WO2003039485A2 - Stable liquid pharmaceutical formulation of igg antibodies - Google Patents

Stable liquid pharmaceutical formulation of igg antibodies Download PDF

Info

Publication number
WO2003039485A2
WO2003039485A2 PCT/US2002/036093 US0236093W WO03039485A2 WO 2003039485 A2 WO2003039485 A2 WO 2003039485A2 US 0236093 W US0236093 W US 0236093W WO 03039485 A2 WO03039485 A2 WO 03039485A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
formulation
buffer
pharmaceutical formulation
liquid pharmaceutical
Prior art date
Application number
PCT/US2002/036093
Other languages
French (fr)
Other versions
WO2003039485A3 (en
Inventor
Elizabet A. Kaisheva
Supriya Gupta
Shanti G. Duvur
Malathy Subramanian
Original Assignee
Protein Design Labs
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to NZ532896A priority Critical patent/NZ532896A/en
Priority to JP2003541777A priority patent/JP5290489B2/en
Priority to IL16167702A priority patent/IL161677A0/en
Priority to ES02802895T priority patent/ES2392073T3/en
Application filed by Protein Design Labs filed Critical Protein Design Labs
Priority to EP02802895A priority patent/EP1441589B1/en
Priority to AT02802895T priority patent/ATE556591T1/en
Priority to DK02802895.9T priority patent/DK1441589T3/en
Priority to CA2466034A priority patent/CA2466034C/en
Priority to AU2002363339A priority patent/AU2002363339B2/en
Publication of WO2003039485A2 publication Critical patent/WO2003039485A2/en
Publication of WO2003039485A3 publication Critical patent/WO2003039485A3/en
Priority to IL161677A priority patent/IL161677A/en
Priority to HK05107139A priority patent/HK1074750A1/en
Priority to LU93314C priority patent/LU93314I2/fr
Priority to CY2016044C priority patent/CY2016044I2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/249Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • 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
    • A61K47/02Inorganic compounds
    • 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
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/246IL-2
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • C07K16/2854Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72 against selectins, e.g. CD62
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • the present invention relates generally to the field of pharmaceutical formulation of antibodies. Specifically, the present invention relates to a stable, liquid, high concentration antibody formulation.
  • This invention is exemplified by a stabilized liquid formulation of Daclizumab, an anti-IL2 receptor antibody; HAIL-12, a humanized anti-JX- 12 monoclonal antibody; and HuEP5C7, a humanized anti-L selectin monoclonal antibody.
  • Immunoglobulins are recognized as possessing characteristics that tend to form aggregates and particulates in solution, and as such, may require filtration before use for intravenous or subcutaneous injection.
  • the formation of protein aggregates and particulates has long been a problem in the development of parenteral immunoglobulin products, especially when the immunoglobulins are formulated at high concentrations.
  • SynagisTM (Medrmmiine) is a humanized monoclonal IgGl antibody produced by recombinant DNA technology, directed to an epitope in the A antigenic site of the T protein of respiratory syncytial virus (RSN).
  • SynagisTM is a composite of human (90%) and murine ( 10%) antibody sequences.
  • SynagisTM is supplied as a sterile lyophilized product for reconstruction with sterile water for injection. Reconstituted SynagisTM is to be administered by intramuscular injection only. Upon reconstitution, SynagisTM contains the following excipients: 47mM histidine, 3.0 mM glycine, 5.6% mannitol, and the active ingredient, IgGl antibody, at a concentration of 100 milligrams per vial. The reconstituted SynagisTM is to be administered within 6 hours of reconstitution.
  • WO 89/11297 discloses a lyophilized monoclonal antibody formulation comprising a lyophilized formulation of 1-25 mg/ml IgG monoclonal antibody, 2-10% maltose, and sodium acetate, phosphate, or citrate buffer having a pH between 3.0 to 6.0.
  • WO 97/45140 discloses an aqueous preparation of anti-CD4 antibody concentrated to approximately 100 mg/ml in 100 mM sodium citrate, 0.05 mM EDTA, pH 6.0.
  • the application discloses a slight rise in turbidity after concentration of the antibody, which likely reflects protein aggregation. Removing this aggregation requires addition of Polysorbate 80 and sterile filtration.
  • WO 90/11091 discloses injectable aqueous compositions comprising about 5 mg/ml of IgM, 2.5-5 % (w/v) human serum albumin, in 8-20 mM phosphate buffer, 270 mM sodium chloride, pH 6.8-7.4.
  • U.S. Patent No. 6,171,586 discloses a stable aqueous pharmaceutical formulation comprising a therapeutically effective amount of an antibody not subjected to prior lyophilization, an acetate buffer from about pH 4.8 to about 5.5, a surfactant, and a polyol, wherein the formulation lacks a tomcifying amount of sodium chloride.
  • U.S. Patent Application Publication No. US 2001/0014326A1 discloses a pre- lyophilized antibody formulation containing 25 mg/ml anti-IgE antibody, 5 mM histidine, pH 6.0, 85 mM sucrose, and 0.01% polysorbate 20.
  • U.S. Patent No. 5,744,132 discloses a composition comprising 1-1000 ⁇ g/ml IL-12 antibody, 2% sucrose, 4.15% mannitol, lOmM sodium succinate, and about 0.02% Tween ® 20, having a pH of about 5.6.
  • U.S. Patent No. 6,267,958 discloses a reconstituted formulation of 100 mg/ml rhuMab E25, in 20 mM histidine, pH 6.0, 340 mM sucrose, 0.04 % polysorbate 20, and 0.9 % benzyl alcohol.
  • U.S. Patent No. 6,267,958 discloses a reconstituted formulation of 100 mg/ml rhuMab E25, in 20 mM histidine, pH 6.0, 340 mM sucrose, 0.04 % polysorbate 20, and 0.9 % benzyl alcohol.
  • 6,165,467 discloses a process for stabilizing a human monoclonal antibody composition produced by hybridoma cell line having accession number HB8307, which comprises dialyzing the human monoclonal antibody in a phosphate salt stabilized buffer solution having a pH from 7.2 to 7.4, said solution comprising 1-20 mg of D- mannitol per mg of said monoclonal antibody, 0.005-0.2 millimole of glycine per mg of said monoclonal antibody, and an amount of pH stabilizing phosphate salt to stabilize the pH of said solution.
  • a stable liquid antibody preparation wherein the antibody concentration is 50 mg/ml or greater; such preparation is suitable for parenteral administration, including intravenous, intramuscular, intraperitoneal, or subcutaneous injection to a human.
  • This invention is directed to a stable liquid pharmaceutical formulation comprising a high concentration, e.g., greater than 50 mg/ml, of an antibody in 20-60 mM succinate buffer or 30-70 mM histidine buffer (pH from about pH 5.5 to about pH 6.5), a tonicity modifier, and about 0.01- 0.1 % polysorbate.
  • This formulation retains the physical, chemical, and biological stability of antibody and prevents the immunoglobulins intended for administration to human subjects from forming aggregates and particulates in the final product.
  • Preferred antibodies of this invention include Daclizumab, a humanized anti-IL-2 receptor monoclonal antibody; HATL-12, a humanized anti-IL-12 monoclonal antibody; and HuEP5C7, a humanized anti-L selectin monoclonal antibody; and Flintozumab, a humanized anti-gamma interferon monoclonal antibody.
  • the liquid antibody formulation is stable at refrigerated temperature (2-8°C) for at least 1 year and preferably 2 years. This liquid formulation is also stable at room temperature (23-27°C) for at least six months. This liquid formulation is suitable for subcutaneous injection.
  • Figure 1 A shows the percent clips formation
  • Figure IB shows the percent aggregates, at various pH levels following a four-week incubation of the sample at 45°C, as assessed by SEC-HPLC.
  • Figure 2 shows the percent of degradation obtained at various pH levels as assessed by cIEF following a four- week incubation of the sample at 45°C.
  • Figure 3 shows the percent of iso-aspartic acid formed at various pH levels as assessed by the Promega IsoQuant kit following a four- week incubation of the sample at 45°C.
  • Figure 4 shows the effect of different buffers over time on potency following incubation at 37°C. DETAILED DESCRIPTION OF THE INVENTION I. Definition
  • buffer encompasses those agents which maintain the solution pH in an acceptable range and may include succinate (sodium), histidine, phosphate' (sodium or potassium), Tris (tris (hydroxymethyi) aminomethane), diethanolamine, and the like.
  • the buffer of this invention has a pH in the range from about 5.5 to about 6.5; and preferably has a pH of about 6.0.
  • Examples of buffers that will control the pH in this range include succinate (such as sodium succinate), gluconate, histidine, citrate phospate and other organic acid buffers.
  • “Pharmaceutically acceptable excipients” are those inert substances that can reasonably be administered to a subject mammal and provide an effective dose of the active ingredient employed. These substances are added to a formulation to stabilize the physical, chemical and biological structure of the antibody. The term also refers to additives that may be needed to attain an isotonic formulation, suitable for the intended mode of administration.
  • pharmaceutical formulation refers to preparations which are in such form as to permit the biological activity of the active ingredients to be unequivocally effective, and which contain no additional components which are toxic to the subjects to which the formulation would be administered.
  • a “stable” formulation is one in which the protein therein essentially retains its physical stability, chemical stability, and biological activity upon storage.
  • Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10:29-90 (1993). Stability can be measured at a selected temperature for a selected time period.
  • a “stable" liquid antibody formulation is a liquid antibody formulation with no significant changes observed at a refrigerated temperature (2-8 °C) for at least 12 months, preferably 2 years, and more preferably 3 years; or at room temperature (23 -27 °C) for at least 3 months, preferably 6 months, and more preferably 1 year.
  • the criteria for stability are as follows. No more than 10%, preferably 5%, of antibody monomer is degraded as measured by SEC-HPLC. The solution is colorless, or clear to slightly opalescent by visual analysis. The concentration, pH and osmolality of the formulation have no more than +/- 10% change. Potency is within 70-130%, preferably 80-120% of the control. No more than 10%, preferably 5% of clipping (hydrolysis) is observed. No more than 10%, preferably 5% of aggregation is formed.
  • An antibody “retains its physical stability” in a pharmaceutical formulation if it shows no significant increase of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering, size exclusion chromatography (SEC-HPLC) and dynamic light scattering.
  • the protein conformation is not altered.
  • the changes of protein conformation can be evaluated by fluorescence spectroscopy, which determines the protein tertiary structure, and by FTIR spectroscopy, which determines the protein secondary structure.
  • An antibody "retains its chemical stability” in a pharmaceutical formulation, if it shows no significant chemical alteration. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein.
  • Degradation processes that often alter the protein chemical structure include hydrolysis or clipping (evaluated by methods such as size exclusion chromatography and SDS-PAGE), oxidation (evaluated by methods such as by peptide mapping in conjunction with mass spectroscopy or MALDI/TOF/MS), deamidation (evaluated by methods such as ion-exchange chromatography, capillary isoelectric focusing, peptide mapping, isoaspartic acid measurement), and isomerization (evaluated by measuring the isoaspartic acid content, peptide mapping, etc.).
  • An antibody "retains its biological activity" in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within a predetermined range of the biological activity exhibited at the time the pharmaceutical formulation was prepared.
  • the biological activity of an antibody can be determined, for example, by an antigen binding ELISA assay.
  • isotonic means that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 270-328 mOsm. Slightly hypotonic osmotic pressure is 250-269 and slightly hypertonic osmotic pressure is 328-350 mOsm. Osmotic pressure can be measured, for example, using a vapor pressure or ice-freezing type osmometer.
  • Tonicity modifiers are those pharmaceutically acceptable inert substances that can be added to the formulation to provide an isotonity of the formulation.
  • Tonicity modifiers suitable for this invention include salts and amino acids.
  • the antibody formulation contains pharmaceutically acceptable excipients.
  • the antibody formulation is formulated such that the antibody retains its physical, chemical and biological activity.
  • the formulation is preferably stable for at least 1 year at refrigerated temperature (2-8°C) and 6 months at room temperature (23-27°C).
  • the analytical methods for evaluating the product stability include size exclusion chromatography (SEC-HPLC), dynamic light scattering test (DLS), differential scanning calorimetery (DSC), iso-asp quantification, potency, UV at 340nm, and UV spectroscopy.
  • SEC size exclusion chromatography
  • DSC differential scanning calorimetery
  • iso-asp quantification potency, UV at 340nm, and UV spectroscopy.
  • SEC J. Pharm. Scien., 83:1645-1650, (1994); Pharm. Res., 11:485 (1994); J. Pharm. Bio. Anal., 15:1928 (1997); J. Pharm. Bio. Anal, 14:1133-1140 (1986)
  • DSC Pharm. Res., 15:200 (1998); Pharm.
  • the iso-Asp content in the samples is measured using the Isoquant Isoaspartate Detection kit (Promega).
  • the kit uses the enzyme Protein Isoaspartyl Methyltransferase (PLMT) to specifically detect the presence of isoaspartic acid residues in a target protein.
  • PLMT Protein Isoaspartyl Methyltransferase
  • PIMT catalyzes the transfer of a methyl group from S-adenosyl-L-methionine to isoaspartic acid at the ce-carboxyl position, generating S-adenosyl-L-homocysteine (SAH) in the process.
  • SAH S-adenosyl-L-homocysteine
  • the potency or bioactivity of an antibody can be measured by its ability to bind to its antigen.
  • the specific binding of an antibody to its antigen can be quantitated by any method known to those skilled in the art, for example, an immunoassay, such as ELISA (enzyme-linked immunosorbant assay).
  • the invention herein relates to a stable aqueous formulation comprising an antibody.
  • the antibody in the formulation is prepared using techniques available in the art for generating antibodies, exemplary methods of which are described in more detail in the following sections.
  • the antibody is directed against an antigen of interest.
  • the antigen is a biologically important polypeptide and administration of the antibody to a mammal may prevent or treat a disorder.
  • antibodies directed against nonpolypeptide antigens are also contemplated.
  • the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor.
  • exemplary antigens include molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha- 1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VDTC, factor LX, tissue factor, and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -
  • the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed cells, is removed, for example, by centrifugation or ultrafiltration. Where the antibody is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography is the preferred purification technique.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody. Protein A can be used to purify antibodies that are based on human Y 1; T 2 , or Y heavy chains (Lindmark et al, J.
  • Protein G is recommended for all mouse isotypes and for human Y 3 (Guss et al, EMBOJ. 5:1567-1575 (1986)).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the antibody comprises a C H3 domain
  • the Bakerbond ABXTM resin J. T. Baker, Phillipsburg, ⁇ . J.
  • Daclizumab (USAN, United States Adopted Names), a humanized anti-IL-2 receptor antibody.
  • Daclizumab is currently being marketed as Zenapax ® for the prevention of organ rejection after renal transplantation and is administered through the intravenous route.
  • Daclizumab is also useful for treating psoriasis, for which, the subcutaneous delivery is the preferred route of administration. For a subcutaneous delivery of antibody, high concentration of antibody is preferred.
  • Daclizumab is a recombinant humanized monoclonal antibody, subclass IgGl. The molecule is composed of two identical heavy chain and two identical light chain subunits. Disulfide bridges link the four chains.
  • Daclizumab monomer is approximately 150,000 daltons in molecular weight. Daclizumab binds to the p55 subunit of the IL-2 receptor expressed on activated T cells. The antigen target is designated CD25. Daclizumab is produced from a GS-NS0 cell line containing the heavy and light chain genes by fed-batch fermentation culture. Bioreactor harvests are processed to remove cells and debris and purified using a combination of ion-exchange and gel filtration chromatography and a series of ultrafiltration and filtration techniques to produce drug substance containing greater than 95% monomeric species.
  • IL-12 anti-interleukin 12
  • IL-12 is a cytokine synthesized by antigen presenting cells. It is composed of two subunits (p35 and p40), both must be present for functional activity.
  • Functional EL- 12 is also called EL- 12p70.
  • This cytokine preferentially acts on T helper cell type 1 (Thl) lymphocytes and natural killer cells by increasing their proliferative rate.
  • Thl T helper cell type 1
  • IFNg interferon gamma
