pharmaceuticals Corporation Treatment of B cell malignancies using anti-CD40L antibodies in combination with anti-CD20 antibodies and/or chemotherapeutics and radiotherapy
US20050123540A1 (en) * 1999-11-08 2005-06-09 Biogen Idec Inc. Treatment of B cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
US6896885B2 (en) * 2000-03-31 2005-05-24 Biogen Idec Inc. Combined use of anti-cytokine antibodies or antagonists and anti-CD20 for treatment of B cell lymphoma
US20020012665A1 (en) * 2000-03-31 2002-01-31 Nabil Hanna Combined use of anti-cytokine antibodies or antagonists and anti-CD20 for treatment of B cell lymphoma
US20050180975A1 (en) * 2000-03-31 2005-08-18 Biogen Idec Inc. Combined use of anti-cytokine antibodies or antagonists and anti-CD20 for treatment of B cell lymphoma
US20020009444A1 (en) * 2000-04-25 2002-01-24 Idec Pharmaceuticals Corporation Intrathecal administration of rituximab for treatment of central nervous system lymphomas
US20050112060A1 (en) * 2000-06-20 2005-05-26 Idec Pharmaceuticals Corporation Treatment of B-cell associated diseases such as malignancies and autoimmune diseases using a cold anti-CD20 antibody/radiolabeled anti-CD22 antibody combination
US20020039557A1 (en) * 2000-06-20 2002-04-04 Christine White Treatment of B-cell associated diseases such as malignances and autoimmune diseases using a cold anti-CD20 antibody/radiolabeled anti-CD22 antibody combination
US6846476B2 (en) * 2000-06-20 2005-01-25 Idec Pharmaceuticals Corporation Treatment of B-cell associated diseases
US20020028178A1 (en) * 2000-07-12 2002-03-07 Nabil Hanna Treatment of B cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
US20020058029A1 (en) * 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
US20070003544A1 (en) * 2000-09-18 2007-01-04 Biogen Idec Inc. Combination therapy for treatment of autoimmune diseases using b- cell depleting/immunoregulatory antibody combination
US20020128448A1 (en) * 2000-10-20 2002-09-12 Idec Pharmaceuticals Corporation Variant IgG3 Rituxan and therapeutic use thereof
US20060121028A1 (en) * 2000-10-20 2006-06-08 Biogen Idec Inc. Variant IgG3 RITUXAN and therapeutic uses thereof
US20070009519A1 (en) * 2001-01-31 2007-01-11 Biogen Idec Inc. Immunoregulatory Antibodies and Uses Thereof
US20020159996A1 (en) * 2001-01-31 2002-10-31 Kandasamy Hariharan Use of CD23 antagonists for the treatment of neoplastic disorders
US20060171950A1 (en) * 2001-01-31 2006-08-03 Biogen Idec Inc. Use of cd23 antagonists for the treatment of neoplastic disorders
US20070020260A1 (en) * 2001-10-25 2007-01-25 Genentech, Inc. Glycoprotein compositions
US20030157108A1 (en) * 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20030103971A1 (en) * 2001-11-09 2003-06-05 Kandasamy Hariharan Immunoregulatory antibodies and uses thereof
US20030180292A1 (en) * 2002-03-14 2003-09-25 Idec Pharmaceuticals Treatment of B cell malignancies using anti-CD40L antibodies in combination with anti-CD20 antibodies and/or chemotherapeutics and radiotherapy
US20060024300A1 (en) * 2002-12-16 2006-02-02 Genentech, Inc. Immunoglobulin variants and uses thereof
US20090155257A1 (en) * 2002-12-16 2009-06-18 Genentech, Inc. Immunoglobulin variants and uses thereof
US20060179501A1 (en) * 2002-12-16 2006-08-10 Chan Andrew C Transgenic mice expressing human cd20
US20060218655A1 (en) * 2002-12-16 2006-09-28 Genentech, Inc. Transgenic mice expressing human cd20 and/or cd16
US20060034835A1 (en) * 2002-12-16 2006-02-16 Genentech, Inc. Immunoglobulin variants and uses thereof
US7767429B2 (en) * 2003-03-05 2010-08-03 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US20040202658A1 (en) * 2003-04-09 2004-10-14 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to TNF-alpha inhibitor
US20080213280A1 (en) * 2003-04-09 2008-09-04 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a tnf-alpha inhibitor
US20060135430A1 (en) * 2003-06-05 2006-06-22 Genentech, Inc. BLy antagonists and uses thereof
US20050095243A1 (en) * 2003-06-05 2005-05-05 Genentech, Inc. Combination therapy for B cell disorders
