MX2008012295A - Anti-igf-1r human monoclonal antibody formulation. - Google Patents

Anti-igf-1r human monoclonal antibody formulation.

Info

Publication number
MX2008012295A
MX2008012295A MX2008012295A MX2008012295A MX2008012295A MX 2008012295 A MX2008012295 A MX 2008012295A MX 2008012295 A MX2008012295 A MX 2008012295A MX 2008012295 A MX2008012295 A MX 2008012295A MX 2008012295 A MX2008012295 A MX 2008012295A
Authority
MX
Mexico
Prior art keywords
igf
histidine
polysorbate
humab
nacl
Prior art date
Application number
MX2008012295A
Other languages
Spanish (es)
Inventor
Hanns-Christian Mahler
Edelbert Grossmann
Astrid Pappenberger
Oliver Boris Stauch
Jan Olaf Stracke
Original Assignee
Hoffmann La Roche
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hoffmann La Roche filed Critical Hoffmann La Roche
Publication of MX2008012295A publication Critical patent/MX2008012295A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • 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
    • 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
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Mycology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Inorganic Chemistry (AREA)
  • Dermatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to an anti-IGF-1R human monoclonal antibody formulation, a process for the preparation and uses thereof.

Description

FORMULATION OF AN ANTI-IGF-1R HUMAN MONOCLONAL ANTIBODY DESCRIPTION OF THE INVENTION The present invention relates to a formulation of a human anti-IGF-1R monoclonal antibody, a process for its preparation and uses thereof. In one aspect, the invention relates to an IGF-IR formulation comprising: between 1 and about 150 mg / mL of huMab IGF-IR, between 0.001 and about 1% of at least one surfactant, and between 1 and about 100 mM of a buffer, a pH of between about 5.0 and about 7.0. IGF-IR (type 1 insulin-like growth factor receptor) is involved in promoting the oncogenic transformation, growth, and survival of cancer cells. High levels of IGF-IR expression have been detected in a wide range of human neoplasms. In addition, high levels of IGF-I and IGF-II expression have been detected in tumors and associated stromal cells and can stimulate the growth of cancer cells in an autocrine or paracrine fashion. Epidemiological studies have correlated in the upper quintile, the plasma levels of IGF-I with an increased risk for prostate, colon, lung, and breast cancer. In addition to these roles in the proliferation of cancer cells, the IGF-IR protects Ref .: 196404 Cells of apoptosis caused by growth factor deprivation, anchorage independence, or treatment with cytotoxic drugs. A promising strategy to inhibit the function of IGF-IR in cancer cells is to apply human anti-IGF-1R antibodies that bind to the extracellular domains of IGF-IR and inhibit receptor activation. Such a fully human antagonistic monoclonal antibody, designated huMab IGF-IR, has been developed which binds specifically to the insulin-like growth factor-1 type receptor (IGF-IR) and inhibits signal transduction and proliferation functions. of the receptor in cancer cells. The antibody comprised in the formulation of the invention was first described in the PCT patent application No. O2005 / 005635 in which the Applicant is the owner and the content thereof, especially the claims are incorporated herein by reference. As described in WO2005 / 005635, said antibody binds to IGF-IR and inhibits the binding of IGF-I and IGF-II to IGF-IR, and is characterized in that: a) it is of the IgGl isotype, b) sample a ratio of IC50 values of inhibition of the binding of IGF-I to IGF-IR to the inhibition of IGF-II binding to IGF-IR from 1: 3 to 3: 1, c) at a concentration of 5 nM inhibits at least one 80%, preferably at least 90%, phosphorylation of IGF-1R in a cell phosphorylation assay using HT29 cells in a medium containing 0.5% heat-inactivated bovine fetal serum (FCS) compared to the same assay without said antibody, and d) at a concentration of 10 μ, does not show IGF-1R stimulating activity measured as the phosphorylation of pkB in a cell phosphorylation assay using 3T3 cells providing from 400,000 to 600,000 IGF-1R molecules per cell in a medium containing 0.5% heat inactivated fetal bovine serum (FCS) compared to the same assay without said antibody. The antibodies included in the formulation according to the invention show benefits for patients in need of antitumor therapy and provide reduction of tumor growth and a significant prolongation of the time of progression. The antibodies comprised in the formulation according to the invention possess novel and inventive properties which give rise to a benefit for a patient suffering from a disease associated with a deregulation of IGF, especially a tumor disease. The antibodies included in the formulation of the invention are characterized by the aforementioned properties - - previously. The properties are therefore especially specific for binding to IGF-1R, inhibiting the binding of IGF-I and IGF-II to IGF-1R in the aforementioned proportion, being of the IgG1 isotype, and not activating IGF-1R signaling even when the cells overexpress IGF-1R at a 200-fold concentration its IC50 value. Antibodies that do not possess an "IGF-I mimetic activity" provide a strong advantage when used as a therapeutic agent. The term "human monoclonal antibody anti-IGF-lR" or "hu Ab IGF-1R" denotes an antibody as described and claimed in WO2005 / 005635, the content of which, especially the claims, are incorporated herein by reference. The term "antibody" encompasses the different forms of antibodies including but not limited to whole antibodies, antibody fragments, human antibodies, humanized antibodies and genetically modified antibodies while retaining the characteristic properties according to the invention. "Antibody fragments" comprises a portion of a full length antibody, generally at least the antigen