WO2013164837A1 - Pharmaceutical formulations of tnf-alpha antibodies - Google Patents

Pharmaceutical formulations of tnf-alpha antibodies Download PDF

Info

Publication number
WO2013164837A1
WO2013164837A1 PCT/IN2013/000129 IN2013000129W WO2013164837A1 WO 2013164837 A1 WO2013164837 A1 WO 2013164837A1 IN 2013000129 W IN2013000129 W IN 2013000129W WO 2013164837 A1 WO2013164837 A1 WO 2013164837A1
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
antibody
buffer
adalimumab
protein
Prior art date
Application number
PCT/IN2013/000129
Other languages
French (fr)
Inventor
Sanjeev Kumar Mendiratta
Sanjay Bandyopadhyay
Chintan G. PATEL
Original Assignee
Cadila Healthcare Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48901138&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2013164837(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to NZ629261A priority Critical patent/NZ629261A/en
Priority to EP21162387.1A priority patent/EP3912639A1/en
Priority to AP2014007922A priority patent/AP2014007922A0/en
Priority to CA2866692A priority patent/CA2866692A1/en
Priority to KR1020167023263A priority patent/KR20160105535A/en
Priority to MX2014010776A priority patent/MX363700B/en
Priority to SG11201405475UA priority patent/SG11201405475UA/en
Priority to KR1020147026991A priority patent/KR101730694B1/en
Priority to JP2014560526A priority patent/JP2015509526A/en
Priority to EP13742519.5A priority patent/EP2822591B1/en
Priority to AU2013255413A priority patent/AU2013255413C1/en
Priority to US14/383,533 priority patent/US10093728B2/en
Priority to EA201491644A priority patent/EA201491644A1/en
Priority to BR112014021325A priority patent/BR112014021325A2/en
Priority to EP18162422.2A priority patent/EP3412310B1/en
Priority to CN201380012986.8A priority patent/CN104159614A/en
Application filed by Cadila Healthcare Limited filed Critical Cadila Healthcare Limited
Publication of WO2013164837A1 publication Critical patent/WO2013164837A1/en
Priority to ZA2014/06414A priority patent/ZA201406414B/en
Priority to PH12014501988A priority patent/PH12014501988B1/en
Priority to HK15101176.7A priority patent/HK1200709A1/en
Priority to US16/123,293 priority patent/US20190071496A1/en
Priority to US17/890,866 priority patent/US20230047111A1/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
    • C07K16/241Tumor Necrosis Factors
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, 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
    • A61K9/14Particulate 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Abstract

The present invention provides certain improved formulations of proteins. Specifically, the present invention provides use of certain excipients that are useful for stabilization of antibody preparations. Additionally, the novel formulation of the present invention prevents the formation of aggregates or fragments or modification of protein in solution.

Description

PHARMACEUTICAL FORMULATIONS
Field of the Invention
The present invention relates to certain improved formulations of proteins. Specifically, the present invention provides use of certain excipients that are useful for stabilization of antibody preparations. Additionally, the novel formulation of the present invention prevents the formation of aggregates or fragments or modification of protein in solution during storage.