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
US20050032130A1 (en) * 2003-07-29 2005-02-10 Genentech, Inc. Neutralizing antibody assay and uses therefor
US20050053602A1 (en) * 2003-08-29 2005-03-10 Genentech, Inc. Therapy of ocular disorders
US20050123546A1 (en) * 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US20090010921A1 (en) * 2003-11-05 2009-01-08 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US20050186206A1 (en) * 2003-12-19 2005-08-25 Genentech, Inc. Detection of CD20 in therapy of autoimmune diseases
US20050191297A1 (en) * 2003-12-19 2005-09-01 Genentech, Inc. Detection of CD20 in transplant rejection
US20070031331A1 (en) * 2004-04-16 2007-02-08 Genentech, Inc. Treatment of Disorders
US20080176257A9 (en) * 2004-04-16 2008-07-24 Genentech, Inc. Assay for Antibodies
US20060099662A1 (en) * 2004-04-16 2006-05-11 Genentech, Inc. Assay for antibodies
US20060002930A1 (en) * 2004-04-16 2006-01-05 Genentech, Inc. Treatment of disorders
US20080075719A1 (en) * 2004-04-16 2008-03-27 Genentech, Inc. Method for Augmenting B Cell Depletion
US20060024295A1 (en) * 2004-06-04 2006-02-02 Genentech, Inc. Method for treating lupus
US20060051345A1 (en) * 2004-06-04 2006-03-09 Genentech, Inc. Method for treating multiple sclerosis
US20060062787A1 (en) * 2004-07-22 2006-03-23 Genentech, Inc. Method for treating Sjogren's syndrome
US20060067930A1 (en) * 2004-08-19 2006-03-30 Genentech, Inc. Polypeptide variants with altered effector function
US20090175854A1 (en) * 2004-09-08 2009-07-09 Genentech, Inc. Methods of using death receptor ligands and CD20 antibodies
US20080044421A1 (en) * 2004-09-08 2008-02-21 Genentech, Inc. Methods of using death receptor ligands and CD20 antibodies
US20060110387A1 (en) * 2004-10-05 2006-05-25 Genentech, Inc. Method for treating vasculitis
US20070025987A1 (en) * 2004-10-05 2007-02-01 Genentech, Inc. Method for Treating Vasculitis
US20060088523A1 (en) * 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulations
US20060134111A1 (en) * 2004-12-17 2006-06-22 Genentech, Inc. Antiangiogenesis therapy of autoimmune disease in patients who have failed prior therapy
US20060172385A1 (en) * 2004-12-22 2006-08-03 Genentech, Inc. Methods for producing soluble membrane-spanning proteins
US20060188495A1 (en) * 2005-01-13 2006-08-24 Genentech, Inc. Treatment method
US20080095771A1 (en) * 2005-01-13 2008-04-24 Genentech, Inc. Treatment Method
US20060233797A1 (en) * 2005-04-15 2006-10-19 Genentech, Inc. Treatment of inflammatory bowel disease (IBD)
US20060240007A1 (en) * 2005-04-22 2006-10-26 Genentech, Inc. Method for treating dementia or Alzheimer's disease
US20070009518A1 (en) * 2005-05-18 2007-01-11 Biogen Idec Inc. Methods for treating fibrotic conditions
US20070071675A1 (en) * 2005-08-19 2007-03-29 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20070212733A1 (en) * 2005-11-23 2007-09-13 Genentech, Inc. Methods and compositions related to B cell assays
US20090004189A1 (en) * 2007-06-18 2009-01-01 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
US20090053786A1 (en) * 2007-07-09 2009-02-26 Yung-Hsiang Kao Prevention of disulfide bond reduction during recombinant production of polypeptides
US20090060913A1 (en) * 2007-09-05 2009-03-05 Thomas Friess Combination therapy with type i and type ii anti-cd20 antibodies
US20090214561A1 (en) * 2007-09-24 2009-08-27 David Robert Close Treatment method
US20090098118A1 (en) * 2007-10-15 2009-04-16 Thomas Friess Combination therapy of a type ii anti-cd20 antibody with an anti-bcl-2 active agent
US20090110688A1 (en) * 2007-10-24 2009-04-30 Georg Fertig Combination therapy of type ii anti-cd20 antibody with a proteasome inhibitor
US20090148435A1 (en) * 2007-10-30 2009-06-11 Genentech, Inc. Antibody purification by cation exchange chromatography
US20090162352A1 (en) * 2007-12-21 2009-06-25 Michael Adler Antibody formulation
US20090169550A1 (en) * 2007-12-21 2009-07-02 Genentech, Inc. Therapy of rituximab-refractory rheumatoid arthritis patients
US20100158903A1 (en) * 2008-09-16 2010-06-24 Craig Smith Methods for treating progressive multiple sclerosis