binding portion or the variable region thereof. Examples of antibody fragments include diabodies, single-chain antibody molecules, immunotoxins, and - - Fierce multispecific antibodies formed from antibody fragments. In addition, the antibody fragments comprise single chain polypeptides having the characteristics of a VH chain, being able to assemble together with the VL chain or a VL chain that binds to IGF-1R, being able to assemble together with a V H chain to a functional antigen-binding region and thus provide the property of inhibiting the binding of IGF-I and IGF-II to IGF-1R. "Antibody fragments" also comprises such fragments that per se are not capable of providing effector functions (ADCC / CDC) but provide this function in a manner according to the invention after combining with an appropriate constant antibody domain (s). The terms "monoclonal antibody" or "monoclonal antibody composition" as used herein refers to a preparation of antibody molecules of a single amino acid composition. Accordingly, the term "human monoclonal antibody" refers to antibodies that exhibit a unique binding specificity possessing variable and constant regions derived from a human germline of immunoglobulin sequences. In one embodiment, human monoclonal antibodies are produced by a hybridoma that includes a B cell obtained from a transgenic non-human animal, eg, a transgenic mouse, which possesses a genome comprising a heavy human chain transgenic and a transgenic light human chain fused with an immortalized cell. The term "chimeric antibody" refers to a monoclonal antibody comprising a variable region, i.e., a binding region, from a source or species and at least a portion of a constant region derived from a different source or species, normally prepared by recombinant DNA techniques. Chimeric antibodies comprising a variable murine region and a human constant region are especially preferred. Such chimeric murine / human antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding murine immunoglobulin variable regions and DNA segments encoding human immunoglobulin constant regions. Other forms of "chimeric antibodies" encompassed by the present invention are those in which the class or subclass has been modified or changed from that of the original antibody. Such "chimeric" antibodies are also referred to as "class change antibodies." Methods for producing chimeric antibodies involve conventional recombinant DNA techniques and transfection of genes that are well known in the art. See, for example, Morrison, S.L., et al., Proc. Nati Acad Sci. USA 81 (1984) 6851-6855; U.S. Patent Nos. 5,202,238 and 5,204,244.
The term "humanized antibody" refers to antibodies in which the framework or "complementarity determining regions" (CDRs) have been modified to comprise the CDR of an immunoglobulin of different specificity when compared to that of the parental immunoglobulin. In a preferred embodiment, a murine CDR is grafted into a framework region of a human antibody to prepare the "humanized antibody." See, e.g., Riechmann, L., et al., Nature 332 (1988) 323-327; and Neuberger, M.S., et al., Nature 314 (1985) 268-270. Particularly preferred CDRs correspond to those representing the sequences recognizing the antigens mentioned above for the chimeric and bifunctional antibodies. The term "human antibody", as used herein, is intended to include antibodies with variable regions and constants derived from a human germline of immunoglobulin sequences. The variable heavy chain is preferably derived from the DP-50 germline sequence (GenBank L06618) and the variable light chain is preferably derived from the germline sequence L6 (GenBank X01668). The constant regions of the antibody are constant regions of the human IgGl type. Such regions can be allotypic and are described in, for example, Johnson, G., and Wu, T.T., Nucleic Acids Res. 28 (2000) 214-218 and in the databases cited there and are useful while retain the induction properties of ADCC and preferably CDC according to the invention. The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant methods, such as antibodies isolated from a host cell such as SP2-0, NSO cells or CHO or an animal (eg, a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected in a host cell. Such recombinant human antibodies possess variable and constant regions derived from the sequences of the human germline of immunoglobulins in a reorganized form. Recombinant human antibodies according to the invention have been subjected to somatic hypermutation in vivo. Thus, the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to the sequences of the human germ lines of VH and VL, may not exist naturally within the human repertoire of germ lines of antibodies in vivo. As used herein, "binding" refers to the binding of an antibody to IGF-1R with an affi of about 10"13 to 10 ~ 8 M (KD), preferably about 10" 13 to 10 ~ 9. M. The term "nucleic acid molecule", as used in used here, intended to include DNA molecules and RNA molecules. A nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA. The "constant domains" are not directly involved in the binding of antibody to an antigen but are involved in effector functions (ADCC, complement binding, and (CDC) .The constant domain of an antibody according to with the invention is of the IgGl type The human constant domains possessing these characteristics are described in detail by Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. 