Background of the Invention
Proteins are large and complex molecules. They are required to be in their native confirmation in order to remain biologically active and not be immunogenic. Proteins having a high pi value or distinct polarity may not show stability in solution at or around physiological pH conditions. Further, at high concentration, protein molecules in solution are susceptible to undergo aggregation or degradation or certain modifications with time during storage. Most common protein degradation pathways known from literature are protein aggregation, deamidation and oxidation [Cleland et al. Critical Reviews in Therapeutic Drug Carrier Systems 10(4): 307-377 (1993)]. Degradation of protein during storage may take place due to chemical instability (i.e. any process which involves modification of the protein by bond formation or cleavage resulting in a new chemical entity) or physical instability (i.e. changes in the higher order structure of the protein). Chemical instability majorly can be result of deamidation, racemization, hydrolysis, oxidation, beta elimination or disulfide exchange. Physical instability can result from denaturation, aggregation, precipitation or adsorption. In an aspect, the present invention discloses suitable formulation of antibody proteins. Antibodies are highly complex molecules and are the fastest growing class of biologies in the pharmaceutical industry due to their therapeutic effectiveness in humans. However, antibodies are subject to undergo aggregation or degradation or denaturation or chemical modifications resulting in the loss of biological activity during the manufacturing process and / or during storage with time. Such protein modifications can also make them immunogenic resulting in the generation of anti-drug antibodies by the patient which can reduce the drug availability during subsequent injections or worse induce an autoimmune reaction. Therefore, there is a need to have stable new formulations for antibody preparations which protect the molecules from aggregation or degradation or chemical modification during the manufacturing process and / or storage in liquid solution even at high protein concentration while preserving the active biological conformation of the antibody molecules.
Liquid pharmaceutical formulation is a primary choice for manufacturers to prepare a stable, safe and effective pharmaceutical preparation of antibody for therapy. Liquid pharmaceutical preparation is also considered to be easy-to-handle for the patients and by the patients. However, a long appreciated problem with liquid formulations of protein therapeutics is that of aggregation, where protein molecules physically stick together, for example, resulting in the formation of either soluble high molecular weight protein aggregates or insoluble protein aggregates, which may cause undesired immunological reactions in patients upon administration. Additionally, a major problem caused by the aggregate formation is that during the administration the formulation may block syringes or pumps rendering it unsafe to patients. Aggregation of protein can also significantly impact its potency, immunogenicity and stability. Another reason of degradation is that unfolding mediated adsorption at interfaces can often be an initiating step for irreversible aggregation in solution. In this respect, proteins tend to adsorb at liquid-solid, liquid-air, and liquid-liquid interfaces. Sufficient exposure of a protein's core at a hydrophobic surface can result in adsorption as a consequence of agitation, temperature or pH induced stresses. Further, proteins also are sensitive to, for example, pH, ionic strength, thermal stress, shear and interfacial stresses, ail of which can lead to aggregation and result in instability. Another consequence of aggregation is particle formation, an important consideration in liquid and lyophilized protein pharmaceuticals.
WO 2004/016286 discloses a liquid formulation for stabilizing antibodies which treat TNF a mediated diseases, comprising the antibody, a buffer system, mannitol, polysorbates and tonicity agents. The formulation uses a citrate-phosphate buffer system. This formulation could be thawed / frozen at least 3 times without any detrimental effect on either the chemical and physicochemical properties or biological activity.
In order to realize the clinical potency of an antibody protein, there is a need for new and improved formulations comprising the antibody molecule(s) in its native conformation, which can be stored under a desired / suitable condition for long-term storage without formation of significant amount of aggregates or fragments or modified variants of the antibody protein, even at high protein concentration. The present invention addresses the above-identified need by providing novel stable formulations comprising an antibody molecule, preferably a monoclonal antibody, along with suitable excipients which makes the formulation stable and having sufficiently low viscosity at around physiological osmolality and which is therefore suitable for administration to mammals, particularly human subjects.
We herein disclose some such formulations which prevent the formation of aggregates during and after formulation while providing a suitable condition for long term storage.
Embodiments of the Invention
The invention provides improved liquid formulation comprising therapeutic amount of proteins preferably antibodies and suitable excipients.
In certain embodiments the present invention provides a liquid formulation suitable for human use which comprises a therapeutic amount of monoclonal antibody -** or antigen binding portion thereof, which binds to tumor necrosis factor (TNF) and suitable excipients optionally selected from suitable buffers, stabilizer(s), surfactantfs) and suitable additives for maintaining osmolality, optionally with other excipients.