Cited By (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090010921A1 (en) * 2003-11-05 2009-01-08 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US8883980B2 (en) 2003-11-05 2014-11-11 Roche Glycart Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US9296820B2 (en) 2003-11-05 2016-03-29 Roche Glycart Ag Polynucleotides encoding anti-CD20 antigen binding molecules with increased Fc receptor binding affinity and effector function
US10287344B2 (en) 2008-07-03 2019-05-14 Mayo Foundation For Medical Education And Research Methods for reducing global chronic inflammation and the presence of melanoma
US9968676B2 (en) 2009-07-31 2018-05-15 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
US10280227B2 (en) 2009-09-11 2019-05-07 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10752696B2 (en) 2009-09-11 2020-08-25 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10377831B2 (en) 2009-09-11 2019-08-13 Genentech, Inc. Highly concentrated pharmaceutical formulations
US9993548B2 (en) 2011-04-29 2018-06-12 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US11779641B2 (en) 2011-04-29 2023-10-10 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US10420835B2 (en) 2011-04-29 2019-09-24 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US9265815B2 (en) 2011-04-29 2016-02-23 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US9289476B2 (en) 2011-04-29 2016-03-22 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US9289477B2 (en) 2011-04-29 2016-03-22 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce cytotoxic T lymphocyte responses
US9295718B2 (en) 2011-04-29 2016-03-29 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce immune responses to therapeutic proteins
US11717569B2 (en) 2011-04-29 2023-08-08 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10039822B2 (en) 2011-04-29 2018-08-07 Selecta Biosciences, Inc. Method for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10004802B2 (en) 2011-04-29 2018-06-26 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US8652487B2 (en) 2011-04-29 2014-02-18 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10441651B2 (en) 2011-04-29 2019-10-15 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US10765741B2 (en) 2011-05-09 2020-09-08 Mayo Foundation For Medical Education And Research Methods for treating VEGF-expressing cancer using preformed nanoparticle complexes comprising albumin-bound paclitaxel and bevacizumab
WO2013036297A1 (en) * 2011-09-06 2013-03-14 Selecta Biosciences, Inc. Therapeutic protein-specific induced tolerogenic dendritic cells and methods of use
WO2013173687A1 (en) 2012-05-18 2013-11-21 Genentech, Inc. High-concentration monoclonal antibody formulations
EP3427721A1 (en) 2012-05-18 2019-01-16 Genentech, Inc. High-concentration monoclonal antibody formulations
US9777067B2 (en) 2012-09-27 2017-10-03 Massachusetts Institute Of Technology HER2- and VEGF-A-binding proteins with enhanced stability
US20150239977A1 (en) * 2012-09-27 2015-08-27 Massachusetts Institute Of Technology Cd20- and egfr-binding proteins with enhanced stability
US10307482B2 (en) 2012-10-01 2019-06-04 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, cetuximab, and albumin
US11648311B2 (en) 2012-10-01 2023-05-16 Mayo Foundation For Medical Education And Research Cancer treatments
US10279036B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Antibody-albumin nanoparticle complexes comprising albumin, bevacizumab, and paclitaxel, and methods of making and using the same