1991), and by Brüggemann, M., Et al., J. Exp. Med. 166 (1987) 1351-1361; Love, TW, et al., Methods Enzymol, 178 (1989) 515-527. shown in Seq ID Nos. 5 to 8 in WO2005 / 005635. Other useful and preferred constant domains are the constant domains of the antibodies obtainable from the hybridoma cell lines deposited with DSMZ for this invention. the invention provide the u complement nion. ADCC and optionally CDC are provided by the combination of variable and constant domains. The "variable region" (variable region of a light chain (VL), the variable region of a heavy chain (VH)) as used herein denotes each pair of light and heavy chains that are directly involved in the binding of the antibody to the antigen. The variable light and heavy human chain domains have the same general structure and each domain comprises four framework regions (FR) whose regions are widely conserved, connected by three "hypervariable regions" (or complementarity determining regions, CDR). The framework regions adopt a β-sheet conformation and the CDRs can form loops connecting the β-sheet structure. The CDRs in each chain are supported in their three-dimensional structure by the framework regions and together with the CDRs of the other chain form the antigen-binding site. The CDR3 regions of the light and heavy chains of the antibody play a particularly important role in the binding specificity / affi of the antibodies according to the invention and therefore provide another object of the invention. The terms "hypervariable region" or "antigen binding portion of an antibody" when used herein refers to amino acid residues of an antibody that are responsible for antigen binding. The hypervariable region comprises amino acid residues of the "complementarity determining regions" or "CDR". The "Frame" or "FR" regions are those variable domain regions different from the hypervariable region residues as defined herein.
Therefore, the light and heavy chains of an antibody comprise the FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4 domains from the N-terminus to the C-terminal end. Especially, CDR3 of the heavy chain is the region that contributes most to antigen binding. The CDR and FR regions are determined according to the standard definition of Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) and / or those residues of a "hypervariable loop". The term "binding to IGF-1R" as used herein means binding of the antibody to IGF-1R in an in vitro assay, preferably in a binding assay in which the antibody is bound to a surface and the binding of IGF- 1R is measured by Surface Plasmon Resonance (RPS). Binding means a binding affinity (KD) of 10 ~ 8 M or less, preferably 1CT13 to 1CT9 M. The binding to IGF-1R can be investigated by a BIAcore assay (Pharmacia Biosensor AB, Uppsala, Sweden). Affinity of the binding is defined by the term ka (constant ratio for the antibody association of the antibody / antigen complex), kd (dissociation constant), and KD (kd / ka). The antibodies according to the invention show a KD of 10"10 M or less The binding of IGF-I and IGF-II to IGF-1R is also inhibited by the antibodies according to the invention.
- - Inhibition is measured as the IC50 in an assay for the binding of IGF-I / IGF-II to IGF-1R on tumor cells. Such assay is described in Example 7. In such assay, the amount of IGF-I or IGF-II or radiolabeled IGF-1R-binding fragments thereof bound to the IGF-1R provided on the surface of tumor cells (e.g. , HT29) is measured without and with incremental concentrations of the antibody. The IC50 values of the antibodies according to the invention for the binding of IGF-I and IGF-II to IGF-1R are not more than 2 nM and the proportion of the IC50 values for the binding of IGF-I / IGF -II to IGF-1R is around 1: 3 to 3: 1. The IC 50 values are measured as the mean or median of the values of at least three independent measurements. Unique IC50 values may be out of range. The term "inhibit the binding of IGF-I and IGF-II to IGF-1R" as used herein, refers to the inhibition of IGF-I or IGF-I binding labeled with I 125 to IGF-1R presented in the surface of HT29 tumor cells (ATCC HTB-38) in an in vitro assay. Inhibition means an IC50 value of 2 nM or less. The term "surfactant" as used herein denotes a pharmaceutically acceptable surfactant. In the formulation of the invention, the amount of surfactant is described as a percentage expressed in weight / volume. The most commonly used unit of weight / volume is mg / mL. The - 1 - Suitable pharmaceutically acceptable surfactants include but are not limited to polyethylene sorbitan fatty acid esters, polyethylene polypropylene glycols, polyoxyethylene stearates and sodium dodecyl sulfates. Preferred polyethylene sorbitan esters of polyethylene (20) -sorbitan (synonymous with polysorbate 20, marketed under the trade name of Tween 20 ™) and polyoxyethylene (20) sorbitan monooleate (synonymous with polysorbate 80 marketed under the brand name of Tween 80 ™). Preferred polyethylene-polypropylene glycols are those marketed under the names Pluronic® F68 or Poloxamer 188 ™. Preferred polyoxyethylene stearates are those marketed under the trade name of Myrj ™. Preferred polyoxyethylene monolauryl ethers are those marketed under the trade name Brij ™. When polyethylene-sorbitan-polyethylene (20) -sorbitan (Tween 20 ™) and polyoxyethylene (20) sorbitan monooleate (Tween 80 ™) esters are used, they are generally used in an amount of about 0.001 to about 1%, preferably between about 0.005 and about 0.1% and even more preferably between about 0.01% and about 0.02% w / v. The term "buffer" as used herein denotes a pharmaceutically acceptable buffer. A suitable pharmaceutically acceptable buffer comprises but is not limited to histidine buffers, citrate buffers, buffers - - of succinate, acetate buffers and phosphate buffers. Preferred buffers comprise L-histidine or mixtures of L-histidine with L-histidine hydrochloride with isotonicity agents and pH adjustment with an acid or a base known in the art. The histidine buffers mentioned above are generally used in an amount of about lmM to about 100mM, preferably between about 5mM to about 50mM and even more preferably about 20mM. Regardless of the buffer used, the pH will be adjusted to a value ranging between about 5.0 and about 7.0 and preferably between about 5.5 and about 6.5 and more preferably about 6.0. The term "isotonicity