In an embodiment the present invention provides a liquid formulation which comprises of monoclonal antibodies or antigen binding portion thereof along with suitable buffer(s) and other excipients optionally selected from one or more stabilizers, surfactants and tonicity agents such as sodium chloride or potassium chloride. In' an embodiment, such formulations can also optionally be lyophilized. Lyophilization ,can be performed by a skilled person using the techniques available in the art which includes various steps like freezing, annealing, primary drying and secondary drying.
In yet another embodiment the present invention provides a liquid formulation also suitable for lyophilization which comprises from about 1 mg / mL to about 160 mg / mL of monoclonal antibody or antigen binding portion thereof and suitable buffers at a concentration of about 5 mM to 100 mM, optionally suitable stabilizers with a concentration of about 1% to 10%, optionally suitable surfactants at a concentration of about 0.001 % to 1% and optionally suitable tonicity agents at a concentration of about 10 mM to about 150 mM. In an embodiment, the present invention provides a liquid formulation buffered between pH 4 to 8.
In another embodiment, the present invention provides a liquid formulation which can be used for parenteral administration. Parenteral administration includes intravenous, subcutaneous, intra peritoneal, intramuscular administration or any other route of delivery generally considered to be falling under the scope of parenteral administration and as is well known to a skilled person.
In another embodiment, the present invention provides a liquid formulation which improves stability and prevents formation of aggregates of protein in the said formulation. Generally, a stable formulation is the one which retains its physical stability and/or chemical stability and/or biological activity over a period of time upon storage.
In a further embodiment, the present invention provides a liquid formulation which can be used for administering to a patient suffering from a disorder in which TNFa activity is detrimental. Such disorders include sepsis, infections, autoimmune diseases, transplant rejection, malignancy, pulmonary disorders, cardiac disorders, ' intestinal disorders, graft- versus-host disease and the like. Such disorders occur due to TNFa activity and are well described in literature such as in US patent No. 6,09,382. Brief Description of the accompanying drawings
Fig 1 shows the level of aggregates of Adalimumab protein formulated in citrate- phosphate buffer with different stabilizer(s), bulking agent and an isotonicity agent during storage under stressed condition. Formulation compositions are described with Examples 1 to 6.
Fig 2 shows the level of principal charged variant of Adalimumab protein formulated in citrate-phosphate buffer with different stabilizer(s), bulking agent and an isotonicity agent during storage under storage condition. Formulation compositions are described with Examples 1 to 6.
Fig 3 shows the level of aggregates of Adalimumab protein formulated in sodium succinate buffer with different stabilizer(s), bulking agent and an isotonicity agent during storage under stressed condition. Formulation compositions are described with Examples 7 to 13. Fig 4 shows the level of principal charged variant of Adalimumab protein formulated in sodium succinate buffer with different stabilizer(s), bulking agent and an isotonicity agent during storage under stressed condition. Formulation compositions are described with Examples 7 to 13.
Fig 5 shows the level of aggregates of Adalimumab protein formulated in sodium succinate buffer with different stabilizer(s), bulking agent and an isotonicity agent during storage under stressed condition. Formulation compositions are described with Examples 14 to 16.
Fig 6 shows the level of principal charged variant of Adalimumab protein formulated in sodium succinate buffer with different stabilizer(s), bulking agent and an isotonicity agent during storage under stressed condition. Formulation compositions are described with Examples 14 to 16.
Detailed description of the invention
The present invention provides novel and improved liquid formulations which can optionally be lyophilized, comprising of suitable amount of therapeutic protein(s), preferably monoclonal antibodies, in suitable buffer(s), one or more suitable stabilizers, and other excipients which are optionally selected from suitable surfactants and tonicity agents. The said formulation prevents formation of aggregates of protein (antibody) and maintains the potency and stability of the therapeutic compound for up to the desired period of time.