US10668151B2 (en) 2012-10-01 2020-06-02 Mayo Foundation For Medical Education And Research Nanoparticle complexes of albumin, paclitaxel, and panitumumab for treatment of cancer
US10279035B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10507243B2 (en) 2012-10-01 2019-12-17 Mayo Foundation For Medical Education And Research Nanoparticle complexes of rituximab, albumin and pacltaxel
US10493150B2 (en) 2012-10-01 2019-12-03 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, alemtuzumab, and albumin
US10478495B2 (en) 2012-10-01 2019-11-19 Mayo Foundation For Medical Education And Research Methods for treating cancer using nanoparticle complexes of paclitaxel, cetuximab, and albumin
US10471145B2 (en) 2012-10-01 2019-11-12 Mayo Foundation For Medical Education And Research Methods for treating cancer using nanoparticle complexes of paclitaxel, rituximab, and albumin
US10441656B2 (en) 2012-10-01 2019-10-15 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, cetuximab, and albumin, and methods of making the same
US10420839B2 (en) 2012-10-01 2019-09-24 Mayo Foundation For Medical Education And Research Methods for treating CD52-expressing cancers using compositions comprising nanoparticle complexes of paclitaxel, alemtuzumab, and albumin
US9757453B2 (en) 2012-10-01 2017-09-12 Mayo Foundation For Medical Education And Research Nanoparticle complexes of anti-CD20 antibodies, albumin and paclitaxel
US10376579B2 (en) 2012-10-01 2019-08-13 Mayo Foundation For Medical Education And Research Nanoparticle complexes of albumin, paclitaxel, and anti-VEGF antibody for treatment of cancer
US10376580B2 (en) 2012-10-01 2019-08-13 Mayo Foundation For Medical Education And Research Methods of treating cancer with antibody-albumin nanoparticle complexes comprising albumin, trastuzumab, and paclitaxel
US10413606B2 (en) 2012-10-01 2019-09-17 Mayo Foundation For Medical Education And Research Methods for treating cancer with nanoparticle complexes of albumin-bound paclitaxel and anti-VEGF antibodies
US10406224B2 (en) 2012-10-01 2019-09-10 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
WO2014165280A1 (en) * 2013-03-12 2014-10-09 Teva Pharmaceutical Industries Ltd. Rituximab induction therapy followed by glatiramer acetate therapy
CN105188751A (zh) * 2013-03-12 2015-12-23 泰华制药工业有限公司 利妥昔单抗诱导疗法继而醋酸格拉替雷疗法
CN105189559A (zh) * 2013-03-15 2015-12-23 塔科达有限责任公司 抗体制剂及其用途
US10668053B2 (en) 2013-05-03 2020-06-02 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US10357483B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods comprising dosing combinations for reducing undesired humoral immune responses
US10434088B2 (en) 2013-05-03 2019-10-08 Selecta Biosciences, Inc. Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US10357482B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US11298342B2 (en) 2013-05-03 2022-04-12 Selecta Biosciences, Inc. Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
CN110559435A (zh) * 2013-09-11 2019-12-13 伊戈尔生物药品股份有限公司 包含离子性液体的液体蛋白质制剂
US11986526B2 (en) 2013-09-11 2024-05-21 Eagle Biologics, Inc. Liquid protein formulations containing 4-ethyl-4-methylmorpholinium methylcarbonate (EMMC)
AU2014337263B2 (en) * 2013-10-16 2019-12-12 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US10376582B2 (en) 2013-10-16 2019-08-13 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
WO2015057910A1 (en) * 2013-10-16 2015-04-23 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US10568951B2 (en) 2013-11-18 2020-02-25 Formycon Ag Pharmaceutical composition of an anti-VEGF antibody