agents" as used herein denotes pharmaceutically acceptable isotonicity agents. Isotonicity agents are used to provide an isotonic formulation. An isotonic formulation is a liquid or a reconstituted liquid of a solid form, for example, a lyophilized form and denotes a solution having the same tonicity as some other solution with which it is compared, such as a physiological salt solution and blood serum. . Suitable isotonicity agents include but are not limited to sodium chloride, potassium chloride, glucose, glycerin and any component of the group of amino acids, sugars and combinations thereof. The - isotonicity agents are generally used in a total amount of between about 5 mM to about 350 mM. The term "liquid" as used herein in conjunction with the formulation according to the invention denotes a formulation that is liquid at a temperature of between at least about 2 to about 8 ° C. The term "freeze-dried" as used herein in conjunction with the formulation according to the invention denotes a formulation that is dried by freezing the formulation and subsequent sublimation of the frozen content ice by any method known in the art on lyophilization, for example commercially available lyophilization devices. The term "amino acid" as used herein denotes an amino acid in an amount of about 1 to about 100 mg / mL comprising but not limited to arginine, glycine, ornithine, lysine, histidine, glutamic acid, asparagic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophan, methionine, serine, proline. The term "sugar" as used herein denotes a pharmaceutically acceptable sugar used in an amount of about 25 mM to about 500 mM. Suitable sugars include but are not limited to trehalose, sucrose, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N-methylglucosamine (also called "Meglumine"), galactosamine and neuraminic acid. The term "stabilizer" refers to pharmaceutically acceptable stabilizers, such as, but not limited to, amino acids and sugars as described in the preceding sections as well as commercially available cyclodextrins and dextrans of any kind and molecular weight as known in the art. . The term "antioxidant" denotes a pharmaceutically acceptable antioxidant. As mentioned above, in one aspect, the invention relates to an IGF-IR formulation comprising: between 1 and about 150 mg / mL of huMab IGF-IR, - between 0.001 and about 1% of less a surfactant, and between 1 and about 100 mM of a buffer, at a pH between about 5.0 and about 7.0. The formulation according to the invention preferably comprises between about 0.001 and about 1% of at least one surfactant. In a certain embodiment, the formulation according to the invention comprises: between 1 and about 150 mg / mL of huMab IGF-IR, - between 0.005 and about 0.05% of at least one - 1 - surfactant, and between 1 and about 100 mM of a buffer, at a pH between about 5.0 and about 7.0. The formulation according to the invention may be in the form of a liquid, in lyophilized form or in the form of reconstituted liquid in a lyophilized form. In a certain embodiment, the formulation according to the invention is a lyophilized formulation. The lyophilized formulation according to the invention has the advantage of an improved stability with respect to the formation of particles and aggregates of higher molecular weight which is normally difficult to achieve with liquid formulations in the same concentration of the anti-IGF-1R monoclonal antibodies. humans described. The formulation according to the invention can be administered by intravenous (i.v.), subcutaneous (s.c.) administration or any other parental means such as those known in the pharmaceutical art. The formulation of the invention may further comprise one or more isotonicity agents in an amount of about 5 mM to about 350 mM. Suitable isotonicity agents can be selected from the group consisting of sodium chloride (NaCl), potassium chloride, sugars comprising glucose, glycerin, amino acids, and combinations thereof.
The formulation of the invention may further comprise a sugar in an amount of about 25 mM to about 500 mM. Suitable sugars can be selected from the group comprised of trehalose, sucrose, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N-methylglucosamine, galactosamine, neuraminic acid and combinations thereof. The formulation of the invention may further comprise one or more of the following ingredients: antioxidants, ascorbic acid, glutathione, preservatives, for example, m-cresol, phenol, benzylalcohol, methylparaben, propylparaben, chlorbutanol, thiomersal, benzalkonium chloride, cyclodextrin, for example, hydroxypropyl-p-cyclodextrin, sulfobutyl-β-cyclodextrin, β-Cyclodextrin, polyethylene glycol, eg, PEG 3000, 3350, 4000, 6000, albumin, human serum albumin (HSA), bovine serum albumin (BSA) ), polyhydrated alcohol, glycerol, ethanol, mannitol, salts, acetate salts (e.g., sodium acetate), magnesium chloride, calcium chloride, tromethamine, EDTA, (e.g., Na-EDTA). The formulation of the invention may further comprise one or more stabilizers as defined above and ingredients also known in the art as "lyoprotectants" such as sugars, sugars of alcohols, amino acids and dextrans as are known in the art. In a certain embodiment, the formulation of the invention It comprises the following formulations, both in its liquid, lyophilized or reconstituted liquid form in a lyophilized form: entity 1 and around 150 mg / mL of huMab IGF-IR, 0.01% Tween 20 p / v, L-histidine 20 mM, NaCl 140 mM, at pH 6.0. The formulation according to the invention also comprises the following specific formulations: - 25 mg / mL of huMab IGF-IR, - polysorbate 20 at 0.01%, - L-histidine 20 mM, -NaCl 140 mM, - at pH 6.0. or - 25 mg / mL of huMab IGF-IR, - polysorbate 20 0.03%, - L-histidine 20 mM, - NaCl 140 mM, - at pH 6.0. or - 25 mg / mL of huMab IGF-IR, - polysorbate 20 at 0.05%, - L-histidine 20 mM, 140 mM NaCl, at pH 6.0. mg / mL of huMab