  • 16G2 (Hoffman La Roche) is a murine antibody raised against LL-12p70. 16G2 has been shown to act in near stoichiometric amounts to EL- 12 in a functional assay-the inhibition of proliferation of activated T cells from human peripheral blood (PBMC). This is an important characteristic because p40 dimers of EL- 12 exist in serum and antibodies raised to the p40 subunit need to be used in excess amounts to neutralize the proliferative capacity of a given amount of EL- 12. 16G2 was humanized at Protein Design Labs. (Fremont, CA) to give rise to HAEL-12 (humanized anti-EL-12, an EgGl antibody).
  • Another preferred antibody is anti-L selectin antibody.
  • Selectins such as L, E, and P-selectin have been found to be associated with tissue damage during the course of ischemia and reperfusion. Neutrophils play an important role in this connection. It is assumed that selectin is required for the recruitment of neutrophils. L-selectin is important for the complete development of damage in skeletal muscle as well as in the lung (Seekamp, et al, Am. J. Pathol 11:592-598 (1994). Mulligan, et al, J. Immunol. 151 :832-840 (1994).
  • HuEP5C7 (SMART Anti-L Selectin) is a humanized anti-L selectin monoclonal antibody, that contains mutant IgG2 Fc, cross reacts with both human E and P selectin antigens. It is currently being developed by Protein Design Labs, Inc. for various indications such as asthma, stroke, trauma, and certain autoimmune diseases.
  • Flintozumab an anti-gamma interferon antibody.
  • Flintozumab is an IgGl humanized monoclonal antibody developed by Protein Design Labs, Inc. for the treatment of immune disorders mediated by interferon-gamma (EFN-g), a proinflammatory cytokine.
  • EFN-g induces the expression of major histocompatibility complex (MHC) class I and/or class U (HLA-DR) antigens, enhances the cytolytic activity of natural killer cells, activates macrophages, and modulates the immunoglobulin isotype profile of the humoral response.
  • MHC major histocompatibility complex
  • HLA-DR class U
  • EFN-g As a lymphokine, EFN-g also enhances the development of T helper cell type 1 (Thl), while suppressing the development of T helper cell type 2 (Th2) cells. Aberrations in the Thl/Th2 ratio have been implicated in a variety of autoimmune conditions.
  • a pharmaceutical formulation comprising the antibody is prepared.
  • the formulation development approach is as follows: selecting the optimum solution pH, selecting buffer type and concentration, evaluating the effect of various excipients of the liquid stability, and optimizing the concentration of the screened excipients using an I-optimal experimental design (Statistics for Experimenters: An Introduction to Design, Data Analysis, and Model Building, Box,
  • compositions of this invention minimize the formation of antibody aggregates and particulates and insure that the antibody maintains its bioactivity over time.
  • the composition is a pharmaceutically acceptable liquid formulation containing a high concentration of an antibody in a buffer having a neutral or slightly acidic pH (pH 5.5-6.5), a surfactant, and a tonicity modifier.
  • the antibody in the composition is a high concentration of 50 mg/ml or greater, preferably 100 mg/ml or greater.
  • a preferred composition of this invention contains Daclizumab, a humanized anti-EL2 receptor antibody; HAEL12, a humanized anti-EL-12 antibody; HaEP5C7, a humanized anti-L selectin antibody; and Flintozumab, a humanized anti-gamma interferon antibody.
  • a buffer of pH 5.5-6.5 is used in the composition.
  • a buffer of pH 6.0-6.5 is preferred.
  • buffers that control the pH in this range include succinate (such as sodium succinate), gluconate, histidine, citrate, phosphate, and other organic acid buffers.
  • Succinate (pKa 5.63) is a preferred buffer for subcutaneous injection. Histidine (pK 5.97) is less preferred because of its susceptibility to oxidization, although such oxidation can be retarded by replacing the vial headspace with N 2 or adding an antioxidant. Citrate and phosphate buffers are much less preferred because it causes a painful reaction when injected subcutaneously. A preferred buffer contains about 20-60 mM sodium succinate.
  • Another preferred buffer is 30-70 mM histidine buffer overlaid with N 2 .
  • a surfactant is also added to the antibody formulation.
  • exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbates 20, 80, such as
  • the amount of surfactant added is such that it reduces aggregation of the formulated antibody and/or minimizes the formation of particulates in the formulation and/or reduces adsorption.
  • the surfactant may be present in the formulation in an amount from about 0.005% to about 0.5%, preferably from about 0.01% to about 0.1%, more preferably from about 0.01% to about 0.05%, and most preferably from about 0.02% to about 0.04%.
  • a tonicity modifier which contributes to the isotonicity of the formulations, is added to the present composition.
  • the tonicity modifier useful for the present invention includes salts and amino acids.
  • Salts that are pharmaceutically acceptable and suitable for this invention include sodium chloride, sodium succinate, sodium sulfate, potassium chloride, magnesium chloride, magnesium sulfate, and calcium chloride.
  • Preferred salts for this invention are NaCl and MgCl 2 .
  • MgCl 2 may also improve the antibody stability by protecting the protein from deamidation.
  • a preferred concentration of NaCl is about 75- 150 mM.
  • a preferred concentration of MgCl 2 is about 1-100 mM.
  • Amino acids that are pharmaceutically acceptable and suitable for this invention include proline, alanine, L- arginine, asparagine, L-aspartic acid, glycine, serine, lysine, and histidine.
  • a preferred amino acid for this invention is proline.
  • a preferred concentration of proline is than 200 mM.
  • EDTA which is commonly used to stabilize a protein formulation, may also be included in the formulation.
  • EDTA as a chelating agent, may inhibit the metal-catalyzed oxidation of the sulfhydryl groups, thus reducing the formation of disulfide-linked aggregates.
  • a preferred concentration of EDTA is 0.01-0.2%.
  • Exemplary liquid compositions are formulations comprising antibody at about 100 mg/ml or greater, about 20-60 mM sodium succinate (pH 6), about 0.01-0.1% polysorbate 20 or 80, and about 75-150 mM NaCl. This formulation retains the stability of biological activity of the monoclonal antibody, and prevents the immunoglobulins intended for administration to human subjects from physical, chemical and biological degradation in the final product.
  • the liquid antibody formulation of this invention is suitable for parenteral administration such as intravenous, intramuscular, intraperitoneal, or subcutaneous injection; particularly suitable for subcutaneous injection.
  • Sample formulations contained 5.0 mg/ml anti-EL2 receptor antibody (Daclizumab) in one of three buffers: 50 mM sodium acetate buffer at pH 4.0 or 5.0, 50 mM histidine at pH 5.5, 6.0, or 6.5, or 50 mM sodium phosphate buffer at pH 7.0 or 8. 5. Independent formulations were incubated at either 5°C or 45° C with 100 RPM shaking for 4 weeks.
  • Daclizumab anti-EL2 receptor antibody
  • the physical and chemical stability of each sample was assessed at 0 and 4 weeks by analytical methods including: pH and visual analysis, UV spectroscopy at 340 nm, size exclusion chromatography (SEC- HPLC), fluorescence spectroscopy, dynamic light scattering (DLS), differential scanning calorimetry (DSC), Promega IsoQuant Assay, capillary isoelectric focusing (cEEF), SDS- PAGE (reduced and non-reduced), and bioactivity assessments (ELISA).
  • SEC-HPLC performed on samples after four weeks of incubation at 45°C showed that clipping is a major degradation pathway for the liquid formulation, as shown in Figure 1 A by the percent of clips recovered at various pH levels with SEC.
  • Figure IB Both the percent of clips and the percent of aggregates (Figure IB) determined by SEC were reduced at the midrange pH values of 5.5 to 6.5.
  • Figure 2 shows the percent of degradation obtained at various pH levels as assessed by cEEF following a four- week incubation of the sample at 45°C. Minimal degradation was obtained at a pH value of about 5.5.
  • Figure 3 shows the percent of iso-aspartic acid formed at various pH levels as assessed by the Promega IsoQuant kit following a four- week incubation of the sample at 45 °C. Iso-aspartic acid formation (deamidation) was minimized at pH values of 6 and 6.5, and increased sharply at pH 8.0.
  • Figure 4 shows the effect of different buffers over time on potency following incubation at 37°C.
  • Highest stability of the antibody formulation was achieved through 8 weeks with 50 mM sodium succinate buffer at pH 6.0.
  • Formulations in histidine alone rapidly (less than 8 weeks) lost their potency as the buffer oxidized.
  • Potency of the formulation remained greater than 80% for at least 12 weeks in either sodium succinate buffer or histidine buffer gassed with N 2 to prevent oxidation.
  • Example 3 Screening of Excipients Objectives
  • surfactants Teween 80 ® and Tween 20 ®
  • salts NaCl and MgCl
  • antioxidants EDTA and methionine
  • amino acids glycine, lysine, serine and proline
  • co-solvents glycerol and ethanol
  • the Daclizumab antibody was in a 67 mM sodium phosphate formulation (without Tween ® 80) at a concentration of 6.6 mg/mL. This material was concentrated to about 30 mg/mL in the Pellicon II (Millipore) unit, and subsequently, buffer exchanged into two selected buffers (50 mM sodium phosphate pH 6.5, and 50 mM sodium succinate pH 6.0) using the 50 mL amicon stir cell (Millipore). During the third and final buffer exchange step, the material was also concentrated to a final concentration of -125 mg/mL. Finally, the antibody was filtered through 0.8 ⁇ m membrane (Uniflo). The post filtration protein concentration was determined to be approximately 100 mg/mL for the phosphate buffer sample and 97 mg/mL for the succinate buffer sample. The target concentration of the excipients at which they were screened is shown in
  • the formulations were prepared by either weighing the required amount of the excipients directly into the vial (e.g. all amino acids) or by preparing concentrated stock solutions of the excipients.
  • the excipients were added to 0.5 mL of the appropriate buffer solution and the pH adjusted to the desired value with either IN HC1 or 10% NaOH. Subsequently, 0.5 mL of the concentrated antibody solution in the appropriate buffer ( ⁇ 100 mg/mL) was added to attain the target concentration of 50 mg/mL. This procedure was adopted to prevent protein degradation due to direct contact with concentrated excipients.
  • the 1 mL solution was split into two vials with 0.5 mL fill each.
  • the other vial was incubated at 45°C with shaking at 100 RPM for 3 weeks and analyzed at the end of that time period.
  • Table 1 List of excipients and their concentrations as used in the study.
  • the samples were analyzed using various analytical techniques. Solution clarity was visually examined by holding the sample vials up against a black background under fluorescent lighting. The solution was inspected for insoluble species and color changes were recorded. Size exclusion chromatography was performed using a Perkin Elmer HPLC unit with diode array detection and two Tosohaas columns connected in series. The samples were diluted approximately 5 fold with the corresponding buffer to bring the concentration to about 1 mg/mL and 100 ⁇ L of the sample was injected onto the column. The sample concentration was measured by UV spectroscopy using the Perkin Elmer Lambda Bio 40 spectrophotometer.
  • the samples from the 3-week time point were analyzed by Capillary Isoelectric Focusing on the BioRAD CE (BioFocus 3000) System. All the samples were diluted to 0.25 mg/mL with water and a 1 : 1 dilution (to a final concentration of 0.125 mg/mL) was made with the pharmalyte solution containing TEMED and two internal pi markers, 8.4 and 10.1.
  • the capillary used was an eCAP with neutral coating (Beckmann, 56 cm length, 50 urn ED).
  • the potency of samples formulated in the succinate buffer with the excipients, Tween-80, EDTA, NaCl and MgCl was tested after 3 weeks of incubation at 5 and 45°C. It was a bio-assay involving KIT-225-K6 cells.
  • succinate buffer all samples except the amino acid containing formulations were clear after three weeks of incubation at 5°C. The samples with proline and lysine were the most turbid. At 45°C, all samples in succinate buffer were clear at the 3 week time point.
  • Table 3 indicates the % monomer for all samples being investigated in this study.
  • no significant change was observed in the % monomer for the 5°C samples in both buffers.
  • all samples indicated a slight drop in the % monomer ( ⁇ 5%).
  • the % monomer varied from 94.08 (methionine) to 97.29 (proline)
  • the % monomer varied from 95.86 (methionine) to 97.55 (Tween-80).
  • the metliionine and glycine containing formulations showed the most significant drop in % monomer. The decrease in % monomer was mostly due to clip formation.
  • Table 3B lists the % aggregate formation in all samples being investigated in this study. It is clear from these results that the increase in aggregate formation during the 3- week duration is minimal for all samples at 5°C in both buffers. After 3 weeks of incubation at 45°C, samples in the phosphate buffer showed an increase in % aggregate ranging from 0.40% (EDTA) to 2.40% (glycine). In the succinate buffer, the aggregate formation was slightly lower; ranging from 0.7 % (metliionine) to 1.09% (glycine) after the 3 week incubation period.
  • One of the hypotheses that supports these results is that if aggregate formation is due to oxidation, it maybe slowed down in the succinate buffer due to the metal chelating properties of the succinate buffer.
  • Table 3C lists the % clip formation in all samples being investigated in this study.
  • the % clipping ranged from -0.2-0.4 % in all samples.
  • the % increase in clips was insignificant over the 3-week period.
  • the % clipping varied from 4.74 (methionine) to 1.5 % (proline, glycerol and ethanol), while in the succinate buffer, the range was 1.48 %(Tween- 80) to 3.44 (methionine).
  • an increase in the clip formation was observed in the amino acid containing formulations.
  • the rate of clip formation appears to be higher in the phosphate buffer. This may be attributed to the pH difference of the Na- succinate and Na-phosphate buffers (pH 6.0 and 6.5, respectively), indicating base catalyzed hydrolysis as being the primary reason for clip formation
  • the stability of formulation was higher in the Na- succinate buffer at pH 6.0, compared with the Na-phosphate buffer at pH 6.5. This is primarily due to base-catalyzed hydrolysis that is accelerated at the higher pH of 6.5, causing an increase in the rate of clip formation.
  • the Na-succinate buffer at pH 6.0 is the selected buffer for all future studies. Results of this study also clearly indicated that in both buffers, the amino acids (glycine, lysine, serine, proline, and methionine) did not have a stabilizing effect on the protein stability. As shown by the data on sample clarity, all amino acid containing formulations indicated the formation of insoluble aggregates at 45°C.
  • the excipient MgCl 2 was selected in this study based on the hypothesis that it might protect the protein against dimidiation. While MgCl 2 precipitated in the Na-phosphate buffer; in the Na-succinate buffer, based on the cEEF data, MgCl 2 has a stabilizing effect on the protein. Ethanol was also included as an excipient to test if it stabilized the protein against deamidation by lowering the dielectric constant of the solution. The results, however, do not support this hypothesis. Finally, Tween-80, EDTA, and NaCl, the excipients most commonly used to stabilize protein formulations, did not show any destabilizing effect on the protein in either buffer.
  • Formulations 1 and 2 were prepared according to Example 3.
  • Formulation 1 100 mg/ml Daclizumab antibody, 30 mM sodium succinate (pH 6.0) 100 mM NaCl and 0.03% Tween ® -80.
  • Formulation 2 same as Formulation 1, plus 0.05% EDTA.
  • Formulation 3 100 mg/ml Daclizumab antibody, 50 mM histidine (pH 6.0), 115 mM NaCl, 0.03% Tween ® -80, purged with nitrogen.
  • Formulation 4 same as Formulation 3, plus 0.05% EDTA.
  • a liquid antibody formulation of 100 mg/ml Daclizumab in 30 mM sodium succinate, pH 6, 100 mM NaCl, and 0.03% Tween ® 80 was tested for stability after one year storage at 25°C. The stability results indicate that the formulation is stable for at least one year at 25°C (Table 7). Table 7. Stability Results of Daclizumab formulation after One- Year Storage at 25 °C.
  • a liquid antibody formulation of 100 mg/ml Daclizumab in 30 mM sodium succinate, pH 6, 100 mM NaCl, and 0.03% Tween ® 80 was incubated at 5°C (2-8°C) and tested for stability at different time points. The stability results indicate that the formulation is stable for at least 18 months at refrigerated temperature (Table 8).
  • Example 8 Stability data of HAIL-12 (histidine buffer) HAIL-12 (anti-EL12 antibody, 50 mg/mL) was formulated in 50 mM Histidine buffer, 120 mM sodium chloride, 0.03% Tween 80, pH 6.0. The on-going stability testing indicates that the formulation is stable at 5°C for at least 9 months (Table 9). Table 9. Stability Results of HAIL-12 at 5 °C.
  • HAIL-12 (50 and 100 mg/mL) was formulated in 40 mM Na-succinate buffer, 100 mM NaCl, and 0.03% Tween-80, pH 6.0.
  • the ongoing stability testing indicates that the formulation is stable at 5, 25, and 37°C for at least 12 weeks (Tables 10 and 11).
  • HuEP5C7 anti-L selectin antibody, 50 and 100 mg/mL was formulated in 50 mM histidine buffer, 125 mM sodium chloride, 0.01% Tween 80, pH 6.0.
  • the on-going stability testing indicates that the formulation is stable for three months at 25°C and 45°C and for at least 9 months at 5°C.
  • the results of the 9-month stability testing at 5°C is shown in Tablel2.
  • the results of the 3 -month accelerated stability testing is shown in Tablel3.