US10800851B2 (en) 2014-02-28 2020-10-13 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
US11713355B2 (en) 2014-02-28 2023-08-01 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
RU2708332C2 (ru) * 2014-06-13 2019-12-06 Мэйо Фаундейшн Фор Медикал Эдьюкейшн Энд Рисерч Лечение лимфом
WO2015191969A1 (en) * 2014-06-13 2015-12-17 Mayo Foundation For Medical Education And Research Treating lymphomas
CN113318239A (zh) * 2014-06-13 2021-08-31 梅约医药教育及研究基金会 治疗淋巴瘤
US10213513B2 (en) 2014-06-16 2019-02-26 Mayo Foundation For Medical Education And Research Treating myelomas
US11285221B2 (en) 2014-06-16 2022-03-29 Mayo Foundation For Medical Education And Research Treating myelomas
US11654185B2 (en) * 2014-07-16 2023-05-23 New York University Use of hyaluronidase for treatment of muscle stiffness
US20190282674A1 (en) * 2014-07-16 2019-09-19 New York University Use of hyaluronidase for treatment of muscle stiffness
US10071114B2 (en) 2014-09-07 2018-09-11 Selecta Biosciences, Inc. Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US11633422B2 (en) 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
US10596112B2 (en) 2014-10-06 2020-03-24 Mayo Foundation For Medical Education And Research Methods of using albumin-antibody nanoparticle complex compositions for treating cancer
US10966923B2 (en) 2014-10-06 2021-04-06 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10772833B2 (en) 2014-10-06 2020-09-15 Mayo Foundation For Medical Education And Research Albumin-CTLA-4 paclitaxel nanopartilce complex compositions and methods of making and using the same
US10780050B2 (en) 2014-10-06 2020-09-22 Mayo Foundation For Medical Education And Research Lyophilized compositions comprosing albumin-EGFR paclitaxel nanoparticle complexes
US10596111B2 (en) 2014-10-06 2020-03-24 Mayo Foundation For Medical Education And Research Methods of making lyophilized compositions comprising albumin-VEGF paclitaxel nanoparticle complexes
US10780049B2 (en) 2014-10-06 2020-09-22 Mayo Foundation For Medical Education And Research Lyophilized compositions comprising albumin-rankl or albumin-GD2 paclitaxel nanoparticle complexes
US10610484B2 (en) 2014-10-06 2020-04-07 Mayo Foundation For Medical Education And Research Methods of using albumin-CD20 paclitaxel nanoparticle complex compositions for treating cancer
US10300016B2 (en) 2014-10-06 2019-05-28 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10391055B2 (en) 2014-10-06 2019-08-27 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US11433023B2 (en) 2014-10-06 2022-09-06 Mayo Foundation For Medical Education And Research Albumin-PD-1 paclitaxel nanoparticle complex compositions and methods of making and using the same
US10624846B2 (en) 2014-10-06 2020-04-21 Mayo Foundation For Medical Education And Research Lyophilized compositions comprising albumin-antibody paclitaxel nanoparticle complexes
US10993911B2 (en) 2014-10-06 2021-05-04 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10993912B2 (en) 2014-10-06 2021-05-04 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10322084B2 (en) 2014-10-06 2019-06-18 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10793630B2 (en) 2014-12-04 2020-10-06 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute myeloid leukemia
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
US9981021B1 (en) 2015-04-09 2018-05-29 Kinetiq, Inc. Subcutaneous therapeutic enzyme formulations, uses, and methods for generating thereof