IGF-1R, polysorbate 20 at 0.01%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0. mg / mL of huMab IGF-1R, polysorbate 20 0.03%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0. mg / mL of huMab IGF-1R, polysorbate 20 at 0.05%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0. mg / mL of huMab IGF-1R, 20 mM L-histidine, 140 mM NaCl, at pH 5.5. - 25 mg / mL of huMab IGF-IR, - polysorbate 20 to 0.01%, - 20 mM L-histidine, - 250 mM dihydrated trehalose, - at pH 5.5. or - 25 mg / mL of huMab IGF-IR, - 20 mM L-histidine, - 250 mM dihydrated trehalose, - at pH 6.0. or - 25 mg / mL of huMab IGF-IR, - 20% 0.01% polysorbate, - 20 mM L-histidine, - 250 mM trehalose dihydrate, - at pH 6.0. or - 25 mg / mL of huMab IGF-IR, - L-histidine 20 mM, - trehalose dihydrate 250 mM, - Polysorbate 20 at 0.05%, - at pH 6.0. or - 25 mg / mL of huMab IGF-IR, - 20 mM L-histidine, - 60 mM dihydrated trehalose, - 0.01% polysorbate 20, - at pH 6.0. or - 25 mg / mL of huMab IGF-1R, - 20 mM Succinate, - 250 mM dihydrated trehalose, - 0.01% polysorbate 20, - at pH 5.5. or - 25 mg / mL of huMab IGF-1R, - 20 mM L-histidine, - 60 mM dihydrated trehalose, - 0.01% polysorbate 20, - at pH 6.0. In a preferred embodiment of the formulation according to the invention, the formulation is in a lyophilized form and comprises after reconstitution with the appropriate amount of water for injection: - 25 mg / mL of huMab IGF-1R, - polysorbate 20 to 0.01 %, - 20 mM L-histidine, - 250 mM dihydrated trehalose, - at pH 5.5.
This formulation shows good stability after storage for approximately 6 months at 2-8 ° C and 25 ° C with species of high molecular weight less than 1.5% and without the formation of visible and sub / visible particles. Agitation and multiple freeze / thaw steps are applied to the liquid formulation to stimulate physical stress conditions that potentially occur during manufacturing, for example, by filtration by pressure and filling, lyophilization and final operations. This formulation was found to be stable after one week of agitation at 5 ° C and 25 ° C. The formation of visible and sub / visible particles could not be detected and there was no significant increase in the fraction of high molecular weight species indicating the formation of soluble aggregates (see figure 1).
EXAMPLES Liquid and lyophilized drug formulations for intravenous administration according to the invention were developed as follows: Preparation of liquid formulations Solutions of approximately 10 and 25 mg / mL of huMab IGF-1R in the production buffer (histidine buffer 10 mM approximately, with 150 mM NaCl approximately) were dialyzed against large volumes of water and the respective salt buffer systems for the final formulation (see table with the exact composition of the formulations). If necessary, the protein concentration was increased by filtration using commercially available centrifugal filter devices) before dialysis and then adjusted to the desired protein concentration by dilution with dialysis buffer. Sugars and salts for protein stabilization and for tonicity adjustment were added to the dialysis buffer as needed. Surfactant was added to the formulations after dialysis as concentrated stock solutions of 2 to 40 times. Alternately, a buffer and concentration exchange was performed using a commercially available tangential flow filtration device, for example, ÁKTA CF (GE Healthcare) with a Sartorius Hydrosart membrane (30,000 Da molecular weight cutoff). Ingredients such as sugars, salts or surfactants were added after buffer exchange using the appropriate amounts of concentrated stock solutions. All formulations were sterile filtered through low-protein filters of 0.22 μ? T? and they were aliquoted aseptically in sterile 6 mL glass vials closed with Teflon-coated rubber stoppers and alucrimp caps. These formulations were stored at different - 5 - temperatures during different time intervals and were removed for the analysis at the times indicated in the individual paragraphs by 1) UV spectrophotometry, 2) Size Exclusion Chromatography (SEC) and 3) dimming of the light to determine the turbidity of the solution. In addition, the analysis for visible particles was performed for each sample using a Seidenader V90-T instrument. The appearance of subvisible particles was assessed using a Royco HIAC device. Preparation of lyophilized formulations The 25 mg / mL solutions of hu ab IGF-1R were prepared as described above for the liquid formulations. All the formulations were filtered sterile through low-binding proteins to 0.22 μp? and they were aliquoted aseptically in sterile glass vials. The vials were partially closed with Teflon-coated rubber stoppers suitable for use in lyophilization processes and transferred to the drying chamber of the lyophilizer. Any lyophilization method known in the art is intended to be within the scope of the invention. For example, the lyophilization process used for this study includes cooling the formulation from room temperature to approx. 5 ° C (pre-cooling) followed by freezing at -40 ° C (Freezing I) at a rate of progression of about 1 ° C / min at 5 ° C / min. The first step - - drying can be applied at a rate of 0.3 to 0.5 ° C / min from -40 ° C to -30 ° C and then maintained at -30 ° C for at least 50 hours at a chamber pressure of approximately 75 to 80 mTorr. A second drying step can be carried out at a progression rate of 0.1 to 0.3 ° C / min from -30 ° C to 25 ° C and maintained at 25 ° C for at least 5 hours at a chamber pressure of about 50 to 80 mTorr. It was found that in the huMab IGF-1R formulations that were dried using the described lyophilization processes, the reconstitution times were conveniently fast, being around < 5 min The lyophilization was carried out in a LyoStar II lyophilizer (FTS Systems, Stone Ridge, NY, USA and Usifroid Orion, Maurepas, France). All lyophilized pastes in this study had a residual water content