Abstract

This invention is directed to a stable liquid pharmaceutical formulation comprising a high concentration, e.g. 50 mg/ml or more, of antibody in about 20-60 mM succinate buffer or 30-70 mM histidine buffer, having pH from about pH 5.5 to about pH 6.5, about 0.01-0.1 % polysorbate, and a tonicity modifier that contributes to the isotonicity of the formulation. This liquid formulation is stable at refrigerated temperature (2-8°C) for at least 1 year, and preferably 2 years. This liquid formulation is suitable for subcutaneous injection. This invention is exemplified by Daclizumab, a humanized anti-IL-2 receptor monoclonal antibody; HAIL-12, a humanized anti-IL-12 monoclonal antibody; HuEP5C7, a humanized anti-L selectin monoclonal antibody; and Flintozumab, a humanized anti-gamma interferon monoclonal antibody.

Description

STABLE LIQUID PHARMACEUTICAL FORMULATION OF IGG ANTIBODIES
FIELD OF THE INVENTION
The present invention relates generally to the field of pharmaceutical formulation of antibodies. Specifically, the present invention relates to a stable, liquid, high concentration antibody formulation. This invention is exemplified by a stabilized liquid formulation of Daclizumab, an anti-IL2 receptor antibody; HAIL-12, a humanized anti-JX- 12 monoclonal antibody; and HuEP5C7, a humanized anti-L selectin monoclonal antibody.
BACKGROUND OF THE INVENTION
Many protein preparations intended for human use require stabilizers to prevent denaturation, aggregation and other alternations to the proteins prior to the use of the preparation. This instability is manifested in the formation of soluble/insoluble particles, and is often increased when the protein preparation is stored over time and during shipping. A major aim in the development of protein drug formulations is to maintain both protein solubility, stability and bioactivity.
Immunoglobulins, in particular, are recognized as possessing characteristics that tend to form aggregates and particulates in solution, and as such, may require filtration before use for intravenous or subcutaneous injection. The formation of protein aggregates and particulates has long been a problem in the development of parenteral immunoglobulin products, especially when the immunoglobulins are formulated at high concentrations. Synagis™ (Medrmmiine) is a humanized monoclonal IgGl antibody produced by recombinant DNA technology, directed to an epitope in the A antigenic site of the T protein of respiratory syncytial virus (RSN). Synagis™ is a composite of human (90%) and murine ( 10%) antibody sequences. Synagis™ is supplied as a sterile lyophilized product for reconstruction with sterile water for injection. Reconstituted Synagis™ is to be administered by intramuscular injection only. Upon reconstitution, Synagis™ contains the following excipients: 47mM histidine, 3.0 mM glycine, 5.6% mannitol, and the active ingredient, IgGl antibody, at a concentration of 100 milligrams per vial. The reconstituted Synagis™ is to be administered within 6 hours of reconstitution. WO 89/11297 discloses a lyophilized monoclonal antibody formulation comprising a lyophilized formulation of 1-25 mg/ml IgG monoclonal antibody, 2-10% maltose, and sodium acetate, phosphate, or citrate buffer having a pH between 3.0 to 6.0.
WO 97/45140 discloses an aqueous preparation of anti-CD4 antibody concentrated to approximately 100 mg/ml in 100 mM sodium citrate, 0.05 mM EDTA, pH 6.0. The application discloses a slight rise in turbidity after concentration of the antibody, which likely reflects protein aggregation. Removing this aggregation requires addition of Polysorbate 80 and sterile filtration.
WO 90/11091 discloses injectable aqueous compositions comprising about 5 mg/ml of IgM, 2.5-5 % (w/v) human serum albumin, in 8-20 mM phosphate buffer, 270 mM sodium chloride, pH 6.8-7.4.
U.S. Patent No. 6,171,586 discloses a stable aqueous pharmaceutical formulation comprising a therapeutically effective amount of an antibody not subjected to prior lyophilization, an acetate buffer from about pH 4.8 to about 5.5, a surfactant, and a polyol, wherein the formulation lacks a tomcifying amount of sodium chloride.
U.S. Patent Application Publication No. US 2001/0014326A1 discloses a pre- lyophilized antibody formulation containing 25 mg/ml anti-IgE antibody, 5 mM histidine, pH 6.0, 85 mM sucrose, and 0.01% polysorbate 20.
U.S. Patent No. 5,744,132 discloses a composition comprising 1-1000 μg/ml IL-12 antibody, 2% sucrose, 4.15% mannitol, lOmM sodium succinate, and about 0.02% Tween® 20, having a pH of about 5.6.
U.S. Patent No. 6,267,958 discloses a reconstituted formulation of 100 mg/ml rhuMab E25, in 20 mM histidine, pH 6.0, 340 mM sucrose, 0.04 % polysorbate 20, and 0.9 % benzyl alcohol. U.S. Patent No. 6,165,467 discloses a process for stabilizing a human monoclonal antibody composition produced by hybridoma cell line having accession number HB8307, which comprises dialyzing the human monoclonal antibody in a phosphate salt stabilized buffer solution having a pH from 7.2 to 7.4, said solution comprising 1-20 mg of D- mannitol per mg of said monoclonal antibody, 0.005-0.2 millimole of glycine per mg of said monoclonal antibody, and an amount of pH stabilizing phosphate salt to stabilize the pH of said solution.
There is a need for a stable liquid antibody preparation, wherein the antibody concentration is 50 mg/ml or greater; such preparation is suitable for parenteral administration, including intravenous, intramuscular, intraperitoneal, or subcutaneous injection to a human.
SUMMARY OF THE INVENTION This invention is directed to a stable liquid pharmaceutical formulation comprising a high concentration, e.g., greater than 50 mg/ml, of an antibody in 20-60 mM succinate buffer or 30-70 mM histidine buffer (pH from about pH 5.5 to about pH 6.5), a tonicity modifier, and about 0.01- 0.1 % polysorbate. This formulation retains the physical, chemical, and biological stability of antibody and prevents the immunoglobulins intended for administration to human subjects from forming aggregates and particulates in the final product. Preferred antibodies of this invention include Daclizumab, a humanized anti-IL-2 receptor monoclonal antibody; HATL-12, a humanized anti-IL-12 monoclonal antibody; and HuEP5C7, a humanized anti-L selectin monoclonal antibody; and Flintozumab, a humanized anti-gamma interferon monoclonal antibody. The liquid antibody formulation is stable at refrigerated temperature (2-8°C) for at least 1 year and preferably 2 years. This liquid formulation is also stable at room temperature (23-27°C) for at least six months. This liquid formulation is suitable for subcutaneous injection.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 A shows the percent clips formation, and Figure IB shows the percent aggregates, at various pH levels following a four-week incubation of the sample at 45°C, as assessed by SEC-HPLC.
Figure 2 shows the percent of degradation obtained at various pH levels as assessed by cIEF following a four- week incubation of the sample at 45°C.
Figure 3 shows the percent of iso-aspartic acid formed at various pH levels as assessed by the Promega IsoQuant kit following a four- week incubation of the sample at 45°C.
Figure 4 shows the effect of different buffers over time on potency following incubation at 37°C. DETAILED DESCRIPTION OF THE INVENTION I. Definition
As used herein, the term "buffer" encompasses those agents which maintain the solution pH in an acceptable range and may include succinate (sodium), histidine, phosphate' (sodium or potassium), Tris (tris (hydroxymethyi) aminomethane), diethanolamine, and the like. The buffer of this invention has a pH in the range from about 5.5 to about 6.5; and preferably has a pH of about 6.0. Examples of buffers that will control the pH in this range include succinate (such as sodium succinate), gluconate, histidine, citrate phospate and other organic acid buffers. "Pharmaceutically acceptable excipients" (vehicles, additives) are those inert substances that can reasonably be administered to a subject mammal and provide an effective dose of the active ingredient employed. These substances are added to a formulation to stabilize the physical, chemical and biological structure of the antibody. The term also refers to additives that may be needed to attain an isotonic formulation, suitable for the intended mode of administration.
The term "pharmaceutical formulation" refers to preparations which are in such form as to permit the biological activity of the active ingredients to be unequivocally effective, and which contain no additional components which are toxic to the subjects to which the formulation would be administered. A "stable" formulation is one in which the protein therein essentially retains its physical stability, chemical stability, and biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10:29-90 (1993). Stability can be measured at a selected temperature for a selected time period. A "stable" liquid antibody formulation is a liquid antibody formulation with no significant changes observed at a refrigerated temperature (2-8 °C) for at least 12 months, preferably 2 years, and more preferably 3 years; or at room temperature (23 -27 °C) for at least 3 months, preferably 6 months, and more preferably 1 year. The criteria for stability are as follows. No more than 10%, preferably 5%, of antibody monomer is degraded as measured by SEC-HPLC. The solution is colorless, or clear to slightly opalescent by visual analysis. The concentration, pH and osmolality of the formulation have no more than +/- 10% change. Potency is within 70-130%, preferably 80-120% of the control. No more than 10%, preferably 5% of clipping (hydrolysis) is observed. No more than 10%, preferably 5% of aggregation is formed.
An antibody "retains its physical stability" in a pharmaceutical formulation if it shows no significant increase of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering, size exclusion chromatography (SEC-HPLC) and dynamic light scattering. In addition the protein conformation is not altered. The changes of protein conformation can be evaluated by fluorescence spectroscopy, which determines the protein tertiary structure, and by FTIR spectroscopy, which determines the protein secondary structure. An antibody "retains its chemical stability" in a pharmaceutical formulation, if it shows no significant chemical alteration. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein. Degradation processes that often alter the protein chemical structure include hydrolysis or clipping (evaluated by methods such as size exclusion chromatography and SDS-PAGE), oxidation (evaluated by methods such as by peptide mapping in conjunction with mass spectroscopy or MALDI/TOF/MS), deamidation (evaluated by methods such as ion-exchange chromatography, capillary isoelectric focusing, peptide mapping, isoaspartic acid measurement), and isomerization (evaluated by measuring the isoaspartic acid content, peptide mapping, etc.).
An antibody "retains its biological activity" in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within a predetermined range of the biological activity exhibited at the time the pharmaceutical formulation was prepared. The biological activity of an antibody can be determined, for example, by an antigen binding ELISA assay.
The term "isotonic" means that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 270-328 mOsm. Slightly hypotonic osmotic pressure is 250-269 and slightly hypertonic osmotic pressure is 328-350 mOsm. Osmotic pressure can be measured, for example, using a vapor pressure or ice-freezing type osmometer.
"Tonicity modifiers" are those pharmaceutically acceptable inert substances that can be added to the formulation to provide an isotonity of the formulation. Tonicity modifiers suitable for this invention include salts and amino acids. II Analytical Methods
The following criteria are important in developing a stable pharmaceutical antibody formulation. The antibody formulation contains pharmaceutically acceptable excipients. The antibody formulation is formulated such that the antibody retains its physical, chemical and biological activity. The formulation is preferably stable for at least 1 year at refrigerated temperature (2-8°C) and 6 months at room temperature (23-27°C).
The analytical methods for evaluating the product stability include size exclusion chromatography (SEC-HPLC), dynamic light scattering test (DLS), differential scanning calorimetery (DSC), iso-asp quantification, potency, UV at 340nm, and UV spectroscopy. SEC (J. Pharm. Scien., 83:1645-1650, (1994); Pharm. Res., 11:485 (1994); J. Pharm. Bio. Anal., 15:1928 (1997); J. Pharm. Bio. Anal, 14:1133-1140 (1986)) measures percent monomer in the product and gives information of the amount of soluble aggregates and clips. DSC (Pharm. Res., 15:200 (1998); Pharm. Res., 9:109 (1982)) gives information of protein denaturation temperature and glass transition temperature. DLS (American Lab., Nov. (1991)) measures mean diffusion coefficient, and gives information of the amount of soluble and insoluble aggregates. UV at 340nm measures scattered light intensity at 340nm and gives information about the amounts of soluble and insoluble aggregates. UN spectroscopy measures absorbance at 278nm and gives information of protein concentration.
The iso-Asp content in the samples is measured using the Isoquant Isoaspartate Detection kit (Promega). The kit uses the enzyme Protein Isoaspartyl Methyltransferase (PLMT) to specifically detect the presence of isoaspartic acid residues in a target protein. PIMT catalyzes the transfer of a methyl group from S-adenosyl-L-methionine to isoaspartic acid at the ce-carboxyl position, generating S-adenosyl-L-homocysteine (SAH) in the process. This is a relatively small molecule, and can usually be isolated and quantitated by reverse phase HPLC using the SAH HPLC standards provided in the kit.
The potency or bioactivity of an antibody can be measured by its ability to bind to its antigen. The specific binding of an antibody to its antigen can be quantitated by any method known to those skilled in the art, for example, an immunoassay, such as ELISA (enzyme-linked immunosorbant assay). III. Preparation of Antibody
The invention herein relates to a stable aqueous formulation comprising an antibody. The antibody in the formulation is prepared using techniques available in the art for generating antibodies, exemplary methods of which are described in more detail in the following sections.
The antibody is directed against an antigen of interest. Preferably, the antigen is a biologically important polypeptide and administration of the antibody to a mammal may prevent or treat a disorder. However, antibodies directed against nonpolypeptide antigens (such as tumor-associated glycolipid antigens; see U.S. Pat. No. 5,091,178) are also contemplated.
Where the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor. Exemplary antigens include molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha- 1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VDTC, factor LX, tissue factor, and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophage inflammatory protein (MIP-1 -alpha); a serum albumin such as human serum albumin; Muellerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; a microbial protein, such as beta- lactamase; DNase; IgE; a cytotoxic T-lymphocyte associated antigen (CTLA), such as CTLA-4; inhibin; activin; vascular endothelial growth factor (NEGF); receptors for hormones or growth factors; protein A or D; rheumatoid factors; a neurotrophic factor such as bone-derived neurotrophic factor (BDΝF), neurotrophin-3, -4, -5, or -6 (ΝT-3, NT4, NT-5, or NT-6), or a nerve growth factor such as NGF-β; platelet-derived growth factor (PDGF); fibroblast growth factor such as aFGF and bFGF; epidermal growth factor (EGF); transforming growth factor (TGF) such as TGF-α and TGF-β, including TGF-βb TGF-β2, TGF-β3, TGF-β4, or TGF-β5; insulin-like growth factor-I and -II (IGF-I and IGF- U); des(l-3)-IGF-I (brain IGF-I), insulin-like growth factor binding proteins; CD proteins such as CD3, CD4, CD8, CD19 and CD20; erythropoietin; osteoinductive factors; immunotoxins; a bone morphogenetic protein (BMP); an interferon such as interferon-α, - β, and -γ; colony stimulating factors (CSFs), e.g., M-CSF, GM-CSF, and G-CSF; interleukins (ILs), e.g., IL-1 to IL-12; receptors to interleukins IL-1 to IL-12; selectins such as L, E, and P-selectin; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor; viral antigen such as, for example, a portion of the AIDS envelope; transport proteins; homing receptors; addressins; regulatory proteins; integrins such as GDI la, CD1 lb, CD1 lc, CD18, an ICAM, NLA-4 and NCAM; a tumor associated antigen such as HER2, HER3 or HER4 receptor; and fragments of any of the above-listed polypeptides.
When using recombinant techniques, the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed cells, is removed, for example, by centrifugation or ultrafiltration. Where the antibody is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants. The antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique. The suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody. Protein A can be used to purify antibodies that are based on human Y1; T2, or Y heavy chains (Lindmark et al, J.
Immunol. Meth. 62:1-13 (1983)). Protein G is recommended for all mouse isotypes and for human Y3 (Guss et al, EMBOJ. 5:1567-1575 (1986)). The matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody comprises a CH3 domain, the Bakerbond ABX™ resin (J. T. Baker, Phillipsburg, Ν. J.) is useful for purification. Other techniques for protein purification such as fractionation on an ion-exchange column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin SEPHAROSET™ chromatography on an anion or cation exchange resin (such as a polyaspartic acid column), chromatofocusing, SDS-PAGE, and ammonium sulfate precipitation are also available depending on the antibody to be recovered.
Preferred antibodies encompassed by the present invention include Daclizumab (USAN, United States Adopted Names), a humanized anti-IL-2 receptor antibody. Daclizumab is currently being marketed as Zenapax® for the prevention of organ rejection after renal transplantation and is administered through the intravenous route. Daclizumab is also useful for treating psoriasis, for which, the subcutaneous delivery is the preferred route of administration. For a subcutaneous delivery of antibody, high concentration of antibody is preferred. Daclizumab is a recombinant humanized monoclonal antibody, subclass IgGl. The molecule is composed of two identical heavy chain and two identical light chain subunits. Disulfide bridges link the four chains. Daclizumab monomer is approximately 150,000 daltons in molecular weight. Daclizumab binds to the p55 subunit of the IL-2 receptor expressed on activated T cells. The antigen target is designated CD25. Daclizumab is produced from a GS-NS0 cell line containing the heavy and light chain genes by fed-batch fermentation culture. Bioreactor harvests are processed to remove cells and debris and purified using a combination of ion-exchange and gel filtration chromatography and a series of ultrafiltration and filtration techniques to produce drug substance containing greater than 95% monomeric species.
Another preferred antibody is anti-interleukin 12 (IL-12) antibody. IL-12 is a cytokine synthesized by antigen presenting cells. It is composed of two subunits (p35 and p40), both must be present for functional activity. Functional EL- 12 is also called EL- 12p70. This cytokine preferentially acts on T helper cell type 1 (Thl) lymphocytes and natural killer cells by increasing their proliferative rate. One downstream effect is the secretion of interferon gamma (IFNg) by Thl cells. Both of these functions (proliferative and EFNg production) can be easily assayed for and were used to detect IL-12 activity in samples. Certain antibodies to JL-12 have been shown to "neutralize" the above activities. Since Thl cells have been implicated in playing a pivotal role in a variety of diseases, an antibody with neutralizing characteristics would have potential therapeutic value. 16G2 (Hoffman La Roche) is a murine antibody raised against LL-12p70. 16G2 has been shown to act in near stoichiometric amounts to EL- 12 in a functional assay-the inhibition of proliferation of activated T cells from human peripheral blood (PBMC). This is an important characteristic because p40 dimers of EL- 12 exist in serum and antibodies raised to the p40 subunit need to be used in excess amounts to neutralize the proliferative capacity of a given amount of EL- 12. 16G2 was humanized at Protein Design Labs. (Fremont, CA) to give rise to HAEL-12 (humanized anti-EL-12, an EgGl antibody).
Another preferred antibody is anti-L selectin antibody. Selectins, such as L, E, and P-selectin have been found to be associated with tissue damage during the course of ischemia and reperfusion. Neutrophils play an important role in this connection. It is assumed that selectin is required for the recruitment of neutrophils. L-selectin is important for the complete development of damage in skeletal muscle as well as in the lung (Seekamp, et al, Am. J. Pathol 11:592-598 (1994). Mulligan, et al, J. Immunol. 151 :832-840 (1994). HuEP5C7 (SMART Anti-L Selectin) is a humanized anti-L selectin monoclonal antibody, that contains mutant IgG2 Fc, cross reacts with both human E and P selectin antigens. It is currently being developed by Protein Design Labs, Inc. for various indications such as asthma, stroke, trauma, and certain autoimmune diseases.
Another preferred antibody is Flintozumab, an anti-gamma interferon antibody. Flintozumab is an IgGl humanized monoclonal antibody developed by Protein Design Labs, Inc. for the treatment of immune disorders mediated by interferon-gamma (EFN-g), a proinflammatory cytokine. EFN-g induces the expression of major histocompatibility complex (MHC) class I and/or class U (HLA-DR) antigens, enhances the cytolytic activity of natural killer cells, activates macrophages, and modulates the immunoglobulin isotype profile of the humoral response. As a lymphokine, EFN-g also enhances the development of T helper cell type 1 (Thl), while suppressing the development of T helper cell type 2 (Th2) cells. Aberrations in the Thl/Th2 ratio have been implicated in a variety of autoimmune conditions.
IV. Preparation of the Formulation
After the antibody of interest is prepared as described above, a pharmaceutical formulation comprising the antibody is prepared. The formulation development approach is as follows: selecting the optimum solution pH, selecting buffer type and concentration, evaluating the effect of various excipients of the liquid stability, and optimizing the concentration of the screened excipients using an I-optimal experimental design (Statistics for Experimenters: An Introduction to Design, Data Analysis, and Model Building, Box,
George E.P. et al., John Wiley and Sons, Inc., 1978).
The compositions of this invention minimize the formation of antibody aggregates and particulates and insure that the antibody maintains its bioactivity over time. The composition is a pharmaceutically acceptable liquid formulation containing a high concentration of an antibody in a buffer having a neutral or slightly acidic pH (pH 5.5-6.5), a surfactant, and a tonicity modifier.