US11065309B2 (en) 2015-04-09 2021-07-20 Kinetiq Therapeutics, Llc Subcutaneous therapeutic enzyme formulations, uses, and methods for generating thereof
US10286045B2 (en) 2015-04-09 2019-05-14 Kinetiq, Inc. Subcutaneous therapeutic enzyme formulations, uses, and methods for generating thereof
US11021543B2 (en) 2015-06-24 2021-06-01 Janssen Biotech, Inc. Immune modulation and treatment of solid tumors with antibodies that specifically bind CD38
US11241387B2 (en) 2015-08-18 2022-02-08 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11660339B2 (en) 2015-10-06 2023-05-30 Mayo Foundation For Medical Education And Research Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US10561726B2 (en) 2015-10-06 2020-02-18 Vavotar Life Sciences LLC Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
EP3827845B1 (en) 2015-11-03 2022-03-30 Janssen Biotech, Inc. Subcutaneous formulations of anti-cd38 antibodies and their uses
US11708419B2 (en) 2015-11-03 2023-07-25 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11708420B2 (en) 2015-11-03 2023-07-25 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11732051B2 (en) 2015-11-03 2023-08-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11566079B2 (en) 2015-11-03 2023-01-31 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
EP3370770B1 (en) 2015-11-03 2021-01-20 Janssen Biotech, Inc. Subcutaneous formulations of anti-cd38 antibodies and their uses
US10781261B2 (en) 2015-11-03 2020-09-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US10702606B2 (en) 2015-12-10 2020-07-07 Menicon Co., Ltd. Peptide composition
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US11351254B2 (en) 2016-02-12 2022-06-07 Mayo Foundation For Medical Education And Research Hematologic cancer treatments
US11305020B2 (en) 2016-03-21 2022-04-19 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US11878061B2 (en) 2016-03-21 2024-01-23 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug
US11655301B2 (en) 2016-04-06 2023-05-23 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US10618969B2 (en) 2016-04-06 2020-04-14 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11548946B2 (en) 2016-09-01 2023-01-10 Mayo Foundation For Medical Education And Research Carrier-PD-L1 binding agent compositions for treating cancers
US11160876B2 (en) 2016-09-01 2021-11-02 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
US11311631B2 (en) 2016-09-06 2022-04-26 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
US11872205B2 (en) 2016-09-06 2024-01-16 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11590098B2 (en) 2016-09-06 2023-02-28 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11427637B2 (en) 2016-09-06 2022-08-30 Mayo Foundation For Medical Education And Research Methods of treating PD-L1 expressing cancer
US11400109B2 (en) * 2016-11-10 2022-08-02 Translate Bio, Inc. Subcutaneous delivery of messenger RNA
US11426451B2 (en) 2017-03-11 2022-08-30 Selecta Biosciences, Inc. Methods and compositions related to combined treatment with antiinflammatories and synthetic nanocarriers comprising an immunosuppressant
US11738082B2 (en) 2017-05-16 2023-08-29 Bhami's Research Laboratory, Pvt. Ltd. High concentration protein formulations with reduced viscosity
US10646569B2 (en) 2017-05-16 2020-05-12 Bhami's Research Laboratory, Pvt. Ltd. High concentration protein formulations with reduced viscosity
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
CN112533947A (zh) * 2018-07-03 2021-03-19 加拉帕戈斯股份有限公司 高浓度液体抗体制剂
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation

Also Published As

Publication number Publication date
CA2771571A1 (en) 2011-03-17
CR20120090A (es) 2012-07-12
KR101782195B1 (ko) 2017-09-26
WO2011029892A3 (en) 2011-09-15
IL249066B (en) 2018-02-28
JP2020011962A (ja) 2020-01-23
JP2013504540A (ja) 2013-02-07
HUE062860T2 (hu) 2023-12-28
US10752696B2 (en) 2020-08-25
IL218165A (en) 2016-12-29
MY174009A (en) 2020-03-03
EP4218715A1 (en) 2023-08-02
DOP2012000064A (es) 2012-05-31
JP2015061839A (ja) 2015-04-02
DK2475353T3 (en) 2016-05-30
IL249066A0 (en) 2017-01-31
AU2010294186B2 (en) 2013-09-19
FI3061445T3 (fi) 2023-08-30
CN102686216A (zh) 2012-09-19
DK3061445T3 (da) 2023-09-11
RS54769B1 (sr) 2016-10-31
KR20170034441A (ko) 2017-03-28
JP6364310B2 (ja) 2018-07-25
CA2771571C (en) 2013-11-12
US10377831B2 (en) 2019-08-13
PE20121178A1 (es) 2012-09-06
UA104909C2 (uk) 2014-03-25
ES2953478T3 (es) 2023-11-13
TN2012000101A1 (en) 2013-09-19
NZ621448A (en) 2015-07-31
JP2017200924A (ja) 2017-11-09
MA33592B1 (fr) 2012-09-01
MX360435B (es) 2018-10-31
PL3061445T3 (pl) 2023-11-06
SMT201600176B (it) 2016-08-31
ES2574052T3 (es) 2016-06-14
EP2475353A2 (en) 2012-07-18
KR20120051094A (ko) 2012-05-21
IL218165A0 (en) 2012-07-31
HRP20160807T1 (hr) 2016-08-26
HRP20231019T1 (hr) 2023-12-08
HUE029078T2 (en) 2017-02-28
US10280227B2 (en) 2019-05-07
ZA201201605B (en) 2019-09-25
SG179030A1 (en) 2012-04-27
BR112012005017B1 (pt) 2021-09-08
TW201429506A (zh) 2014-08-01
HK1174553A1 (zh) 2013-06-14
TWI515020B (zh) 2016-01-01
JP5674788B2 (ja) 2015-02-25
MX360432B (es) 2018-10-31
US20190367629A1 (en) 2019-12-05
ECSP12011722A (es) 2012-04-30
EP3061445B1 (en) 2023-07-05
MY197773A (en) 2023-07-13
EP2475353B1 (en) 2016-04-13
HK1208176A1 (zh) 2016-02-26
US20150086537A1 (en) 2015-03-26
PL2475353T3 (pl) 2016-10-31
MX2012002861A (es) 2012-04-20
AU2010294186A1 (en) 2012-03-08
EP4218716A1 (en) 2023-08-02
BR112012005017B8 (pt) 2021-09-21
EA201792552A1 (ru) 2018-09-28
PH12016501002B1 (en) 2018-01-29
AR078161A1 (es) 2011-10-19
BR112012005017A2 (pt) 2020-10-27
CO6440527A2 (es) 2012-05-15
TW201114449A (en) 2011-05-01
LT3061445T (lt) 2023-09-25
EP3064196A1 (en) 2016-09-07
CN104399075B (zh) 2018-11-06
EP3061445A1 (en) 2016-08-31
KR20140006112A (ko) 2014-01-15
JP6934919B2 (ja) 2021-09-15
PH12016501002A1 (en) 2018-01-29
US20160137742A1 (en) 2016-05-19
CN104399075A (zh) 2015-03-11
PE20151425A1 (es) 2015-09-18
CY1117941T1 (el) 2017-05-17
CN102686216B (zh) 2014-11-05
GT201200069A (es) 2014-05-26
SI3061445T1 (sl) 2023-10-30
SI2475353T1 (sl) 2016-08-31
EA031972B1 (ru) 2019-03-29
WO2011029892A2 (en) 2011-03-17
PT3061445T (pt) 2023-08-28
CL2012000620A1 (es) 2013-09-06
US20210054092A1 (en) 2021-02-25
DOP2016000110A (es) 2016-08-15
NZ598653A (en) 2014-03-28
KR101782301B1 (ko) 2017-09-26
EA201200475A1 (ru) 2012-10-30
TWI515021B (zh) 2016-01-01

Similar Documents

Publication Publication Date Title
US10752696B2 (en) Highly concentrated pharmaceutical formulations
AU2013266987B2 (en) Highly concentrated pharmaceutical formulations comprising anti - CD20 antibody
CN111202845A (zh) 包含抗cd20抗体的药物配制剂及其制备方法和应用
EA041673B1 (ru) Высококонцентрированные фармацевтические композиции, содержащие антитело к cd20, их применение и способ лечения с помощью таких композиций

Legal Events

Date Code Title Description
AS Assignment

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ADLER, MICHAEL;MAHLER, HANNS-CHRISTIAN;STAUCH, OLIVER BORIS;REEL/FRAME:024970/0156

Effective date: 20100908

Owner name: GENENTECH, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:024970/0264

Effective date: 20100909

STCB Information on status: application discontinuation

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