of approximately 0.1 to 5.0% as determined by the Karl-Fischer method. The lyophilized vials were stored at different temperatures for different time intervals. The lyophilized formulations were reconstituted with the respective volume of water for injection (API) before analysis by 1) UV spectrophotometry, 2) Determination of reconstitution time 3) Size Exclusion Chromatography (SEC) and 4 ) darkening of the light to determine the turbidity of the solution. In addition, the analysis for visible particles was performed for each sample using a Seidenader V90-T instrument (Seidenader, Marktschwaben, Germany). The appearance of subvisible particles was assessed using a Royco HIAC device. Size Exclusion Chromatography (SEC) was performed to detect soluble high molecular weight (aggregates) and low molecular weight hydrolysis products in the formulations. The method used a Merck Hitachi 7000 HPLC instrument or a Waters Alliance 2795 with UV detector (detection wavelength? (280 nm) Both instruments were equipped with a TSK G3000 SWXL column, the method used K2HP04 0.2M / KCL 0.25M, pH 7.0 as a mobile phase, the flow rate was 0.5 mL / min (isocritic acid), the execution time was 30 min at a column temperature of 25 ° C. UV spectroscopy for the determination of protein concentration It was performed after dilution of the samples at an antibody concentration of 0.5 mg / mL in a Uvikon 932 (Kontron Instruments) at a wavelength of 278 nm and in a Varian Cary Bio UV spectrophotometer at 280 nm respectively. of the turbidity, the darkening of the light in FTU (turbidity units) was measured using a HACH 2100AN turbidimeter at room temperature Compositions of liquid product formulations huMAb IGF-1R according to the invention and stability data after 3 months of storage at 2-8 ° C Formulation A is a liquid formulation with the composition 25 mg / mL of huMab IGF-1R, 20 mM L-histidine, 140 NaCl mM, polysorbate 20 at 0.01%, at pH 6.0.
Formulation B is a liquid formulation with the 25 mg / mL composition of huMab IGF-1R, 20 mM L-histidine, 140 mM NaCl, 0.03% polysorbate 20, at pH 6.0.
Formulation C is a liquid formulation with the 25 mg / mL composition of huMab IGF-1R, 20 mM L-histidine, 140 mM NaCl, 0.05% polysorbate 20, at pH 6.0.
Concentration Species of high weight Turbidez Molecular protein time (%) in initial FTU 24.42 0.85 10.1 4 weeks 21.57 1.02 10.5 8 weeks 25.0 1.36 10.6 12 weeks 23.97 1.51 10.2 Formulation D is a liquid composition 10 mg / mL composition of huMab IGF-1R, L-histidine 20 140 mM, polysorbate 20 0.01%, at pH 6.0 Formulation E is a liquid formulation with the 10 mg / mL composition of huMab IGF-1R, 20 mM L-histidine, 140 mM NaCl, 0.03% polysorbate 20, at pH 6.0. Concentration High Turbidity Species Protein time molecular weight (%) in initial FTU 9.98 0.69 4.26 4 weeks 9.68 0.79 4.72 8 weeks 9.68 1.05 4.54 12 weeks 9.62 1.59 4.26 - - Formulation F is a 10 mg / mL composition liquid formulation of huMab IGF-1R, 140 mM L-histidine 20, 0.05% polysorbate 20, at pH 6.0 Formulation G is a liquid formulation with the composition 10 mg / mL of huMab IGF-1R, 20 mM L-histidine, 140 mM NaCl, at pH 5.5.
Formulation H is a liquid formulation with the composition 25 mg / mL of huMab IGF-1R, L-histidine 20 mM, trehalose dihydrate 250 mM, polysorbate 20 0.01%, at pH 5.5 Concentration High Turbidity Species Protein time molecular weight (%) in initial FTU 27.1 1.29 3.58 4 weeks 26.9 1.27 3.74 8 weeks 29.33 1.43 5.03 12 weeks 26.44 1.38 4.10 Formulation I is a liquid formulation with the composition 25 mg / mL of hu ab IGF-1R, 20 mM L-histidine, 250 mM dihydrated trehalose, at pH 6.0 Formulation J is a liquid formulation with the composition 25 mg / mL of huMab IGF -1R, 20 mM L-histidine, 250 mM dihydrated trehalose, 0.01% polysorbate 20, at pH 6.0 Formulation K is a liquid formulation with the composition 25 mg / mL of huMab IGF-1R, 20 mM L-histidine, trehalose dihydrate 250 mM, polysorbate 20 0.05%, at pH 6.0 Formulation L is a liquid formulation with the composition 25 mg / mL of huMab IGF-1R, L-histidine 20 mM, trehalose dihydrate 60 mM, polysorbate 20 0.01%, at pH 6.0 Formulation M is a liquid formulation with the 25 mg / mL composition of huMab IGF-1R, 20 mM Succinate, 250 mM dihydrated trehalose, 0.01% polysorbate 20, at pH 5.5 Concentration High Turbidity Species Protein time molecular weight (%) in initial FTU 26.13 1.49 7.06 4 weeks 27.93 1.46 7.25 8 weeks 26.59 1.62 8.20 12 weeks 26.89 1.62 7.10 Compositions of lyophilized product of huMAb IGF-1R according to the invention and stability data after 3 months of storage at 2-8 ° C Formulation N is a lyophilized formulation with the composition of the reconstituted solution of 25 mg / mL of huMab IGF -1R, 20 mM L-histidine, 60 mM dihydrated trehalose, 0.01% polysorbate 20, at pH 6.0 Formulation O is a lyophilized formulation with the reconstituted solution composition of 25 mg / mL of huMab IGF-1R, 20 mM L-histidine, 60 mM sucrose, 0.01% polysorbate 20, at pH 6.0 Concentration High Turbidity Species Protein time molecular weight (%) in initial FTU 24.77 0.93 5.77 4 weeks 24.81 0.93 6.79 8 weeks 24.11 0.88 5.38 12 weeks 24.32 1.03 393 Formulation P is a lyophilized formulation with the composition of the reconstituted solution of 25 mg / mL of huMab IGF-1R, 20 mM Succinate, 250 mM dihydrated trehalose, 0.01% polysorbate 20, at pH 5.5 Formulation Q is a lyophilized formulation with the reconstituted solution composition of 25 mg / mL of huMab IGF-1R, 20 mM L-histidine, 250 mM dihydrated trehalose, 0.01% polysorbate 20, at pH 5.5 Concentration High Turbidity Species Protein time molecular weight (%) in initial FTU 27.11 1.33 3.59 4 weeks 28.28 1.22 3.49 8 weeks 27.81 1.30 4.65 12 weeks 28.22 1.18 4.04 It is noted that in relation to this date, the best method known to the applicant to carry out the aforementioned invention, is that which is clear from the present description of the invention.