The antibody in the composition is a high concentration of 50 mg/ml or greater, preferably 100 mg/ml or greater. A preferred composition of this invention contains Daclizumab, a humanized anti-EL2 receptor antibody; HAEL12, a humanized anti-EL-12 antibody; HaEP5C7, a humanized anti-L selectin antibody; and Flintozumab, a humanized anti-gamma interferon antibody.
A buffer of pH 5.5-6.5 is used in the composition. A buffer of pH 6.0-6.5 is preferred. Examples of buffers that control the pH in this range include succinate (such as sodium succinate), gluconate, histidine, citrate, phosphate, and other organic acid buffers.
Succinate (pKa 5.63) is a preferred buffer for subcutaneous injection. Histidine (pK 5.97) is less preferred because of its susceptibility to oxidization, although such oxidation can be retarded by replacing the vial headspace with N2 or adding an antioxidant. Citrate and phosphate buffers are much less preferred because it causes a painful reaction when injected subcutaneously. A preferred buffer contains about 20-60 mM sodium succinate.
Another preferred buffer is 30-70 mM histidine buffer overlaid with N2.
A surfactant is also added to the antibody formulation. Exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbates 20, 80, such as
Tween® 20, Tween® 80) or poloxamers (e.g. poloxamer 188). The amount of surfactant added is such that it reduces aggregation of the formulated antibody and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. The surfactant may be present in the formulation in an amount from about 0.005% to about 0.5%, preferably from about 0.01% to about 0.1%, more preferably from about 0.01% to about 0.05%, and most preferably from about 0.02% to about 0.04%. A tonicity modifier, which contributes to the isotonicity of the formulations, is added to the present composition. The tonicity modifier useful for the present invention includes salts and amino acids. Salts that are pharmaceutically acceptable and suitable for this invention include sodium chloride, sodium succinate, sodium sulfate, potassium chloride, magnesium chloride, magnesium sulfate, and calcium chloride. Preferred salts for this invention are NaCl and MgCl2. MgCl2 may also improve the antibody stability by protecting the protein from deamidation. A preferred concentration of NaCl is about 75- 150 mM. A preferred concentration of MgCl2 is about 1-100 mM. Amino acids that are pharmaceutically acceptable and suitable for this invention include proline, alanine, L- arginine, asparagine, L-aspartic acid, glycine, serine, lysine, and histidine. A preferred amino acid for this invention is proline. A preferred concentration of proline is than 200 mM.
EDTA, which is commonly used to stabilize a protein formulation, may also be included in the formulation. EDTA, as a chelating agent, may inhibit the metal-catalyzed oxidation of the sulfhydryl groups, thus reducing the formation of disulfide-linked aggregates. A preferred concentration of EDTA is 0.01-0.2%.
/ Exemplary liquid compositions are formulations comprising antibody at about 100 mg/ml or greater, about 20-60 mM sodium succinate (pH 6), about 0.01-0.1% polysorbate 20 or 80, and about 75-150 mM NaCl. This formulation retains the stability of biological activity of the monoclonal antibody, and prevents the immunoglobulins intended for administration to human subjects from physical, chemical and biological degradation in the final product.
The liquid antibody formulation of this invention is suitable for parenteral administration such as intravenous, intramuscular, intraperitoneal, or subcutaneous injection; particularly suitable for subcutaneous injection.
The invention is illustrated further by the following examples, which are not to be construed as limiting the invention in scope of the specific procedures described in them.
EXAMPLES
Example 1 : Optimization of pH
To identify the optimum formulation for pH range and to identify major degradation pathways, a pH profile study was conducted. Sample formulations contained 5.0 mg/ml anti-EL2 receptor antibody (Daclizumab) in one of three buffers: 50 mM sodium acetate buffer at pH 4.0 or 5.0, 50 mM histidine at pH 5.5, 6.0, or 6.5, or 50 mM sodium phosphate buffer at pH 7.0 or 8. 5. Independent formulations were incubated at either 5°C or 45° C with 100 RPM shaking for 4 weeks. The physical and chemical stability of each sample was assessed at 0 and 4 weeks by analytical methods including: pH and visual analysis, UV spectroscopy at 340 nm, size exclusion chromatography (SEC- HPLC), fluorescence spectroscopy, dynamic light scattering (DLS), differential scanning calorimetry (DSC), Promega IsoQuant Assay, capillary isoelectric focusing (cEEF), SDS- PAGE (reduced and non-reduced), and bioactivity assessments (ELISA). SEC-HPLC performed on samples after four weeks of incubation at 45°C showed that clipping is a major degradation pathway for the liquid formulation, as shown in Figure 1 A by the percent of clips recovered at various pH levels with SEC. Both the percent of clips and the percent of aggregates (Figure IB) determined by SEC were reduced at the midrange pH values of 5.5 to 6.5. Figure 2 shows the percent of degradation obtained at various pH levels as assessed by cEEF following a four- week incubation of the sample at 45°C. Minimal degradation was obtained at a pH value of about 5.5.
Figure 3 shows the percent of iso-aspartic acid formed at various pH levels as assessed by the Promega IsoQuant kit following a four- week incubation of the sample at 45 °C. Iso-aspartic acid formation (deamidation) was minimized at pH values of 6 and 6.5, and increased sharply at pH 8.0.
The results from this experiment indicate that pH 5.5 to 6.5 and preferable pH 6.0 to 6.5, are the optimal pH which minimize antibody degradation and aggregation.
Example 2: Optimization of Buffers
En this experiment, independent formulations contained 5.0 mg/ml Daclizumab antibody in 50 mM sodium succinate, pH 6.0; and 50 mM histidine, pH 6.0, with and without N2 gassing. Sodium citrate buffer was not included because of reports of pain on subcutaneous injection. The bioactivity (potency) at time 0, and after 4, 8, and 12 weeks of incubation at 37° C was measured by ELISA using microplates coated with recombinant human JL2 alpha receptor (EL-2 sRα) antigen, and goat anti-human IgG-HRP conjugate.
Figure 4 shows the effect of different buffers over time on potency following incubation at 37°C. Highest stability of the antibody formulation was achieved through 8 weeks with 50 mM sodium succinate buffer at pH 6.0. Formulations in histidine alone rapidly (less than 8 weeks) lost their potency as the buffer oxidized. Potency of the formulation remained greater than 80% for at least 12 weeks in either sodium succinate buffer or histidine buffer gassed with N2 to prevent oxidation. Example 3 : Screening of Excipients Objectives
This study was conducted to screen various excipients for the formulation of Daclizumab antibody at 50 mg/mL. From the pH optimization study conducted earlier (Example 1), the formulation stability was maximized in the pH range of 6.0-6.5. Therefore in this study, excipients were screened in two buffers; 50 mM phosphate, pH 6.5 and 50 mM succinate, pH 6.0. The stability of antibody was monitored in the two buffers for 3 weeks at 5°C and 45 °C with shaking at 100 RPM at a concentration of 50 mg/mL. The excipients examined included: surfactants (Tween 80® and Tween 20®), salts (NaCl and MgCl ), antioxidants (EDTA and methionine), amino acids (glycine, lysine, serine and proline), and co-solvents (glycerol and ethanol). Various analytical techniques (clarity, pH, SEC-HPLC, UV-Vis, and cEEF) were used to characterize the excipient-containing formulations. Sample Preparation
The Daclizumab antibody was in a 67 mM sodium phosphate formulation (without Tween® 80) at a concentration of 6.6 mg/mL. This material was concentrated to about 30 mg/mL in the Pellicon II (Millipore) unit, and subsequently, buffer exchanged into two selected buffers (50 mM sodium phosphate pH 6.5, and 50 mM sodium succinate pH 6.0) using the 50 mL amicon stir cell (Millipore). During the third and final buffer exchange step, the material was also concentrated to a final concentration of -125 mg/mL. Finally, the antibody was filtered through 0.8 μm membrane (Uniflo). The post filtration protein concentration was determined to be approximately 100 mg/mL for the phosphate buffer sample and 97 mg/mL for the succinate buffer sample. The target concentration of the excipients at which they were screened is shown in
Table 1. The formulations were prepared by either weighing the required amount of the excipients directly into the vial (e.g. all amino acids) or by preparing concentrated stock solutions of the excipients. The excipients were added to 0.5 mL of the appropriate buffer solution and the pH adjusted to the desired value with either IN HC1 or 10% NaOH. Subsequently, 0.5 mL of the concentrated antibody solution in the appropriate buffer (~100 mg/mL) was added to attain the target concentration of 50 mg/mL. This procedure was adopted to prevent protein degradation due to direct contact with concentrated excipients. The 1 mL solution was split into two vials with 0.5 mL fill each. One vial was used for initial T=0 analysis and then stored at 2-8°C for the 3 week time point analysis at 2-8 °C. The other vial was incubated at 45°C with shaking at 100 RPM for 3 weeks and analyzed at the end of that time period.
Table 1: List of excipients and their concentrations as used in the study.
Figure imgf000016_0001
Analytical Methods
At each of the two time points, the samples were analyzed using various analytical techniques. Solution clarity was visually examined by holding the sample vials up against a black background under fluorescent lighting. The solution was inspected for insoluble species and color changes were recorded. Size exclusion chromatography was performed using a Perkin Elmer HPLC unit with diode array detection and two Tosohaas columns connected in series. The samples were diluted approximately 5 fold with the corresponding buffer to bring the concentration to about 1 mg/mL and 100 μL of the sample was injected onto the column. The sample concentration was measured by UV spectroscopy using the Perkin Elmer Lambda Bio 40 spectrophotometer.
The samples from the 3-week time point were analyzed by Capillary Isoelectric Focusing on the BioRAD CE (BioFocus 3000) System. All the samples were diluted to 0.25 mg/mL with water and a 1 : 1 dilution (to a final concentration of 0.125 mg/mL) was made with the pharmalyte solution containing TEMED and two internal pi markers, 8.4 and 10.1. The capillary used was an eCAP with neutral coating (Beckmann, 56 cm length, 50 urn ED). The potency of samples formulated in the succinate buffer with the excipients, Tween-80, EDTA, NaCl and MgCl was tested after 3 weeks of incubation at 5 and 45°C. It was a bio-assay involving KIT-225-K6 cells.
Results
There were 24 samples at time point T=0 as 12 different excipients were monitored in two different buffers. At the 3 week time point, there were 48 samples to be analyzed (12 different excipients x 2 temperatures x 2 buffers = 48). Assays performed include concentration determination by UV-Vis, pH, Clarity, SEC-HPLC, and CEEF.
(a) Sample Clarity
Sample appearance is indicated in Table 2. All samples were clear in both the buffers at the initial time point T = 0. At the 3 week time point, all samples in the phosphate buffer except the one containing lysine were clear at 5°C. In the same buffer, at 45°C, the samples containing amino acids (glycine, serine, proline and lysine) appeared clear but had some thread like floaties in the vials. The sample with MgCl2 had clear crystals settled in the bottom of the vial.
In the succinate buffer, all samples except the amino acid containing formulations were clear after three weeks of incubation at 5°C. The samples with proline and lysine were the most turbid. At 45°C, all samples in succinate buffer were clear at the 3 week time point.
Table 2: Sample clarity determined by fluorescence light at T=0 and T=4 weeks at 5 and 45°C in the Na-succinate (pH 6.0) and Na-phosphate (pH 6.5) buffers.
Figure imgf000018_0001
(b) SEC-HPLC
Results of SEC-HPLC are tabulated in Table 3(A-C). Table 3 A indicates the % monomer for all samples being investigated in this study. The % monomer at T=0 for all samples was >99%. At the three weeks time point, no significant change was observed in the % monomer for the 5°C samples in both buffers. However, at 45°C, all samples indicated a slight drop in the % monomer (<5%). For samples formulated in the phosphate buffer, the % monomer varied from 94.08 (methionine) to 97.29 (proline), while for the samples formulated in the succinate buffer, the % monomer varied from 95.86 (methionine) to 97.55 (Tween-80). En both the buffers, the metliionine and glycine containing formulations showed the most significant drop in % monomer. The decrease in % monomer was mostly due to clip formation.
Table 3B lists the % aggregate formation in all samples being investigated in this study. It is clear from these results that the increase in aggregate formation during the 3- week duration is minimal for all samples at 5°C in both buffers. After 3 weeks of incubation at 45°C, samples in the phosphate buffer showed an increase in % aggregate ranging from 0.40% (EDTA) to 2.40% (glycine). In the succinate buffer, the aggregate formation was slightly lower; ranging from 0.7 % (metliionine) to 1.09% (glycine) after the 3 week incubation period. One of the hypotheses that supports these results is that if aggregate formation is due to oxidation, it maybe slowed down in the succinate buffer due to the metal chelating properties of the succinate buffer.
Table 3C lists the % clip formation in all samples being investigated in this study. At the initial time point, the % clipping ranged from -0.2-0.4 % in all samples. For all samples incubated at 5°C, the % increase in clips was insignificant over the 3-week period. At 45°C, a significant increase in the rate of clip formation was observed. For samples formulated in the phosphate buffer, the % clipping varied from 4.74 (methionine) to 1.5 % (proline, glycerol and ethanol), while in the succinate buffer, the range was 1.48 %(Tween- 80) to 3.44 (methionine). In general, an increase in the clip formation was observed in the amino acid containing formulations. Further, the rate of clip formation appears to be higher in the phosphate buffer. This may be attributed to the pH difference of the Na- succinate and Na-phosphate buffers (pH 6.0 and 6.5, respectively), indicating base catalyzed hydrolysis as being the primary reason for clip formation
Table 3A: % Monomer as determined by SEC at T=0 and 3 weeks at 5°C and 45°C in the Na-succinate (pH 6.0) and Na-phosphate (pH 6.5) buffers.
Figure imgf000019_0001
Table 3B: % Aggregate as determined by SEC at T=0 and 3 weeks at 5°C and 45°C in the Na-succinate (pH 6.0) and Na-phosphate (pH 6.5) buffers.
Figure imgf000020_0001
Table 3C: % Clipping as determined by SEC at T=0 and 3 weeks at 5°C and 45°C in the Na-succinate (pH 6.0) and Na-phosphate (pH 6.5) buffers.
Figure imgf000020_0002
(c) Capillary Electrophoresis
All the samples from this study were analyzed by capillary electrophoresis (cEEF) on the BioRAD system. A typical cEEF profile of Daclizumab shows four peaks. Typically on accelerated aging at high temperatures, the area of the main isoform peak decreases followed by an increase in the other isoform peaks, which indicates the conversion of one isoform to another isoform. The % degradation is calculated by percent decrease in the peak area of the main isoform:
% Degradation = [Peak Area at T=0 - Peak Area at 45°C1 xl00%
[Peak Area at T=0]
Our results indicate that the 45°C samples are more degraded in the phosphate buffer (pH 6.5) when compared with similar samples in the succinate buffer (pH 6.0).The best electropherograms were seen for the excipients, EDTA, NaCl, lysine and MgCl2. The % degradation after 3 weeks for the 5°C versus the 45°C could not be calculated for the samples containing Tween 80, Tween 20, serine and proline as their electropherograms were very collapsed and the peaks indistinguishable.
(d) Potency Based on the results of this study, the Na-succinate buffer appears to be more promising than the Na-phosphate buffer. Thus, potency assessments were done for the most stabilizing excipients in the Na-succinate buffer only. This included the formulations containing Tween-80, EDTA, NaCl and MgCl , subject to three weeks of incubation at 5 and 45 °C. Results (Table 4) showed that the potency of all formulations was within specifications, indicating that the underlying chemical and physical degradation processes are not significantly altering the protein activity.
Table 4: Potency results of selected formulations in succinate buffer at T=3 weeks at 5 and 45°C.
Figure imgf000021_0001
Discussion
Based on the results of this study, the stability of formulation was higher in the Na- succinate buffer at pH 6.0, compared with the Na-phosphate buffer at pH 6.5. This is primarily due to base-catalyzed hydrolysis that is accelerated at the higher pH of 6.5, causing an increase in the rate of clip formation. Thus, the Na-succinate buffer at pH 6.0 is the selected buffer for all future studies. Results of this study also clearly indicated that in both buffers, the amino acids (glycine, lysine, serine, proline, and methionine) did not have a stabilizing effect on the protein stability. As shown by the data on sample clarity, all amino acid containing formulations indicated the formation of insoluble aggregates at 45°C.
The excipient MgCl2 was selected in this study based on the hypothesis that it might protect the protein against dimidiation. While MgCl2 precipitated in the Na-phosphate buffer; in the Na-succinate buffer, based on the cEEF data, MgCl2 has a stabilizing effect on the protein. Ethanol was also included as an excipient to test if it stabilized the protein against deamidation by lowering the dielectric constant of the solution. The results, however, do not support this hypothesis. Finally, Tween-80, EDTA, and NaCl, the excipients most commonly used to stabilize protein formulations, did not show any destabilizing effect on the protein in either buffer.
Further experiments were conducted in the Na-succinate buffer at pH 6.0; the effect of the excipients (MgCl2, Tween-80, NaCl, and EDTA) was further examined on the protein stability. The results indicate that to formulate an antibody at 100 mg/mL with 100 mM NaCl, the optimal concentration of Tween 80 falls in the range of 0.02-0.03%. Results also indicate that increasing the salt concentration (100-150 mM) could further stabilize the formulation. Thus, the concentration of NaCl should be maximized while maintaining the tonicity requirements. The results also indicate that the stability of the Tween 80 and NaCl containing formulation could be enhanced by adding EDTA in the concentration range of 0.35-0.5%. The addition of MgCl2 in the concentration range of 0-50 mM also could have a favorable effect. The results also indicate that the excipient concentrations for the most stable formulation are: 150 mM NaCl, 0.05% Tween 80, 0.03-0.04% EDTA and 60-70 mM MgCl2, however, these conditions are not practical because they do not provide isotonic conditions. Example 4: Stability Data of Two Daclizumab Antibody Formulations in Succinate Buffer
Formulations 1 and 2 were prepared according to Example 3. Formulation 1: 100 mg/ml Daclizumab antibody, 30 mM sodium succinate (pH 6.0) 100 mM NaCl and 0.03% Tween®-80.
Formulation 2: same as Formulation 1, plus 0.05% EDTA.
The stability results of Formulations 1 and 2 at T= 0, 2 weeks, 4 weeks, 8 weeks, and 12 weeks are shown as follows at 5, 25, and 37°C (Table 5).
Figure imgf000023_0001
T=12 Weeks
Example 5: Stability Data of Two Daclizumab Formulations in Histidine Buffer
Formulations 3 and 4 are prepared according to Example 3.
Formulation 3: 100 mg/ml Daclizumab antibody, 50 mM histidine (pH 6.0), 115 mM NaCl, 0.03% Tween®-80, purged with nitrogen.
Formulation 4: same as Formulation 3, plus 0.05% EDTA.
The stability results of Formulations 3 and 4 at T= 0, 2 weeks, 4 weeks, 8 weeks, and 12 weeks are shown as follows at 5, 25, and 37°C (Table 6).
Table 6: Stability results of Formulations 3 and 4. T=0
Figure imgf000024_0002
T=4 Weeks
Figure imgf000025_0001
Example 6. Stability Data of Daclizumab Formulation at Room Temperature for One Year
A liquid antibody formulation of 100 mg/ml Daclizumab in 30 mM sodium succinate, pH 6, 100 mM NaCl, and 0.03% Tween® 80 was tested for stability after one year storage at 25°C. The stability results indicate that the formulation is stable for at least one year at 25°C (Table 7). Table 7. Stability Results of Daclizumab formulation after One- Year Storage at 25 °C.
Figure imgf000026_0001
Example 7. Stability Data of Daclizumab Formulation at 5°C for 18 Months
A liquid antibody formulation of 100 mg/ml Daclizumab in 30 mM sodium succinate, pH 6, 100 mM NaCl, and 0.03% Tween® 80 was incubated at 5°C (2-8°C) and tested for stability at different time points. The stability results indicate that the formulation is stable for at least 18 months at refrigerated temperature (Table 8).
Table 8. Stability Results of Daclizumab at 5°C.
Figure imgf000026_0002
Example 8. Stability data of HAIL-12 (histidine buffer) HAIL-12 (anti-EL12 antibody, 50 mg/mL) was formulated in 50 mM Histidine buffer, 120 mM sodium chloride, 0.03% Tween 80, pH 6.0. The on-going stability testing indicates that the formulation is stable at 5°C for at least 9 months (Table 9). Table 9. Stability Results of HAIL-12 at 5 °C.
Figure imgf000027_0001
Example 9. Stability data of HAIL-12 (succinate buffer)
HAIL-12 (50 and 100 mg/mL) was formulated in 40 mM Na-succinate buffer, 100 mM NaCl, and 0.03% Tween-80, pH 6.0. The ongoing stability testing indicates that the formulation is stable at 5, 25, and 37°C for at least 12 weeks (Tables 10 and 11).
Table 10. Stability Results of HAIL-12 (50 mg/mL)at various temperatures. T=0
Figure imgf000027_0002
T=12 Weeks
Table 11. Stability Results of HAIL-12 (100 mg/ml) at various temperatures. T=0
Figure imgf000028_0002
Example 11. Stability data of HuEP5C7.
HuEP5C7 (anti-L selectin antibody, 50 and 100 mg/mL) was formulated in 50 mM histidine buffer, 125 mM sodium chloride, 0.01% Tween 80, pH 6.0. The on-going stability testing indicates that the formulation is stable for three months at 25°C and 45°C and for at least 9 months at 5°C. The results of the 9-month stability testing at 5°C is shown in Tablel2. The results of the 3 -month accelerated stability testing is shown in Tablel3.
Table 12. Stability Results of HuEP5C7 at 5°C.
Figure imgf000028_0001
Figure imgf000028_0003
Table 13. Stability Results of HuEP5C7 at various temperatures.
T=3 months
Figure imgf000029_0001
The invention, and the manner and process of making and using it, are now described in such full, clear, concise and exact terms as to enable any person skilled in the art to which it pertains, to make and use the same. It is to be understood that the foregoing describes preferred embodiments of the present invention and that modifications may be made therein without departing from the scope of the present invention as set forth in the claims. To particularly point out and distinctly claim the subject matter regarded as invention, the following claims conclude this specification.