In such embodiment the protein is an antibody or antigen binding portion thereof. In a preferred embodiment the antibody is selected from suitable polyclonal, monoclonal, recombinant antibodies, single chain antibodies, hybrid antibodies, chimeric antibodies, humanized antibodies, or fragments thereof, isolated human antibodies or antibody portions thereof or antibody-like molecules containing one or two binding sites for an antigen and an Fc-part of an immunoglobulin. Adalimumab and Infliximab are examples of antibodies or more suitably, monoclonal antibodies. Infliximab is example of chimeric antibody. Adalimumab is example of human antibody. Etanercept is an example of antibody like molecules. In a preferred embodiment, antibodies used in the formulation are human antibodies. In a more preferred embodiment, the antibodies used in the formulation are human antibodies directed to TNFa including human TNFa. In a still further embodiment, the formulation includes D2E7 and combination of D2E7 with other antibodies. D2E7 antibody which is known by the generic name Adalimumab, with its high affinity binding to hTNFa with low dissociation kinetics and high neutralizing capacity is described in US patent Nos. 6,090,382 and US 6,258,562, as well as several publications such as Ann Rheum Dis 1999;58:(Suppl I) 170-172, Ann Rheum Dis 2000; 59(suppl. Ι):ϊ44— i45 etc. all of which are incorporated herein in their entirety. Adalimumab is available in the market with the brand name HUMIRA®. The names Adalimumab and D2E7 whenever used in the specification represent the same human monoclonal antibody as described in the above references.
In a preferred embodiment, the monoclonal antibody is Adalimumab or antigen binding portion, thereof.
In some embodiments the monoclonal antibodies or antigen binding portion thereof is generally present in a therapeutic amount of up to 160 mg /; mL. In a preferred embodiment the therapeutic amount is about 1 mg / mL to about 100 mg / mL. In a more preferred embodiment the therapeutic amount is about 1 mg / mL to about 50 mg / mL.
The liquid formulation comprises a suitable buffer along with other pharmaceutically acceptable excipients, which stabilizes the pharmaceutical preparation. Suitable buffers which can be used are selected from those which are known in the art and can be found in the literature. In an embodiment the suitable buffers comprise but are not limited to histidine-buffers, citrate-buffers, succinate- buffers, acetate-buffers, phosphate-buffers, phosphate buffered saline, citrate and phosphate buffer, tromethamine buffers and the like or their suitable mixtures.
In a preferred embodiment the suitable buffer comprises of a succinate-buffer or acetate-buffer or histidine buffer. In a still preferred embodiment the suitable buffer comprises a succinate-buffer or an acetate-buffer. In a further preferred embodiment the suitable buffer comprises a succinate-buffer. Succinate buffer can be prepared by dissolving sodium succinate in sterile water or Water for Injection (WFI) or by titrating succinic acid with sodium hydroxide.
The buffers are generally used in concentrations of about ImM to about 100 mM. In a preferred embodiment the buffer concentration is about 5 mM to about 50 mM. In a more preferred embodiment the buffer concentration is about 10 mM to about 20 mM. In a still more preferred embodiment the buffer concentration is about 10 mM.
In an embodiment the liquid formulation maintains a pH value ranging from 4.0 to about 8.0 depending on the monoclonal antibody being used. In a preferred embodiment the buffer used maintains the pH of the formulation in the range of about 5.0 to 5.5. In a more preferred embodiment the pH is maintained to about 5.2.
The liquid formulation further comprises suitable surfactants which are pharmaceutically acceptable excipients used to protect the protein formulations against various stress conditions, like agitation, shearing, exposure to high temperature etc. The suitable surfactants include but are not limited to polyoxyethylensorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers (e.g. Brij), alkylphenylpolyoxyethylene ethers (e.g. Triton-X), polyoxyethylene-polyoxypropylene copolymer (e.g. Poloxamer, Pluronic), sodium dodecyl sulphate (SDS) and the like. In a preferred embodiment the suitable surfactant is polyoxyethylenesorbitan-fatty acid esters (Tweens). In a more preferred embodiment the polyoxyethylenesorbitan-fatty acid esters are polysorbate 20, (sold under the trademark Tween 20™) and polysorbate 80 (sold under the trademark Tween 80™). In another preferred embodiment the suitable surfactant is polyethylene- polypropylene copolymers which are sold under the names Pluronic (R) F68 o Poloxamer 188™. In another preferred embodiment the suitable surfactant is alkylphenolpolyoxyethylene esters which are sold under the trade name Triton-X.