Claims (10)

  1. CLAIMS Having described the invention as above, the content of the following claims is claimed as property: 1. Formulation characterized in that it is selected from a group consisting of: - between about 1 and about 150 mg / mL of huMab IGF-1R, - 0.01% Tween 20 p / v, - 20 mM L-histidine, - 140 mM NaCl, - at pH 6.0. or - 25 mg / mL of huMab IGF-1R, - polysorbate 20 at 0.01%, - L-histidine 20 mM, - NaCl 140 mM, - at pH 6.0; or - 25 mg / mL of huMab IGF-1R, - polysorbate 20 0.03%, - L-histidine 20 mM, - NaCl 140 mM, - at pH 6.0; or 25 mg / mL of huMab IGF-IR, polysorbate 20 at 0.05%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0; 10 mg / mL of huMab IGF-IR, polysorbate 20 at 0.01%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0; 10 mg / mL of huMab IGF-IR, polysorbate 20 0.03%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0; 10 mg / mL of huMab IGF-IR, polysorbate 20 at 0.05%, L-histidine 20 mM, NaCl 140 mM, at pH 6.0; 25 mg / mL of huMab IGF-IR, - 20 mM L-histidine, - 140 mM NaCl, - at pH 5.5; or - 25 mg / mL of huMab IGF-IR, - polysorbate 20 to 0.01%, - 20 mM L-histidine, - 250 mM dihydrated trehalose, - at pH 5.5; or - 25 mg / mL of huMab IGF-IR, - 20 mM L-histidine, - 250 mM dihydrated trehalose, - at pH 6.0; or - 25 mg / mL of huMab IGF-IR, - polysorbate 20 to 0.01%, - 20 mM L-histidine, 250 mM dihydrated trehalose, - at pH 6.0; or - 25 mg / mL of huMab IGF-IR, - L-histidine 20 mM, - trehalose dihydrate 250 mM, - polysorbate 20 0.05%, - at pH 6.0; or - 25 mg / mL of huMab IGF-1R, - 20 mM L-histidine, - 60 mM dihydrated trehalose, - 0.01% polysorbate 20, - at pH 6.0; or - 25 mg / mL of huMab IGF-1R, - 20 mM Succinate, - 250 mM dihydrated trehalose, - 0.01% polysorbate 20, - at pH 5.5; or - 25 mg / mL of huMab IGF-1R, - 20 mM L-histidine, - 60 mM dihydrated trehalose, - 0.01% polysorbate 20, - at pH 6.0.
  2. 2. Formulation according to claim 1, characterized in that it is in the form of liquid, in a lyophilized form or in a reconstituted liquid form of a lyophilized form.
  3. 3. Formulation according to any of claims 1 or 2, characterized in that it can administered by intravenous (i.v.) or subcutaneous (s.c.) administration or any other parental administration.
  4. 4. Formulation according to any of claims 1 to 3, characterized in that it further comprises one or more isotonicity agents in an amount of about 5 mM to about 350 mM.
  5. 5. Formulation according to claim 4, characterized in that the isotonicity agents are selected from the group consisting of sodium chloride (NaCl), potassium chloride, sugars such as glucose, glycerin, amino acids and combinations thereof.
  6. 6. Formulation according to any of claims 1 to 5, characterized in that it also comprises a sugar in an amount of about 25 mM to about 500 mM.
  7. Formulation according to claim 6, characterized in that the sugars are selected from the group consisting of trehalose, sucrose, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N-methylglucosamine ("Meglumine" ), galactosamine and neuraminic acid.
  8. 8. Formulation according to any of claims 1 to 7, characterized in that it also comprises one or more ingredients selected from the group consisting of pharmaceutically acceptable: antioxidants, ascorbic acid, glutathione, preservatives, in particular, m-cresol, phenol, benzylalcohol, methylparaben, propylparaben, chlorbutanol, thiomersal, benzalkonium chloride, cyclodextrin, in particular, hydroxypropyl-p-cyclodextrin, sulfobutylethia-β-cyclodextrin, β-Cyclodextrin, polyethylene glycol, in particular, PEG 3000, 3350, 4000 or 6000, albumin, human serum albumin (HSA), bovine serum albumin (BSA), polyhydrated alcohol, glycerol, ethanol, mannitol, salts, acetate salts, in particular sodium acetate, magnesium chloride, calcium chloride, tromethamine, EDTA, in particular Na-EDTA.
  9. 9. Use of a formulation according to any of claims 1 to 8, for the preparation of a medicament useful for treating diseases modulated by the IGF-1R receptor.
  10. 10. Use according to claim 9, wherein the disease is selected from the group consisting of breast cancer, colorectal cancer, non-small cell lung cancer (NSCLC) and prostate cancer.
MX2008012295A 2006-03-28 2007-03-19 Anti-igf-1r human monoclonal antibody formulation. MX2008012295A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP06111848 2006-03-28
PCT/EP2007/052569 WO2007110339A1 (en) 2006-03-28 2007-03-19 Anti-igf-1r human monoclonal antibody formulation