Claims

What is claimed is:
1. A stable liquid pharmaceutical formulation comprising: about 20-60 mM succinate buffer having pH from about pH 5.5 to about pH 6.5, about 0.01 % - 0.1 % polysorbate, a tonicity modifier that contributes to isotonicity of the formulation, and greater than 50 mg/ml antibody.
2. A stable liquid pharmaceutical formulation comprising: about 30-70 mM histidine buffer having pH from about pH 5.5 to about pH 6.5, about 0.01 % - 0.1 % polysorbate, a tonicity modifier that contributes to isotonicity of the formulation, and greater than 50 mg/ml antibody.
3. The liquid pharmaceutical formulation according to Claim 1 or 2, wherein said antibody concentration is greater than 100 mg/ml.
4. The stable liquid pharmaceutical formulation according to Claim 1 or 2, wherein said tonicity modifier is NaCl or MgCl2.
5. The stable liquid pharmaceutical formulation according to Claim 4, wherein said NaCl is at 75-150 mM.
6. The stable liquid pharmaceutical formulation according to Claim 4, wherein said MgCl2 is at 1-100 mM.
7. The stable liquid pharmaceutical formulation according to Claim 1 or 2, wherein said pH is about pH 6.0 to 6.5.
8. The stable liquid pharmaceutical formulation according to Claim 1 or 2, wherein said polysorbate is at a concentration of about 0.02-0.04 %.
9. The stable liquid pharmaceutical formulation according to Claim 1 or 2, further comprising 0.01 to 0.5 % EDTA.
10. The stable liquid pharmaceutical formulation according to Claim 1 or 2, wherein said formulation is stable at about 2-8°C for at least one year.
11. The stable liquid pharmaceutical formulation according to Claim 1 or 2, wherein said formulation is stable at about 23-27°C for at least 6 months.
12. A liquid pharmaceutical formulation comprising: about 20-60 mM succinate buffer having pH of from about pH 5.5 to about pH 6.5, about 0.01 % - 0.05 % polysorbate, about 75-150 mM sodium chloride, and greater than 50 mg/ml antibody selected from the group consisting of Daclizumab, Flintozumab, HAEL-12, and HuEP5C7.
13. A liquid pharmaceutical formulation comprising: about 30-70 mM histidine buffer having pH of from about pH 5.5 to about pH 6.5, about 0.01 % - 0.05 % polysorbate, about 75-150 mM sodium chloride, and greater than 50 mg/ml antibody selected from the group consisting of Daclizumab, Flintozumab, HAEL-12, and HuEP5C7.
PCT/US2002/036093 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of igg antibodies WO2003039485A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
AT02802895T ATE556591T1 (en) 2001-11-08 2002-11-08 STABLE PHARMACEUTICAL LIQUID FORMULATION OF IGG ANTIBODIES
IL16167702A IL161677A0 (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of igg antibodies
ES02802895T ES2392073T3 (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of IGG antibodies
CA2466034A CA2466034C (en) 2001-11-08 2002-11-08 Stable aqueous pharmaceutical formulations of daclizumab antibodies
EP02802895A EP1441589B1 (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of igg antibodies
JP2003541777A JP5290489B2 (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of IGG antibody
DK02802895.9T DK1441589T3 (en) 2001-11-08 2002-11-08 Stable, liquid, pharmaceutical composition of IgG antibodies
NZ532896A NZ532896A (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of IGG antibodies including daclizumab and fontolizumab
AU2002363339A AU2002363339B2 (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of IGG antibodies
IL161677A IL161677A (en) 2001-11-08 2004-04-29 Stable liquid pharmaceutical formuation comprising succinate buffer, polysorbate, sodium chloride and a daclizumab antibody
HK05107139A HK1074750A1 (en) 2001-11-08 2005-08-17 Stable liquid pharmaceutical formulation of igg antiboddies
LU93314C LU93314I2 (en) 2001-11-08 2016-11-17
CY2016044C CY2016044I2 (en) 2001-11-08 2016-12-09 STABLE PHARMACEUTICAL PREPARATION IN LIQUID FORM OF IGG ANTIBODIES

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33750901P 2001-11-08 2001-11-08
US60/337,509 2001-11-08

Publications (2)

Publication Number Publication Date
WO2003039485A2 true WO2003039485A2 (en) 2003-05-15
WO2003039485A3 WO2003039485A3 (en) 2004-02-12

Family

ID=23320826

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/036093 WO2003039485A2 (en) 2001-11-08 2002-11-08 Stable liquid pharmaceutical formulation of igg antibodies

Country Status (17)

Country Link
US (3) US20030138417A1 (en)
EP (1) EP1441589B1 (en)
JP (2) JP5290489B2 (en)
KR (1) KR100913714B1 (en)
CN (1) CN1292655C (en)
AT (1) ATE556591T1 (en)
AU (1) AU2002363339B2 (en)
CA (1) CA2466034C (en)
CY (1) CY2016044I2 (en)
DK (1) DK1441589T3 (en)
ES (1) ES2392073T3 (en)
HK (1) HK1074750A1 (en)
IL (2) IL161677A0 (en)
LU (1) LU93314I2 (en)
NZ (1) NZ532896A (en)
PT (1) PT1441589E (en)
WO (1) WO2003039485A2 (en)