In a preferred embodiment the surfactant is Polysorbate 80 or Polysorbate 20. In a more preferred embodiment the surfactant is Polysorbate 80.
The surfactants are generally used in concentrations of about 0.001 % to about 1 %. In a preferred embodiment surfactant concentration is about 0.01 % to about 1%.
The liquid formulation further comprises one or more suitable stabilizer(s) which are pharmaceutically acceptable excipients, which protect the active pharmaceutical ingredient from chemical and/or physical degradation during manufacturing, storage and application. In an embodiment the stabilizers include but are not limited to suitable sugars, amino acids, polyols, cyclodextrines and the like or suitable derivative or mixtures thereof.
In one such embodiment the sugar is a monosaccharide or an oligosaccharide. Monosaccharide sugars include but are not limited to glucose, fructose, galactose, mannose, sorbose, ribose, deoxyribose and the like or amino sugars, like neuraminic acid. An oligosaccharide includes but is not limited to sucrose, trehalose, lactose, maltose and raffmose and the like or suitable mixtures, thereof.
In another embodiment the polyols which can be used as stabilizers include but are not limited to mannitol, sorbitol, dextran, glycerol, arabitol, propylene glycol, polyethylene glycol and the like or suitable combinations thereof. In a preferred embodiment the suitable polyol is sorbitol.
In another preferred embodiment the stabilizer is a polyol preferably sorbitol. In an embodiment the sorbitol is present in amount about 1% to about 10%.
In another such embodiment the amino acids which can be used as stabilizers include but are not limited to arginine, glycine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophane, methionine, serine, proline, cysteine / cystine and the like or suitable combination of any the above.
In a preferred embodiment the suitable amino acid is arginine or lysine.
In an embodiment the amino acid is present in amount about 0.5% to about
10%.
In another embodiment cyclodextrines or derivative thereof, which can be used as stabilizers, includes but are not limited to a-cyclodextrin, β-cyclodextrih, γ- cyclodextrin, or their hydroxypropylated, hydroxyethylated, ethylated or methylated derivatives thereof or Sulfobutyl ether beta-cyclodextrin (SBE-beta-CD) or a branched cyclodextrins or cyclodextrin polymers or suitable mixture thereof. In a preferred embodiment the suitable cyclodextrin variant is hydroxypropylated cyclo beta-dextrin (ΗΡ-β-CD).
In a preferred embodiment the cyclodextrin or derivative is present in amount about 0.2% to about 10%.
In another embodiment the liquid formulation optionally comprises tonicity agents such as sodium chloride or potassium chloride. In a preferred embodiment, the tonicity agent is sodium chloride which is present in amount about 10 mM to about 150 raM.
The formulation may additionally further comprise one or suitable other excipients which are well known to a person skilled in the art. In some embodiments, the liquid formulation maintains the storage stability in terms of not allowing any further protein aggregation or modifications as compared to time point zero of stability.
In some embodiments, the liquid formulation maintains the stability during the process of formulation.
To estimate the level of aggregates and the principal charged variant of Adalimumab protein, analytical HP-size exclusion chromatography and HP-ion exchange chromatography were performed, respectively.
The said analytical methods used in the present invention are well known to a skilled person and a brief description of the same is provided below merely for the sake of reference only.
HP-Size exclusion chromatography (HP-SEC):
Samples were analyzed to estimate the aggregates by HP-size exclusion chromatography (HP-SEC) using TSK gel G3000 SWXL column (7.8 mm I.D χ 30 cm L). Samples were loaded and eluted isocratically using sodium phosphate buffer at a flow rate of 0.5 mL / min. Elution was monitored at UV 214 nm.