Publications (1)

Publication Number Publication Date
MX2008012295A true MX2008012295A (en) 2008-10-09

Family

ID=37025222

Family Applications (1)

Application Number Title Priority Date Filing Date
MX2008012295A MX2008012295A (en) 2006-03-28 2007-03-19 Anti-igf-1r human monoclonal antibody formulation.

Country Status (18)

Country Link
EP (1) EP1998806A1 (en)
JP (1) JP2009531371A (en)
KR (1) KR20080104160A (en)
CN (1) CN101410137A (en)
AR (1) AR060130A1 (en)
AU (1) AU2007229554A1 (en)
BR (1) BRPI0709229A2 (en)
CA (1) CA2647111A1 (en)
CL (1) CL2007000797A1 (en)
CR (1) CR10295A (en)
EC (1) ECSP088778A (en)
IL (1) IL193904A0 (en)
MA (1) MA30345B1 (en)
MX (1) MX2008012295A (en)
NO (1) NO20083895L (en)
RU (1) RU2008142359A (en)
TW (1) TW200815029A (en)
WO (1) WO2007110339A1 (en)

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2007331712A1 (en) * 2006-12-11 2008-06-19 F. Hoffmann-La Roche Ag Abeta antibody parenteral formulation
GB0702888D0 (en) * 2007-02-14 2007-03-28 Glaxo Group Ltd Novel Antibodies
BRPI0809112A2 (en) * 2007-03-22 2014-08-26 Imclone Llc STABLE ANTIBODY FORMULATIONS
CN101687038A (en) * 2007-07-10 2010-03-31 霍夫曼-拉罗奇有限公司 Novel formulation
US20100249384A1 (en) * 2007-11-29 2010-09-30 Stefan Hepbildikler Immunoglobulin aggregates
CN105126099A (en) * 2007-12-21 2015-12-09 弗·哈夫曼-拉罗切有限公司 Antibody formulation
WO2010069858A1 (en) * 2008-12-19 2010-06-24 F. Hoffmann-La Roche Ag Pharmaceutical composition
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
US9345661B2 (en) 2009-07-31 2016-05-24 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
AR078161A1 (en) 2009-09-11 2011-10-19 Hoffmann La Roche VERY CONCENTRATED PHARMACEUTICAL FORMULATIONS OF AN ANTIBODY ANTI CD20. USE OF THE FORMULATION. TREATMENT METHOD
BR112012013148A2 (en) * 2009-12-29 2017-03-21 F Hoffmann - La Roche Ag pharmaceutical formulation and use
CN102905692B (en) * 2010-05-28 2015-09-16 诺沃—诺迪斯克有限公司 Comprise the stable multi-dose compositions of antibody and antiseptic
KR102031020B1 (en) 2011-03-31 2019-10-14 머크 샤프 앤드 돔 코포레이션 Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
CN102363040B (en) * 2011-10-21 2013-05-01 北京锐瑟科技有限公司 Antimicrobial peptide preparation for mucosal tissues
WO2014177460A1 (en) 2013-04-29 2014-11-06 F. Hoffmann-La Roche Ag Human fcrn-binding modified antibodies and methods of use
CN103505729B (en) * 2013-05-24 2015-10-28 华北制药集团新药研究开发有限责任公司 A kind of stable rabies virus human antibody combination preparation
CN104707146B (en) * 2013-12-16 2019-04-16 浙江海正药业股份有限公司 A kind of pharmaceutical composition containing adalimumab
EP3842455A1 (en) 2014-01-15 2021-06-30 F. Hoffmann-La Roche AG Fc-region variants with improved protein a-binding
CA2950392A1 (en) * 2014-05-28 2015-12-03 Nono Inc. Lyophilized formulation of tat-nr2b9c with acetylation scavenger
AR102198A1 (en) 2014-10-09 2017-02-08 Regeneron Pharma PROCESS TO REDUCE SUBVISIBLE PARTICLES IN A PHARMACEUTICAL FORMULATION
BR112017024610A2 (en) 2015-06-24 2018-07-31 F. Hoffmann-La Roche Ag specific affinity antitransferrin receptor antibodies
CA2999079A1 (en) * 2015-09-28 2017-04-06 Jiangsu Hengrui Medicine Co., Ltd. Stable anti-pd-1 antibody pharmaceutical preparation and application thereof in medicine
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
RU2753390C1 (en) 2015-10-02 2021-08-13 Ф. Хоффманн-Ля Рош Аг Bispecific antibodies to human cd20/human transferrin receptor and methods for their use
CN106199007B (en) * 2016-08-03 2017-04-05 烟台普罗吉生物科技发展有限公司 Protein protective agent
CN109641050A (en) 2016-08-16 2019-04-16 里珍纳龙药品有限公司 Method for being quantified to the individual antibody in mixture
CN109923411B (en) 2016-10-25 2022-05-31 里珍纳龙药品有限公司 Method and system for chromatographic data analysis
RU2019138507A (en) 2017-05-02 2021-06-02 Мерк Шарп И Доум Корп. ANTIBODY AGAINST LAG3 AND JOINT ANTIBODY AGAINST LAG3 AND ANTIBODY AGAINST PD-1
JOP20190260A1 (en) 2017-05-02 2019-10-31 Merck Sharp & Dohme Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
TW202005694A (en) 2018-07-02 2020-02-01 美商里珍納龍藥品有限公司 Systems and methods for preparing a polypeptide from a mixture
CN114324882B (en) * 2020-10-12 2022-12-27 广东菲鹏生物有限公司 Protein stabilizer and application thereof

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2190087T3 (en) * 1997-06-13 2003-07-16 Genentech Inc STABILIZED FORMULATION OF AN ANTIBODY.
CN1854157B (en) * 2001-01-05 2013-01-02 辉瑞大药厂 Antibodies to insulin-like growth factor i receptor
WO2003009817A2 (en) * 2001-07-25 2003-02-06 Protein Design Labs, Inc. Stable lyophilized pharmaceutical formulation of igg antibodies
CA2519113C (en) * 2003-04-02 2012-06-05 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
LT2335725T (en) * 2003-04-04 2017-01-25 Genentech, Inc. High concentration antibody and protein formulations
AR046071A1 (en) * 2003-07-10 2005-11-23 Hoffmann La Roche ANTIBODIES AGAINST RECEIVER I OF THE INSULINAL TYPE GROWTH FACTOR AND THE USES OF THE SAME
DE602004029581D1 (en) * 2003-08-13 2010-11-25 Pfizer Prod Inc MODIFIED HUMANESE IGF-1R ANTIBODIES

Also Published As

Publication number Publication date
IL193904A0 (en) 2011-08-01
KR20080104160A (en) 2008-12-01
AU2007229554A1 (en) 2007-10-04
CA2647111A1 (en) 2007-10-04
TW200815029A (en) 2008-04-01
BRPI0709229A2 (en) 2011-06-28
RU2008142359A (en) 2010-05-10
CL2007000797A1 (en) 2008-01-25
AR060130A1 (en) 2008-05-28
EP1998806A1 (en) 2008-12-10
WO2007110339A1 (en) 2007-10-04
ECSP088778A (en) 2008-10-31
CR10295A (en) 2008-10-06
CN101410137A (en) 2009-04-15
NO20083895L (en) 2008-10-24
JP2009531371A (en) 2009-09-03
MA30345B1 (en) 2009-04-01

Similar Documents

Publication Publication Date Title
MX2008012295A (en) Anti-igf-1r human monoclonal antibody formulation.
RU2648152C2 (en) Stable and soluble antibodies inhibiting vegf
US20100158919A1 (en) Pharmaceutical Composition
KR102274714B1 (en) Cyclodextrin and antibody-drug conjugate formulations
US20100260766A1 (en) Stable antibody formulations
JP2020518599A (en) Stable preparation of programmed death receptor 1 (PD-1) antibody and method of using the same
MX2008015852A (en) Lyophilized formulations of anti-egfr antibodies.
KR20180100439A (en) A pharmaceutical composition comprising a bispecific antibody construct
JP7475335B2 (en) CSF-1R antibody preparation
TW201420601A (en) Stable formulations of antibodies to TSLP
TW201036650A (en) Lyophilised antibody formulation
WO2007095337A2 (en) Antibody formulation
TWI761869B (en) Formulation containing anti-cd47 / pd-l1 bispecific antibody and preparation and use thereof
JP2023538669A (en) Pharmaceutical formulations containing BITE, bispecific antibodies, and methionine
CA3064470A1 (en) Pharmaceutical composition comprising c-met antibody-drug conjugate and use thereof
KR20220044286A (en) Formulations comprising anti-PD-1/HER2 bispecific antibodies, methods for their preparation and uses
CN114832102A (en) Stable anti-EGFR antibody composition

Legal Events

Date Code Title Description
FG Grant or registration