Cited By (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004001007A2 (en) * 2002-06-21 2003-12-31 Idec Pharmaceuticals Corporation Buffered formulations for concentrating antibodies and methods of use thereof
WO2006081587A2 (en) * 2005-01-28 2006-08-03 Wyeth Stabilized liquid polypeptide formulations
WO2006083689A2 (en) * 2005-01-28 2006-08-10 Elan Pharma International Limited Anti a beta antibody formulation
WO2006096490A2 (en) * 2005-03-08 2006-09-14 Pharmacia & Upjohn Company Llc ANTI-MAdCAM ANTIBODY COMPOSITIONS
WO2007037607A1 (en) * 2005-09-27 2007-04-05 Lg Life Sciences, Ltd. Hfsh aqueous formulation
WO2007127665A2 (en) * 2006-04-26 2007-11-08 Wyeth Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
WO2007087344A3 (en) * 2006-01-25 2007-12-13 Taro Pharmaceuticals Usa Inc Anti-histamine compositions and use thereof
EP1909838A2 (en) * 2005-07-29 2008-04-16 Amgen Inc. Formulations that inhibit protein aggregation
WO2008045373A2 (en) * 2006-10-06 2008-04-17 Amgen Inc. Stable antibody formulations
WO2008051363A2 (en) * 2006-10-20 2008-05-02 Amgen Inc. Stable polypeptide formulations
EP1917030A2 (en) * 2005-08-03 2008-05-07 Immunogen, Inc. Immunoconjugate formulations
JP2008520551A (en) * 2004-10-20 2008-06-19 ジェネンテック・インコーポレーテッド Antibody preparation
WO2009041621A1 (en) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anti-il-6 receptor antibody
WO2009041613A1 (en) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
EP2196476A1 (en) * 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
WO2010089522A1 (en) * 2009-07-16 2010-08-12 Arecor Limited Stable formulation of a therapeutic protein
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
JP2011184446A (en) * 2003-12-22 2011-09-22 Pfizer Products Inc Cd40 antibody formulation and method
US8034339B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
FR2961107A1 (en) * 2010-06-15 2011-12-16 Lab Francais Du Fractionnement HUMAN IMMUNOGLOBULIN COMPOSITION STABILIZED
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
WO2012037534A1 (en) * 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral ph
CN103217525A (en) * 2013-03-21 2013-07-24 上海执诚生物科技股份有限公司 Composition for improving cystatin C latex coated antibody stability, stabilizer containing the same, preparation method and application thereof
EP1610820B2 (en) 2003-04-04 2013-08-21 Genentech, Inc. High concentration antibody and protein formulations
WO2013165791A1 (en) * 2012-05-03 2013-11-07 Boehringer Ingelheim International Gmbh Anti-il-23p19 antibodies
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
AU2012200284B2 (en) * 2006-10-06 2014-03-06 Amgen Inc. Stable Antibody Formulations
US8697397B2 (en) 2007-08-07 2014-04-15 Chugai Seiyaku Kabushiki Kaisha Method of producing heterogeneous protein
EP2727602A1 (en) 2012-10-31 2014-05-07 Takeda GmbH Method for preparation of a high concentration liquid formulation of an antibody
US8741601B2 (en) 2009-04-22 2014-06-03 Chugai Seiyaku Kabushiki Kaisha Method for producing a cell capable of high-yield production of heteroproteins
AU2012200203B2 (en) * 2005-03-08 2014-07-03 Pfizer Products Inc. Anti-CTLA-4 Antibody Compositions
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8796210B2 (en) 2008-07-16 2014-08-05 Arecor Limited Stable formulation of growth hormone comprising lactate anion
US8796007B2 (en) 2007-03-15 2014-08-05 Chugai Seiyaku Kabushiki Kaisha Method for production of polypeptide
WO2014141152A3 (en) * 2013-03-15 2014-12-04 Glaxosmithkline Intellectual Property (No.2) Limited Low concentration antibody formulations
AU2014268186A1 (en) * 2006-04-26 2014-12-11 Wyeth Llc Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US8945564B2 (en) 2006-04-21 2015-02-03 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
WO2015075201A1 (en) 2013-11-21 2015-05-28 Genmab A/S Antibody-drug conjugate lyophilised formulation
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US9068212B2 (en) 2007-10-24 2015-06-30 Chugai Seiyaku Kabushiki Kaisha Method for producing a polypeptide using a cell that overexpresses a bicarbonate transporter
US9095567B2 (en) 2010-06-04 2015-08-04 Wyeth Llc Vaccine formulations
WO2015173782A1 (en) * 2014-05-16 2015-11-19 Glaxosmithkline Intellectual Property Management Limited Antibody formulation
US9238687B2 (en) 2006-04-13 2016-01-19 Chugai Seiyaku Kabushiki Kaisha Method for recombinant production of a desired polypeptide using a mammalian cell co-expressing a taurine transporter polypeptide
US9328169B2 (en) 2004-01-09 2016-05-03 Pfizer Inc. Human antibodies that bind human MAdCAM
US9441036B2 (en) 2010-11-04 2016-09-13 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
AU2014240252B2 (en) * 2005-03-08 2016-10-06 Pfizer Products Inc Anti-CTLA-4 Antibody Compositions
US9487589B2 (en) 2011-06-30 2016-11-08 Genentech, Inc. Anti-c-met-antibody formulations
WO2016205037A1 (en) * 2015-06-17 2016-12-22 Eli Lilly And Company Anti-cgrp antibody formulation
EP3127921A1 (en) 2007-09-26 2017-02-08 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substition in cdr
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
WO2017121867A1 (en) 2016-01-13 2017-07-20 Genmab A/S Formulation for antibody and drug conjugate thereof
US9732152B2 (en) 2002-08-16 2017-08-15 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9802993B2 (en) 2007-10-15 2017-10-31 Chugai Seiyaku Kabushiki Kaisha Method for producing a cell for protein production by treating a cell overexpressing a taurine transporter with methotrexate
US9849181B2 (en) 2012-08-31 2017-12-26 Bayer Healthcare Llc High concentration antibody and protein formulations
WO2018027195A1 (en) * 2016-08-05 2018-02-08 Abbvie Biotherapeutics Inc. Compositions containing reduced amounts of daclizumab acidic isoforms and methods for preparing the same
EP2526963B1 (en) 2010-01-20 2018-04-25 Chugai Seiyaku Kabushiki Kaisha Stabilized Antibody-Containing Liquid Formulations
EP2697369B1 (en) 2011-03-25 2018-06-27 F.Hoffmann-La Roche Ag Novel protein purification methods
US10059763B2 (en) 2014-09-03 2018-08-28 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
USRE47150E1 (en) 2010-03-01 2018-12-04 Bayer Healthcare Llc Optimized monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
US10413593B2 (en) 2014-10-24 2019-09-17 Merck Sharp & Dohme Corp. Co-agonists of the glucagon and GLP-1 receptors
AU2018204779B2 (en) * 2006-04-26 2019-10-31 Wyeth Llc Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
CN110418803A (en) * 2017-03-01 2019-11-05 免疫医疗有限公司 Anti- RSV monoclonal antibody preparation
US10507241B2 (en) 2014-07-24 2019-12-17 Boehringer Ingelheim International Gmbh Biomarkers useful in the treatment of IL-23A related diseases
US10745475B2 (en) 2013-08-30 2020-08-18 Takeda Gmbh Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
US11034760B2 (en) 2015-05-07 2021-06-15 Swedish Orphen Biovitrum AG Methods and compositions for diagnosis and treatment of disorders in patients with elevated levels of CXCL9 and other biomarkers
WO2021146336A1 (en) * 2020-01-13 2021-07-22 Aptevo Research And Development Llc Methods and compositions for preventing adsorption of therapeutic proteins to drug delivery system components
US11091543B2 (en) 2015-05-07 2021-08-17 Swedish Orphan Biovitrum Ag Methods, compositions and dosing regimens for treating or preventing interferon-gamma related indications
WO2021233853A1 (en) * 2020-05-19 2021-11-25 F. Hoffmann-La Roche Ag The use of chelators for the prevention of visible particle formation in parenteral protein solutions
US11633476B2 (en) 2017-05-02 2023-04-25 Merck Sharp & Dohme Llc Stable formulations of programmed death receptor 1 (PD-1) antibodies and methods of use thereof
US11801300B2 (en) 2014-12-22 2023-10-31 Novartis Ag Pharmaceutical products and stable liquid compositions of IL-17 antibodies
US11802156B2 (en) 2017-07-14 2023-10-31 Pfizer Inc. Antibodies to MAdCAM
US11845798B2 (en) 2017-05-02 2023-12-19 Merck Sharp & Dohme Llc Formulations of anti-LAG3 antibodies and co-formulations of anti-LAG3 antibodies and anti-PD-1 antibodies

Families Citing this family (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8703126B2 (en) 2000-10-12 2014-04-22 Genentech, Inc. Reduced-viscosity concentrated protein formulations
AU1344102A (en) * 2000-10-12 2002-04-22 Genentech Inc Reduced-viscosity concentrated protein formulations
PT1441589E (en) * 2001-11-08 2012-08-13 Abbott Biotherapeutics Corp Stable liquid pharmaceutical formulation of igg antibodies
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
US20040002451A1 (en) * 2002-06-20 2004-01-01 Bruce Kerwin Compositions of pegylated soluble tumor necrosis factor receptors and methods of preparing
MY150740A (en) * 2002-10-24 2014-02-28 Abbvie Biotechnology Ltd Low dose methods for treating disorders in which tnf? activity is detrimental
AU2003293543A1 (en) * 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
US20040208869A1 (en) * 2003-01-30 2004-10-21 Medimmune, Inc. Uses of anti-integrin alphanubeta3 antibody formulations
US20040208870A1 (en) * 2003-01-30 2004-10-21 Medimmune, Inc. Stabilized high concentration anti-integrin alphanubeta3 antibody formulations
WO2004075913A1 (en) 2003-02-28 2004-09-10 Chugai Seiyaku Kabushiki Kaisha Stabilized preparation containing protein
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
DK1691833T3 (en) * 2003-11-28 2010-05-03 Micromet Ag Compositions comprising polypeptides
US20060008415A1 (en) * 2004-06-25 2006-01-12 Protein Design Labs, Inc. Stable liquid and lyophilized formulation of proteins
LT2586459T (en) 2005-03-25 2017-09-25 Regeneron Pharmaceuticals, Inc. Vegf antagonist formulations
MX2007014148A (en) * 2005-05-19 2008-01-11 Amgen Inc Compositions and methods for increasing the stability of antibodies.
ES2776657T3 (en) 2005-06-14 2020-07-31 Amgen Inc Self-buffering protein formulations
KR20080025174A (en) * 2005-06-23 2008-03-19 메디뮨 인코포레이티드 Antibody formulations having optimized aggregation and fragmentation profiles
WO2007033230A2 (en) * 2005-09-12 2007-03-22 Novimmune S.A. Anti-cd3 antibody formulations
CA2625998C (en) 2005-10-06 2015-12-01 Xencor, Inc. Optimized anti-cd30 antibodies
JP5405122B2 (en) * 2005-12-21 2014-02-05 ワイス・エルエルシー Low viscosity protein formulations and uses thereof
AR059066A1 (en) 2006-01-27 2008-03-12 Amgen Inc COMBINATIONS OF THE ANGIOPOYETINE INHIBITOR -2 (ANG2) AND THE VASCULAR ENDOTELIAL GROWTH FACTOR INHIBITOR (VEGF)
NZ611859A (en) 2006-04-05 2014-12-24 Abbvie Biotechnology Ltd Antibody purification
LT2944306T (en) 2006-06-16 2021-02-25 Regeneron Pharmaceuticals, Inc. Vegf antagonist formulations suitable for intravitreal administration
CA2790018C (en) * 2006-12-21 2015-02-03 Amgen Inc. Formulations
WO2009009406A1 (en) * 2007-07-06 2009-01-15 Smithkline Beecham Corporation Antibody formulations
WO2009064838A1 (en) * 2007-11-15 2009-05-22 Amgen, Inc. Aqueous formulation of erythropoiesis stimulating protein stablised by antioxidants for parenteral administration
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
PE20091174A1 (en) 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd LIQUID FORMULATION WITH HIGH CONCENTRATION OF ANTIBODY CONTENT
CA2710775A1 (en) * 2007-12-28 2009-07-09 Bioinvent International Ab Formulation
EP2328559B1 (en) * 2008-09-19 2015-01-07 F. Hoffmann-La Roche AG Formulation comprising antibody against p-selectin
WO2010062896A1 (en) * 2008-11-28 2010-06-03 Abbott Laboratories Stable antibody compositions and methods for stabilizing same
WO2011026948A1 (en) * 2009-09-03 2011-03-10 Ablynx N.V. Stable formulations of polypeptides and uses thereof
US10005830B2 (en) 2009-03-05 2018-06-26 Ablynx N.V. Antigen binding dimer-complexes, methods of making/avoiding and uses thereof
US9265834B2 (en) 2009-03-05 2016-02-23 Ablynx N.V. Stable formulations of polypeptides and uses thereof
EP2403874A1 (en) * 2009-03-06 2012-01-11 Genentech, Inc. Antibody formulation
JP2012520098A (en) * 2009-03-30 2012-09-06 エフ.ホフマン−ラ ロシュ アーゲー How to prevent fogging of glass
RU2560701C2 (en) * 2009-05-04 2015-08-20 Эббви Байотекнолоджи Лтд. Stable compositions with high concentrations of proteins of human antibodies against tnf-alpha
CA2765220A1 (en) * 2009-07-14 2011-01-20 Biogen Idec Ma Inc. Methods for inhibiting yellow color and peroxide formation in a composition
EP2473851A1 (en) 2009-08-31 2012-07-11 Abbott Biotherapeutics Corp. Use of an immunoregulatory nk cell population for monitoring the efficacy of anti-il-2r antibodies in multiple sclerosis patients
US8821879B2 (en) 2009-09-04 2014-09-02 Xoma Technology Ltd. Anti-botulism antibody coformulations
EP2473191B1 (en) * 2009-09-04 2017-08-23 XOMA Technology Ltd. Antibody coformulations
BR112012011463A2 (en) 2009-10-30 2017-05-02 Abbott Biotherapeutics Corp use of nk cell immunoregulatory populations to predict the efficacy of anti-il-2r antibodies in multiple sclerosis patients.
US20110158987A1 (en) * 2009-12-29 2011-06-30 F. Hoffmann-Laroche Ag Novel antibody formulation
SI2521568T1 (en) 2010-01-06 2019-01-31 Dyax Corp. Plasma kallikrein binding proteins
US20120294866A1 (en) * 2010-01-19 2012-11-22 F. Hoffmann-La Roche Ag Pharmaceutical formulation for proteins
WO2011109365A2 (en) 2010-03-01 2011-09-09 Progenics Pharmaceuticals, Inc. Concentrated protein formulations and uses thereof
JO3400B1 (en) 2010-09-30 2019-10-20 Ferring Bv Pharmaceutical composition of carbetocin
CA2815689C (en) 2010-11-11 2016-11-22 Abbvie Biotechnology Ltd. Improved high concentration anti-tnf.alpha. antibody liquid formulations
EP2471554A1 (en) * 2010-12-28 2012-07-04 Hexal AG Pharmaceutical formulation comprising a biopharmaceutical drug
CN103635489B (en) 2011-01-06 2016-04-13 戴埃克斯有限公司 Blood plasma prekallikrein associated proteins
KR20200077622A (en) 2011-01-13 2020-06-30 리제너론 파아마슈티컬스, 인크. Use of a vegf antagonist to treat angiogenic eye disorders
UY34105A (en) 2011-06-03 2012-07-31 Lg Life Sciences Ltd STABLE LIQUID FORMULATION OF ETANERCEPT
MX354988B (en) 2011-10-25 2018-03-28 Prothena Biosciences Ltd Antibody formulations and methods.
CA2864539C (en) * 2012-02-16 2022-06-07 Santarus, Inc. Antibody formulations
BR112014028600B1 (en) * 2012-05-18 2022-11-22 Genentech, Inc SUSPENSION FORMULATIONS COMPRISING HIGH CONCENTRATION MONOCLONAL ANTIBODY, ITS METHOD OF PREPARATION, ITS USE AND DEVICE FOR SUBCUTANEOUS ADMINISTRATION, AND METHOD FOR PRODUCING AN ARTICLE OF MANUFACTURING
US9216219B2 (en) 2012-06-12 2015-12-22 Novartis Ag Anti-BAFFR antibody formulation
BR112015000203A2 (en) * 2012-07-09 2017-06-27 Coherus Biosciences Inc etanercept formulations exhibiting marked reduction in sub-visible particles
FR2994390B1 (en) 2012-08-10 2014-08-15 Adocia METHOD FOR LOWERING THE VISCOSITY OF HIGH CONCENTRATION PROTEIN SOLUTIONS
US8613919B1 (en) 2012-08-31 2013-12-24 Bayer Healthcare, Llc High concentration antibody and protein formulations
KR102238677B1 (en) * 2012-09-07 2021-04-12 코히러스 바이오사이언시즈, 인코포레이티드 Stable aqueous formulations of adalimumab
UA117466C2 (en) * 2012-12-13 2018-08-10 Мерк Шарп Енд Доме Корп. SOLUTION FORMULATIONS OF ENGINEERED ANTI-IL-23p19 ANTIBODIES
JP2016515524A (en) 2013-03-15 2016-05-30 アッヴィ バイオテクノロジー リミテッド Anti-CD25 antibodies and their use
CN105051068A (en) 2013-03-15 2015-11-11 戴埃克斯有限公司 Anti-plasma kallikrein antibodies
RU2015144033A (en) 2013-03-15 2017-04-26 Эббви Байотекнолоджи Лтд. ANTIBODIES AGAINST CD25 AND THEIR APPLICATION
KR20140119396A (en) 2013-03-29 2014-10-10 삼성전자주식회사 Liquid formulation containing a protein drug
CN116585468A (en) 2014-01-21 2023-08-15 武田药品工业株式会社 Plasma kallikrein binding proteins and their use in the treatment of hereditary angioedema
CN106459210A (en) 2014-03-27 2017-02-22 戴埃克斯有限公司 Compositions and methods for treatment of diabetic macular edema
WO2015169742A1 (en) * 2014-05-07 2015-11-12 Takeda Gmbh Liquid formulation comprising gm-csf neutralizing compound
US20160074515A1 (en) 2014-06-20 2016-03-17 Reform Biologics, Llc Viscosity-reducing excipient compounds for protein formulations
US11357857B2 (en) 2014-06-20 2022-06-14 Comera Life Sciences, Inc. Excipient compounds for protein processing
US10478498B2 (en) 2014-06-20 2019-11-19 Reform Biologics, Llc Excipient compounds for biopolymer formulations
US20170204149A1 (en) 2014-06-23 2017-07-20 Novartis Ag Hsa-gdf-15 fusion polypeptide and use thereof
US10588980B2 (en) 2014-06-23 2020-03-17 Novartis Ag Fatty acids and their use in conjugation to biomolecules
WO2015200080A1 (en) 2014-06-23 2015-12-30 Novartis Ag Site specific protein modifications
ES2756275T3 (en) 2014-11-07 2020-04-27 Sesen Bio Inc Enhanced anti-IL-6 antibodies
MX2017009534A (en) 2015-01-23 2018-04-10 Novartis Ag Synthetic apelin fatty acid conjugates with improved half-life.
SG10202101105XA (en) 2015-08-13 2021-03-30 Amgen Inc Charged depth filtration of antigen-binding proteins
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
FI3384049T3 (en) 2015-12-03 2023-09-25 Regeneron Pharma Methods of associating genetic variants with a clinical outcome in patients suffering from age-related macular degeneration treated with anti-vegf
WO2017096262A1 (en) 2015-12-04 2017-06-08 Jomoco, Corp. Compositions and methods to mitigate or prevent an immune response to an immunogenic therapeutic molecule in non-human primates
EA201891388A1 (en) 2015-12-11 2018-11-30 Дайэкс Корп. PLASMA KALLIKREIN INHIBITORS AND THEIR APPLICATION FOR THE TREATMENT OF THE EXPOSURE OF HEREDITARY ANGIONEUROTIC DOMESTIC
US20190000923A1 (en) 2015-12-22 2019-01-03 Novartis Ag Methods of treating or ameliorating metabolic disorders using growth differentiation factor 15 (gdf-15)
WO2017147293A1 (en) * 2016-02-23 2017-08-31 Eleven Biotherapeutics, Inc. Il-6 antagonist formulations and uses thereof
WO2017184880A1 (en) 2016-04-20 2017-10-26 Coherus Biosciences, Inc. A method of filling a container with no headspace
MX2019004580A (en) 2016-10-21 2019-08-12 Amgen Inc Pharmaceutical formulations and methods of making the same.
WO2018080196A2 (en) 2016-10-28 2018-05-03 (주)셀트리온 Stable pharmaceutical formulation
WO2018156180A1 (en) 2017-02-24 2018-08-30 Kindred Biosciences, Inc. Anti-il31 antibodies for veterinary use
GB201703062D0 (en) * 2017-02-24 2017-04-12 Arecor Ltd Stabilized antibody protein solutions
US11608357B2 (en) 2018-08-28 2023-03-21 Arecor Limited Stabilized antibody protein solutions
WO2018156367A1 (en) * 2017-02-24 2018-08-30 Kindred Biosciences, Inc. Anti-il31 antibodies for veterinary use
EP3372241A1 (en) 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
EP3372242A1 (en) 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
JP7179717B2 (en) * 2017-03-31 2022-11-29 Meiji Seikaファルマ株式会社 Aqueous formulation, aqueous formulation containing syringe, antibody protein disaggregation agent, and antibody protein disaggregation method
EP3612217A4 (en) * 2017-04-18 2020-12-30 Dr. Reddy's Laboratories Limited Stable liquid pharmaceutical composition
RU2766590C2 (en) * 2017-05-16 2022-03-15 Цзянсу Хэнжуй Медисин Ко., Лтд. Pharmaceutical composition based on antibody to pd-l1 and its application
CA3074565A1 (en) 2017-09-05 2019-03-14 Merck Sharp & Dohme Corp. Compounds for reducing the viscosity of biological formulations
WO2019055902A1 (en) * 2017-09-18 2019-03-21 Amgen Inc. Vegfr-fc fusion protein formulations
TW202344266A (en) 2017-12-22 2023-11-16 瑞士商諾華公司 Methods of treating metabolic disorders with fgf21 variants
JP7235770B2 (en) 2018-05-10 2023-03-08 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Formulations Containing High Concentrations of VEGF Receptor Fusion Proteins
US11519020B2 (en) 2018-05-25 2022-12-06 Regeneron Pharmaceuticals, Inc. Methods of associating genetic variants with a clinical outcome in patients suffering from age-related macular degeneration treated with anti-VEGF
JP2022513626A (en) 2018-11-26 2022-02-09 ノバルティス アーゲー LPL-GPIHBP1 fusion polypeptide
US20230287122A1 (en) * 2018-11-29 2023-09-14 Harbour Biomed Therapeutics Limited Anti-pd-l1 antibody preparation
EP3914282A1 (en) 2019-01-25 2021-12-01 Ospedale San Raffaele S.r.l. Inhibitor of dux4 and uses thereof
WO2021011404A1 (en) * 2019-07-12 2021-01-21 Contrafect Corporation Therapeutic protein formulations comprising antibodies and uses thereof
GB201913697D0 (en) 2019-09-23 2019-11-06 King S College London DAP10/DAP12 fusion polypeptides
GB202003277D0 (en) 2020-03-06 2020-04-22 King S College London Therapeutic agents
GB202007655D0 (en) 2020-05-22 2020-07-08 King S College London Chimeric nkg2d protein
EP4171622B1 (en) 2021-03-23 2024-05-01 King's College London Compositions comprising nkg2d, cxcr2, and dap10/dap12 fusion polypeptides and methods of use thereof
GB202214120D0 (en) 2022-09-27 2022-11-09 King S College London Compositions comprising NKG2D, CXCR2, and DAP10/DAP12 fusion polypeptides and methods of use thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997045140A1 (en) * 1996-05-24 1997-12-04 Glaxo Group Limited Concentrated antibody preparation
WO1998022136A2 (en) * 1996-11-19 1998-05-28 Roche Diagnostics Gmbh Stable lyophilized pharmaceutical substances from monoclonal or polyclonal antibodies
WO1998056418A1 (en) * 1997-06-13 1998-12-17 Genentech, Inc. Stabilized antibody formulation
US6267958B1 (en) * 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO2002030463A2 (en) * 2000-10-12 2002-04-18 Genentech, Inc. Reduced-viscosity concentrated protein formulations