HP-Ion Exchange Chromatography (HP-IEC):
Samples were analyzed to estimate the principal charged variant by HP-IEC using analytical cation exchange column. Samples were loaded and eluted using salt gradient at a flow rate of 1.0 mL / min. Elution was monitored at UV 280 nm.
The present invention is illustrated further in the following examples which are provided for illustration purpose and should not be construed as being a limitation to the scope of the invention.
Examples
The following non-limiting examples describe the different formulations which can be prepared as per the present invention. It will be appreciated that other excipients may be added as are necessary to these formulations and such addition of excipients are within the scope of a person skilled in the art and are to be included within the scope of the present invention.
Screening of excipients is carried out by formulating the Adalimumab protein in different compositions with different excipients and exposing them to higher temperature over the period of time. Formulations of Adalimumab protein were prepared in the presence of different buffering agent(s), stabilizer(s), bulking agent(s) and isotonicity agent(s) as exemplified below.
Example 1
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Dibasic sodium phosphate dihydrate 1.53 mg / mL
Monobasic sodium phosphate dihydrate 0.86 mg / mL
Sodium citrate 0.3 mg / mL
Citric acid 1.3 mg / mL
Mannitol 12 mg / mL
Sodium Chloride 6.16 mg / mL
Polysorbate 80 1 mg / mL Adalimumab protein was purified as per the technique known in the art. In this example, the purified Adalimumab protein was formulated in the presence of citrate- phosphate buffer along with a stabilizer, a bulking agent and an isotonicity agent at a desired concentration as described above. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and the volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 1 and 2. Example 2
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients Dibasic sodium phosphate dihydrate 1.53 mg / mL
Monobasic sodium phosphate dihydrate 0.86 mg / mL
Sodium citrate 0.3 mg / mL
Citric acid 1.3 mg / mL
Mannitol 12 mg / mL
Arginine 5 mg / mL
Sodium Chloride 6.16 mg / mL
Polysorbate 80 1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the same formulation medium as described with Example 1 in the presence of Arginine. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume wasi made up to the desired level with Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 1 and 2.
Example 3
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Dibasic sodium phosphate dihydrate 1.53 mg / mL Monobasic sodium phosphate dihydrate 0.86 mg / mL
Sodium citrate 0.3 mg / mL
Citric acid 1.3 mg / mL
Mannitol 12 mg / mL
Arginine 5 mg / mL
Sorbitol lO mg / mL 6.16 mg / mL
1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the same formulation medium as described in Example 1 with the addition of arginine and sorbitol. pH of the formulation medium is adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container- closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 1 and 2. Example 4
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Dibasic sodium phosphate dihydrate 1.53 mg / mL
Monobasic sodium phosphate dihydrate 0.86 mg / mL
Sodium citrate 0.3 mg / mL
Citric acid 1.3 mg / mL
Mannitol 12 mg / mL
Hydroxypropyl beta cyclodextrin 20 mg / mL
Sodium Chloride 6.16 mg / mL
Polysorbate 80 1 mg / mL Adalimumab protein was prepared as per the technique known in the art and formulated in the same formulation medium as described in Example 1 with the addition of hydroxypropyl beta cyclodextrin. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed mainly for presence of aggregates and HC-LysO content by HP-Size exclusion chromatography and HP-IEC respectively. Obtained results. are shown in Figures 1 and 2.
Example 5
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Dibasic sodium phosphate dihydrate 1.53 mg / mL
Monobasic sodium phosphate dihydrate 0.86 mg / mL
Sodium citrate 0.3 mg / mL
Citric acid 1.3 mg / mL
Mannitol 12 mg / mL
Hydroxypropyl beta cyclodextrin 20 mg / mL
Sorbitol 10 mg / mL
Sodium Chloride 6.16 mg / mL
Polysorbate 80 1 mg / mL Adalimumab protein was prepared as per the technique known in the art and formulated in the same formulation medium as described with Example 1 in the presence of hydroxypropyl beta cyclodextrin and sorbitol. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 1 and 2.