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5091178A (en) * 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
DE68908175T2 (en) 1988-05-27 1994-03-03 Centocor Inc FREEZE DRIED FORMULATION FOR ANTIBODY PRODUCTS.
WO1990006764A1 (en) * 1988-12-15 1990-06-28 Invitron Corporation Use of basic amino acids to solubilize immunoglobulins
EP0465513A1 (en) 1989-03-27 1992-01-15 Centocor, Inc. FORMULATIONS FOR STABILIZING OF IgM ANTIBODIES
US6165467A (en) * 1991-07-20 2000-12-26 Yoshihide Hagiwara Stabilized human monoclonal antibody preparation
JP2000510813A (en) * 1995-02-06 2000-08-22 ジェネテイックス・インスティテュート・インコーポレイテッド Formulation for IL-12
JPH11510170A (en) * 1995-07-27 1999-09-07 ジェネンテック インコーポレーテッド Protein Formula
US6685940B2 (en) * 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
US6171586B1 (en) * 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
KR101222450B1 (en) 1999-03-25 2013-01-16 애보트 게엠베하 운트 콤파니 카게 Human antibodies that bind human IL-12 and methods for producing
US6914128B1 (en) * 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
GB0113179D0 (en) * 2001-05-31 2001-07-25 Novartis Ag Organic compounds
PT1441589E (en) * 2001-11-08 2012-08-13 Abbott Biotherapeutics Corp Stable liquid pharmaceutical formulation of igg antibodies

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6267958B1 (en) * 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO1997045140A1 (en) * 1996-05-24 1997-12-04 Glaxo Group Limited Concentrated antibody preparation
WO1998022136A2 (en) * 1996-11-19 1998-05-28 Roche Diagnostics Gmbh Stable lyophilized pharmaceutical substances from monoclonal or polyclonal antibodies
WO1998056418A1 (en) * 1997-06-13 1998-12-17 Genentech, Inc. Stabilized antibody formulation
WO2002030463A2 (en) * 2000-10-12 2002-04-18 Genentech, Inc. Reduced-viscosity concentrated protein formulations

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1441589A2 *

Cited By (185)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8642044B2 (en) 1997-12-02 2014-02-04 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US8034339B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8034348B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US8535673B2 (en) 1997-12-02 2013-09-17 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
WO2004001007A3 (en) * 2002-06-21 2004-07-01 Idec Pharma Corp Buffered formulations for concentrating antibodies and methods of use thereof
WO2004001007A2 (en) * 2002-06-21 2003-12-31 Idec Pharmaceuticals Corporation Buffered formulations for concentrating antibodies and methods of use thereof
US9750808B2 (en) 2002-08-16 2017-09-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US9732152B2 (en) 2002-08-16 2017-08-15 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9738714B2 (en) 2002-08-16 2017-08-22 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9950066B2 (en) 2002-08-16 2018-04-24 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8961964B2 (en) 2003-04-04 2015-02-24 Genentech, Inc. High concentration antibody and protein formulations
EP1610820B2 (en) 2003-04-04 2013-08-21 Genentech, Inc. High concentration antibody and protein formulations
US10034940B2 (en) 2003-04-04 2018-07-31 Genentech, Inc. High concentration antibody and protein formulations
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
JP2014205692A (en) * 2003-12-22 2014-10-30 ファイザー・プロダクツ・インク CD40 antibody formulation and methods
JP2011184446A (en) * 2003-12-22 2011-09-22 Pfizer Products Inc Cd40 antibody formulation and method
US9328169B2 (en) 2004-01-09 2016-05-03 Pfizer Inc. Human antibodies that bind human MAdCAM
US10259872B2 (en) 2004-01-09 2019-04-16 Pfizer, Inc. Antibodies to MAdCAM
EP3498294A1 (en) 2004-10-20 2019-06-19 Genentech, Inc. Antibody formulations
US8372396B2 (en) 2004-10-20 2013-02-12 Genetech, Inc. Antibody formulations
EP2371388A2 (en) 2004-10-20 2011-10-05 Genentech, Inc. Antibody formulations
JP2014159469A (en) * 2004-10-20 2014-09-04 Genentech Inc Antibody formulations
JP2008520551A (en) * 2004-10-20 2008-06-19 ジェネンテック・インコーポレーテッド Antibody preparation
JP2012176970A (en) * 2004-10-20 2012-09-13 Genentech Inc Antibody formulation
US9017671B2 (en) 2004-10-20 2015-04-28 Genentech, Inc. Method of treating cancer with a pharmaceutical formulation comprising a HER2 antibody
JP2016065091A (en) * 2004-10-20 2016-04-28 ジェネンテック, インコーポレイテッド Antibody formulations
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
WO2006083689A3 (en) * 2005-01-28 2006-10-19 Neuralab Ltd Anti a beta antibody formulation
JP2008528612A (en) * 2005-01-28 2008-07-31 エラン・フアルマ・インターナシヨナル・リミテツド Anti-Abeta antibody preparation
US8318164B2 (en) 2005-01-28 2012-11-27 Janssen Alzheimer Immunotherapy Anti A beta antibody formulation
EA021507B1 (en) * 2005-01-28 2015-07-30 Янссен Сайенсиз Айрлэнд Юси Anti a beta antibody formulation
EP2392353A1 (en) * 2005-01-28 2011-12-07 Janssen Alzheimer Immunotherapy Anti A beta antibody formulation
WO2006083689A2 (en) * 2005-01-28 2006-08-10 Elan Pharma International Limited Anti a beta antibody formulation
AU2006211184B2 (en) * 2005-01-28 2011-06-16 Janssen Alzheimer Immunotherapy Anti a beta antibody formulation
WO2006081587A2 (en) * 2005-01-28 2006-08-03 Wyeth Stabilized liquid polypeptide formulations
EA015147B1 (en) * 2005-01-28 2011-06-30 Элан Фарма Интернэшнл Лимитед Stabilized antibody formulation to beta-amyloid peptide and use thereof
AU2006211184B8 (en) * 2005-01-28 2011-07-07 Janssen Alzheimer Immunotherapy Anti a beta antibody formulation
WO2006081587A3 (en) * 2005-01-28 2006-10-12 Wyeth Corp Stabilized liquid polypeptide formulations
US9487581B2 (en) 2005-03-08 2016-11-08 Pfizer Inc. Anti-CTLA-4 antibody compositions
AU2012200203B2 (en) * 2005-03-08 2014-07-03 Pfizer Products Inc. Anti-CTLA-4 Antibody Compositions
AU2014240252B2 (en) * 2005-03-08 2016-10-06 Pfizer Products Inc Anti-CTLA-4 Antibody Compositions
EP2620450A3 (en) * 2005-03-08 2014-01-08 Pfizer Products Inc. Anti-CTLA-4 antibody compositions
AU2006220829B2 (en) * 2005-03-08 2011-10-13 Pfizer Products Inc. Anti-CTLA-4 antibody compositions
WO2006096490A3 (en) * 2005-03-08 2006-12-28 Pharmacia & Upjohn Co Llc ANTI-MAdCAM ANTIBODY COMPOSITIONS
EP2311491A1 (en) * 2005-03-08 2011-04-20 Pharmacia & Upjohn Company LLC Composition comprising an antibody against macrophage colony-stimulating factor (M-CSF) and a chelating agent
WO2006096491A3 (en) * 2005-03-08 2007-03-29 Pharmacia & Upjohn Co Llc Anti-ctla-4 antibody compositions
JP2015110656A (en) * 2005-03-08 2015-06-18 ファイザー・プロダクツ・インク Anti-ctla-4 antibody composition
JP2006249081A (en) * 2005-03-08 2006-09-21 Pharmacia & Upjohn Co Llc Anti-ctla-4 antibody composition
JP2006249084A (en) * 2005-03-08 2006-09-21 Pharmacia & Upjohn Co Llc ANTI-MadCAM ANTIBODY COMPOSITION
JP2006249085A (en) * 2005-03-08 2006-09-21 Pharmacia & Upjohn Co Llc Platform antibody composition
WO2006096461A3 (en) * 2005-03-08 2006-12-21 Pharmacia & Upjohn Co Llc Composition comprising an antibody against macrophage colony-stimulating factor (m-csf) and a chelating agent
JP2012167120A (en) * 2005-03-08 2012-09-06 Pharmacia & Upjohn Co Llc Anti-ctla-4 antibody composition
WO2006096491A2 (en) * 2005-03-08 2006-09-14 Pharmacia & Upjohn Company Llc Anti-ctla-4 antibody compositions
WO2006096490A2 (en) * 2005-03-08 2006-09-14 Pharmacia & Upjohn Company Llc ANTI-MAdCAM ANTIBODY COMPOSITIONS
EP1909838A2 (en) * 2005-07-29 2008-04-16 Amgen Inc. Formulations that inhibit protein aggregation
US9114179B2 (en) 2005-08-03 2015-08-25 Immunogen, Inc. Immunoconjugate formulations
EP1917030A2 (en) * 2005-08-03 2008-05-07 Immunogen, Inc. Immunoconjugate formulations
EP1917030A4 (en) * 2005-08-03 2011-03-09 Immunogen Inc Immunoconjugate formulations
WO2007037607A1 (en) * 2005-09-27 2007-04-05 Lg Life Sciences, Ltd. Hfsh aqueous formulation
WO2007087344A3 (en) * 2006-01-25 2007-12-13 Taro Pharmaceuticals Usa Inc Anti-histamine compositions and use thereof
US8569278B2 (en) 2006-01-25 2013-10-29 Taro Pharmaceuticals North America, Inc. Anti-histamine compositions and use thereof
US9238687B2 (en) 2006-04-13 2016-01-19 Chugai Seiyaku Kabushiki Kaisha Method for recombinant production of a desired polypeptide using a mammalian cell co-expressing a taurine transporter polypeptide
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8945564B2 (en) 2006-04-21 2015-02-03 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
US8562999B2 (en) 2006-04-26 2013-10-22 Wyeth Llc Formulations which stabilize and inhibit precipitation of immunogenic compositions
AU2014268186A1 (en) * 2006-04-26 2014-12-11 Wyeth Llc Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
WO2007127665A2 (en) * 2006-04-26 2007-11-08 Wyeth Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
US7935787B2 (en) 2006-04-26 2011-05-03 Wyeth Llc Formulations which stabilize and inhibit precipitation of immunogenic compositions
WO2007127665A3 (en) * 2006-04-26 2008-11-13 Wyeth Corp Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
EP2679244A3 (en) * 2006-04-26 2014-01-15 Wyeth LLC Formulations which stabilize and inhibit precipitation of immunogenic compositions
AU2018204779B2 (en) * 2006-04-26 2019-10-31 Wyeth Llc Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
WO2008045373A3 (en) * 2006-10-06 2008-10-16 Amgen Inc Stable antibody formulations
WO2008045373A2 (en) * 2006-10-06 2008-04-17 Amgen Inc. Stable antibody formulations
AU2012200284B2 (en) * 2006-10-06 2014-03-06 Amgen Inc. Stable Antibody Formulations
AU2007307107B2 (en) * 2006-10-06 2011-11-03 Amgen Inc. Stable antibody formulations
WO2008051363A3 (en) * 2006-10-20 2008-09-12 Amgen Inc Stable polypeptide formulations
US7705132B2 (en) 2006-10-20 2010-04-27 Amgen Inc. Stable polypeptide formulations
US8241632B2 (en) 2006-10-20 2012-08-14 Amgen Inc. Stable polypeptide formulations
WO2008051363A2 (en) * 2006-10-20 2008-05-02 Amgen Inc. Stable polypeptide formulations
US9181540B2 (en) 2007-03-15 2015-11-10 Chugai Seiyaku Kabushiki Kaisha Method for production of polypeptide
US8796007B2 (en) 2007-03-15 2014-08-05 Chugai Seiyaku Kabushiki Kaisha Method for production of polypeptide
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8697397B2 (en) 2007-08-07 2014-04-15 Chugai Seiyaku Kabushiki Kaisha Method of producing heterogeneous protein
US9499618B2 (en) 2007-08-07 2016-11-22 Chugai Seiyaku Kabushiki Kaisha Method of producing heterogeneous protein
US9284374B2 (en) 2007-08-07 2016-03-15 Chugai Seiyaku Kabushiki Kaisha Method of producing heterogeneous protein
EP3789400A1 (en) 2007-09-26 2021-03-10 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
EP3059246A1 (en) 2007-09-26 2016-08-24 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
EP3415529A1 (en) 2007-09-26 2018-12-19 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
EP3689912A1 (en) 2007-09-26 2020-08-05 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
WO2009041613A1 (en) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
WO2009041621A1 (en) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anti-il-6 receptor antibody
EP3127921A1 (en) 2007-09-26 2017-02-08 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substition in cdr
EP4339294A2 (en) 2007-09-26 2024-03-20 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
US9802993B2 (en) 2007-10-15 2017-10-31 Chugai Seiyaku Kabushiki Kaisha Method for producing a cell for protein production by treating a cell overexpressing a taurine transporter with methotrexate
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
US9068212B2 (en) 2007-10-24 2015-06-30 Chugai Seiyaku Kabushiki Kaisha Method for producing a polypeptide using a cell that overexpresses a bicarbonate transporter
US8796210B2 (en) 2008-07-16 2014-08-05 Arecor Limited Stable formulation of growth hormone comprising lactate anion
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
WO2010066762A1 (en) * 2008-12-10 2010-06-17 Novartis Ag Antibody formulation
EP2196476A1 (en) * 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
RU2745601C2 (en) * 2008-12-10 2021-03-29 Новартис Аг Formulations containing antibodies
EP3072906A1 (en) * 2008-12-10 2016-09-28 Novartis Ag Antibody formulation
AU2009324371B2 (en) * 2008-12-10 2013-10-10 Novartis Ag Antibody formulation
EP3792282A1 (en) * 2008-12-10 2021-03-17 Novartis AG Antibody formulation
US8623367B2 (en) 2008-12-10 2014-01-07 Novartis Ag Antibody formulation
US8741601B2 (en) 2009-04-22 2014-06-03 Chugai Seiyaku Kabushiki Kaisha Method for producing a cell capable of high-yield production of heteroproteins
WO2010089522A1 (en) * 2009-07-16 2010-08-12 Arecor Limited Stable formulation of a therapeutic protein
AU2020200395B2 (en) * 2010-01-20 2021-07-08 Chugai Seiyaku Kabushiki Kaisha Stabilized antibody-containing liquid formulations
US11612562B2 (en) 2010-01-20 2023-03-28 Chugai Seiyaku Kabushiki Kaisha Solution preparation containing stabilized antibody
EP2526963B1 (en) 2010-01-20 2018-04-25 Chugai Seiyaku Kabushiki Kaisha Stabilized Antibody-Containing Liquid Formulations
USRE47150E1 (en) 2010-03-01 2018-12-04 Bayer Healthcare Llc Optimized monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
US9095567B2 (en) 2010-06-04 2015-08-04 Wyeth Llc Vaccine formulations
WO2011157950A1 (en) * 2010-06-15 2011-12-22 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Stabilised human immunoglobulin composition
FR2961107A1 (en) * 2010-06-15 2011-12-16 Lab Francais Du Fractionnement HUMAN IMMUNOGLOBULIN COMPOSITION STABILIZED
EA027353B1 (en) * 2010-09-17 2017-07-31 Баксалта Инкорпорейтид STABILIZATION OF IMMUNOGLOBULINS THROUGH AQUEOUS FORMULATION WITH HISTIDINE AT WEAK ACIDIC TO NEUTRAL pH
AU2011301807C1 (en) * 2010-09-17 2016-02-11 Baxalta GmbH Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral pH
WO2012037534A1 (en) * 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral ph
US8795658B2 (en) 2010-09-17 2014-08-05 Baxter International Inc. Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral pH
US9855331B2 (en) 2010-09-17 2018-01-02 Baxalta Incorporated Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral pH
EA032336B1 (en) * 2010-09-17 2019-05-31 Баксалта Инкорпорейтид Stable aqueous immunoglobulin composition and method of stabilization thereof
AU2011301807B2 (en) * 2010-09-17 2015-07-30 Baxalta GmbH Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral pH
WO2012037530A1 (en) * 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins and other proteins through aqueous formulations with sodium chloride at weak acidic to neutral ph
US9441036B2 (en) 2010-11-04 2016-09-13 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US10202448B2 (en) 2010-11-04 2019-02-12 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
EP2697369B1 (en) 2011-03-25 2018-06-27 F.Hoffmann-La Roche Ag Novel protein purification methods
US10906934B2 (en) 2011-03-25 2021-02-02 Genentech, Inc. Protein purification methods
US9487589B2 (en) 2011-06-30 2016-11-08 Genentech, Inc. Anti-c-met-antibody formulations
EP4039275A1 (en) * 2012-05-03 2022-08-10 Boehringer Ingelheim International GmbH Anti-il-23p19 antibodies
WO2013165791A1 (en) * 2012-05-03 2013-11-07 Boehringer Ingelheim International Gmbh Anti-il-23p19 antibodies
EP3326649A1 (en) * 2012-05-03 2018-05-30 Boehringer Ingelheim International GmbH Anti-il-23p19 antibodies
AU2018200239B2 (en) * 2012-05-03 2019-10-17 Boehringer Ingelheim International Gmbh Anti-IL-23p19 antibodies
US11078265B2 (en) 2012-05-03 2021-08-03 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US9849181B2 (en) 2012-08-31 2017-12-26 Bayer Healthcare Llc High concentration antibody and protein formulations
WO2014068021A1 (en) 2012-10-31 2014-05-08 Takeda Gmbh Method for preparation of a high concentration liquid formulation of an antibody
EP2727602A1 (en) 2012-10-31 2014-05-07 Takeda GmbH Method for preparation of a high concentration liquid formulation of an antibody
WO2014141152A3 (en) * 2013-03-15 2014-12-04 Glaxosmithkline Intellectual Property (No.2) Limited Low concentration antibody formulations
US10537638B2 (en) 2013-03-15 2020-01-21 Glaxosmithkline Intellectual Property (No.2) Limited Low concentration antibody formulations
EP3686217A1 (en) * 2013-03-15 2020-07-29 GlaxoSmithKline Intellectual Property (No.2) Limited Low concentration antibody formulations
CN103217525A (en) * 2013-03-21 2013-07-24 上海执诚生物科技股份有限公司 Composition for improving cystatin C latex coated antibody stability, stabilizer containing the same, preparation method and application thereof
US11795216B2 (en) 2013-08-30 2023-10-24 Takeda Pharmaceutical Company Limited Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
US10745475B2 (en) 2013-08-30 2020-08-18 Takeda Gmbh Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
WO2015075201A1 (en) 2013-11-21 2015-05-28 Genmab A/S Antibody-drug conjugate lyophilised formulation
RU2743681C2 (en) * 2014-05-16 2021-02-24 Глэксосмитклайн Интеллекчуал Проперти Менеджмент Лимитед Antibody composition
EP3715371A1 (en) * 2014-05-16 2020-09-30 GlaxoSmithKline Intellectual Property Management Limited Belimumab formulation
US11179463B2 (en) 2014-05-16 2021-11-23 Glaxosmithkline Intellectual Property Management Limited BLyS antibody formulation
EP3719038A1 (en) * 2014-05-16 2020-10-07 GlaxoSmithKline Intellectual Property Management Limited Belimumab formulation
WO2015173782A1 (en) * 2014-05-16 2015-11-19 Glaxosmithkline Intellectual Property Management Limited Antibody formulation
AU2015260758B2 (en) * 2014-05-16 2017-11-02 Glaxosmithkline Intellectual Property Management Limited Antibody formulation
US10507241B2 (en) 2014-07-24 2019-12-17 Boehringer Ingelheim International Gmbh Biomarkers useful in the treatment of IL-23A related diseases
US11680096B2 (en) 2014-09-03 2023-06-20 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
US10059763B2 (en) 2014-09-03 2018-08-28 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
US10793629B2 (en) 2014-09-03 2020-10-06 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
US10413593B2 (en) 2014-10-24 2019-09-17 Merck Sharp & Dohme Corp. Co-agonists of the glucagon and GLP-1 receptors
US11801300B2 (en) 2014-12-22 2023-10-31 Novartis Ag Pharmaceutical products and stable liquid compositions of IL-17 antibodies
US11091543B2 (en) 2015-05-07 2021-08-17 Swedish Orphan Biovitrum Ag Methods, compositions and dosing regimens for treating or preventing interferon-gamma related indications
US11034760B2 (en) 2015-05-07 2021-06-15 Swedish Orphen Biovitrum AG Methods and compositions for diagnosis and treatment of disorders in patients with elevated levels of CXCL9 and other biomarkers
US11236158B2 (en) 2015-05-07 2022-02-01 Swedish Orphan Biovitrum Ag Methods, compositions and dosing regimens for treating or preventing interferon-gamma related indications
KR102000867B1 (en) * 2015-06-17 2019-07-16 일라이 릴리 앤드 캄파니 Anti-CGRP antibody preparation
EA037580B1 (en) * 2015-06-17 2021-04-16 Эли Лилли Энд Компани Anti-cgrp antibody formulation
WO2016205037A1 (en) * 2015-06-17 2016-12-22 Eli Lilly And Company Anti-cgrp antibody formulation
US11498959B2 (en) 2015-06-17 2022-11-15 Eli Lilly And Company Anti-CGRP antibody formulation
KR20180002858A (en) * 2015-06-17 2018-01-08 일라이 릴리 앤드 캄파니 Anti-CGRP antibody preparation
WO2017121867A1 (en) 2016-01-13 2017-07-20 Genmab A/S Formulation for antibody and drug conjugate thereof
WO2018027195A1 (en) * 2016-08-05 2018-02-08 Abbvie Biotherapeutics Inc. Compositions containing reduced amounts of daclizumab acidic isoforms and methods for preparing the same
KR102359181B1 (en) 2017-03-01 2022-02-08 메디뮨 리미티드 Anti-RSV monoclonal antibody preparations
AU2018227502B2 (en) * 2017-03-01 2022-08-11 Medimmune Limited Anti-RSV monoclonal antibody formulation
GB2564175B (en) * 2017-03-01 2021-12-08 Medimmune Ltd Anti-RSV monoclonal antibody formulation
EP3589648A4 (en) * 2017-03-01 2020-12-23 Medimmune Limited Anti-rsv monoclonal antibody formulation
US11667698B2 (en) 2017-03-01 2023-06-06 Medimmune Limited Anti-RSV monoclonal antibody formulation
CN110418803A (en) * 2017-03-01 2019-11-05 免疫医疗有限公司 Anti- RSV monoclonal antibody preparation
CN110418803B (en) * 2017-03-01 2024-01-19 免疫医疗有限公司 anti-RSV monoclonal antibody formulations
KR20190125318A (en) * 2017-03-01 2019-11-06 메디뮨 리미티드 Anti-RSV Monoclonal Antibody Formulations
US11286294B2 (en) 2017-03-01 2022-03-29 Medimmune Limited Anti-RSV monoclonal antibody formulation
US11845798B2 (en) 2017-05-02 2023-12-19 Merck Sharp & Dohme Llc Formulations of anti-LAG3 antibodies and co-formulations of anti-LAG3 antibodies and anti-PD-1 antibodies
US11633476B2 (en) 2017-05-02 2023-04-25 Merck Sharp & Dohme Llc Stable formulations of programmed death receptor 1 (PD-1) antibodies and methods of use thereof
US11802156B2 (en) 2017-07-14 2023-10-31 Pfizer Inc. Antibodies to MAdCAM
WO2021146336A1 (en) * 2020-01-13 2021-07-22 Aptevo Research And Development Llc Methods and compositions for preventing adsorption of therapeutic proteins to drug delivery system components
WO2021233853A1 (en) * 2020-05-19 2021-11-25 F. Hoffmann-La Roche Ag The use of chelators for the prevention of visible particle formation in parenteral protein solutions

Also Published As

Publication number Publication date
KR20050044365A (en) 2005-05-12
AU2002363339B2 (en) 2008-02-07
CA2466034C (en) 2012-12-18
CY2016044I1 (en) 2017-07-12
JP2011068675A (en) 2011-04-07
US8465739B2 (en) 2013-06-18
ES2392073T3 (en) 2012-12-04
CN1292655C (en) 2007-01-03
PT1441589E (en) 2012-08-13
NZ532896A (en) 2007-08-31
ATE556591T1 (en) 2012-05-15
CA2466034A1 (en) 2003-05-15
CN1612689A (en) 2005-05-04
KR100913714B1 (en) 2009-08-24
HK1074750A1 (en) 2005-11-25
EP1441589A2 (en) 2004-08-04
WO2003039485A3 (en) 2004-02-12
US20110070231A1 (en) 2011-03-24
JP5290489B2 (en) 2013-09-18
JP2005508981A (en) 2005-04-07
IL161677A0 (en) 2004-09-27
EP1441589A4 (en) 2007-07-04
US20110318343A1 (en) 2011-12-29
DK1441589T3 (en) 2012-08-06
US20030138417A1 (en) 2003-07-24
EP1441589B1 (en) 2012-05-09
IL161677A (en) 2010-06-16
LU93314I2 (en) 2019-11-20
CY2016044I2 (en) 2017-07-12

Similar Documents

Publication Publication Date Title
US8465739B2 (en) Stable aqueous pharmaceutical formulations of daclizumab antibodies
AU2002363339A1 (en) Stable liquid pharmaceutical formulation of IGG antibodies
US8298530B2 (en) Stable lyophilized pharmaceutical formulation of IgG antibodies
EP2568960B1 (en) Liquid formulation of polypeptides containing an fc domain of an immunoglobulin
WO2004039337A2 (en) Stable liquid pharmaceutical formulation of antibodies that are prone to isomerization
AU2003211991B2 (en) Antibody-containing solution formulations
US20110158987A1 (en) Novel antibody formulation
AU2006330858A1 (en) Protein formulations with reduced viscosity and uses thereof
CN110787292B (en) Programmed cell death receptor 1 antibody preparation and application thereof
JP4422485B2 (en) Lyophilized preparation containing immune cytokines
US20040170632A1 (en) Liquid formulation comprising cetuximab and a fatty acid ester of polyoxyethylene sorbitan
CN111375057A (en) Pharmaceutical formulation comprising anti-Her 2 monoclonal antibody
EP3849519B1 (en) A composition comprising a protein and a polyalkoxy fatty acyl surfactant

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 161677

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2466034

Country of ref document: CA

Ref document number: 01203/DELNP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2002802895

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2002363339

Country of ref document: AU

Ref document number: 1020047006962

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2003541777

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 532896

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 20028267273

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2002802895

Country of ref document: EP