Example 6
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Dibasic sodium phosphate dihydrate 1.53 mg / mL
Monobasic sodium phosphate dihydrate 0.86 mg / mL
Sodium citrate 0.3 mg / mL
Citric acid 1.3 mg / mL
Mannitol 12 mg / mL
Lysine 5 mg / mL
Sodium Chloride 6.16 mg / mL
Polysorbate 80 1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the same formulation medium as described in Example 1 with the addition of lysine. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container- closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 1 and 2. Example 7
Formulation composition
Active Ingredient Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride l lO mM
Sorbitol 50 mg / mL
Arginine 10 mg / mL
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer containing arginine and sorbitol as stabilizers, a bulking agent and an isotonicity agent at a desired concentration as described above in formulation composition. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock: solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4.
Example 8
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride 110 mM
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the absence of stabilizers. The formulation medium comprises a succinate buffer, a bulking agent and an isotonicity agent at a desired concentration as described above. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container- closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4. Example 9
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride l lO mM
Arginine lO mg / mL
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer further comprising arginine as a stabilizer, a bulking agent and an isotonicity agent at a desired concentration as described above. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container- closure systems (like vials, syringes etc.) for storage. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4. Example 10
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride l lO mM
Trehalose 50 mg / mL
Arginine lO mg / mL
Polysorbate 80 O.l mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer along with arginine and trehalose as stabilizers, a bulking agent and an isotonicity agent at a desired concentration as described above in the formulation composition. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4.
Example 11
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride 110 mM 50 mg / mL
10 mg / mL
0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer with arginine and raffinose as stabilizers, a suitable bulking agent and an isotonicity agent at a desired concentration as described above in the formulation composition. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4. Example 12
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride 110 mM
Sorbitol 10 mg / mL
Arginine 5 mg / mL
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer containing arginine and sorbitol as stabilizers at a reduced concentration compared to the earlier Example 7 to reduce the osmolality. A bulking agent and an isotonicity agent were added at a desired concentration as described above. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4.
Example 13
Formulation composition
A ctive Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 10 mM
Sodium chloride 100 mM
Sorbitol lO mg / mL
Arginine 5 mg / mL
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer with arginine and sorbitol as stabilizers, a suitable bulking agent and an isotonicity agent as described above in the formulation composition. The concentration of an isotonicity agent was reduced in comparison to the earlier Example 12 to further reduce the osmolality of the formulation medium. pH of the formulation medium is adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 3 and 4.
Example 14
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride 110 mM
Hydroxypropyl beta cyclodextrin 20 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer further comprising hydroxypropyl beta cyclodextrin as a stabilizer along with a suitable isotonicity agent at a desired concentration as described above in the formulation composition. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 5 and 6.
Example 15
Formulation composition
Active Ingredient
Adalimumab 50 mg / mL
Inactive Ingredients
Sodium succinate 20 mM
Sodium chloride 110 mM Hydroxypropyl beta cyclodextrin 20 mg / mL
Arginine 10 mg / mL
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer with arginine and hydroxypropyl beta cyclodextrin as stabilizers along with a bulking agent and an isotonicity agent at a desired concentration as described above in the formulation composition. pH of the formulation medium is adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 5 and 6.
Example 16
Formulation composition
Active Ingredient
Adalimumab . 50 mg / mL
Inactive Ingredients
Sodium succinate 20 raM
Sodium chloride 110 raM
Hydroxypropyl beta cyclodextrin 20 mg / mL
Polysorbate 80 0.1 mg / mL
Adalimumab protein was prepared as per the technique known in the art and formulated in the presence of succinate buffer containing hydroxypropyl beta cyclodextrin as a stabilizer along with a bulking agent and an isotonicity agent at a desired concentration as described above in the formulation composition. pH of the formulation medium was adjusted to around pH 5.2. Excipients were added to the protein solution from respective stock solutions to adjust the final concentration and volume was made up to the desired level with sterile water or Water for Injection. The formulated bulk was distributed in suitable container-closure systems (like vials, syringes etc.) for storage. Samples were exposed to the higher temperature for checking the degradation over a period of time. Samples at different time points were taken out and analyzed to estimate the aggregates and the principal charged variant of Adalimumab protein by HP-Size exclusion chromatography and HP-IEC, respectively. Results are shown in Figures 5 and 6.
The formulations described with Examples 7 to 16 are also prepared using acetate buffers or histidine buffer. Such formulations are also to be considered as being encompassed by the present invention.
The formulations of the present invention are stable when kept at 2-8 °C.
The formulation of the invention can be used in similar indications as those described in U. S. Patent Nos. 6,090,382 and 6,258,562 each of which is incorporated by reference herein.
The language "effective amount" of the formulation is that amount necessary or sufficient to inhibit TNFot activity, e.g. prevent the various morphological and somatic symptoms of a detrimental TNFot activity-associated state. In one embodiment, an effective amount of the formulation is the amount sufficient to inhibit detrimental TNFot activity. The effective amount can vary depending on such factors as the size and weight of the subject, or the type of illness. One of ordinary skill in the art would be able to study the aforementioned factors and make the determination regarding the effective amount of the TNFot activity inhibiting formulation without undue experimentation.
The formulation of the present invention can be administered to the subject either prior to or after the onset of detrimental TNFot activity. Further, several divided dosages, as well as staggered dosages can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. The dose can be titrated based on the exigencies of the therapeutic or prophylactic situation.
Actual dosage levels of the active ingredients (antibody) in the pharmaceutical formulation of this invention may be varied so as to obtain an amount of the active , ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the antibody found in the formulation, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known to a medical practitioner who can easily prescribe the effective amount of the pharmaceutical composition of the invention.
In general, a suitable daily dose of a formulation of the invention will be that amount of the formulation that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above and well known to a skilled practitioner.
It is to be noted that dosage values may vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
The invention provides a pharmaceutical formulation with an extended shelf life, which, in one embodiment, is used to inhibit TNFa activity in a subject suffering from a disorder in which TNFa activity is detrimental, comprising administering to the subject an antibody or antibody portion of the invention such that TNFa activity in the subject is inhibited. Preferably, the TNFa is human TNFa and the subject is a human subject.

Claims

1. A liquid pharmaceutical formulation comprising an effective amount of an antibody or antigen binding portion thereof directed to TNFa in a buffer system, a surfactant, a tonicity agent and a stabilizer selected from amino acids and cyclodextriries.
2. The formulation as claimed in claim 1, wherein the antibody or antigen binding portion thereof is polyclonal or monoclonal or recombinant antibodies or single chain antibodies or hybrid antibodies or chimeric antibodies or humanized antibodies or fragments thereof or isolated human antibodies or antibody portions thereof.
3. The formulation as claimed in claims 1 & 2, wherein the antibody is Adalimumab or Infliximab.
4. The formulation as claimed in claims 1-3, wherein the antibody is Adalimumab or antigen binding portion thereof.
5. The formulation as claimed in claim 1, wherein the concentration of antibody is between 1 mg / mL to 160 mg / mL preferably 1 mg / mL to 100 mg / mL.
6. The formulation as claimed in claim 5, wherein the concentration of antibody is 50 mg / mL.
7. The formulation as claimed in claim 1, wherein the pH of the formulation is between pH 4 and pH 8.
8. The formulation as claimed in claim 7, wherein the pH of the formulation is between pH 5.0 and pH 5.5, preferably at pH 5.2.
9. The formulation as claimed in claim 1, wherein the buffer system is selected from histidine-buffers, citrate-buffers, succinate-buffers, acetate-buffers, phosphate-buffers, phosphate buffered saline, citrate and phosphate buffer, tromethamine buffers and suitable mixtures thereof.
10. The formulation as claimed in claim 9, wherein the buffer system is a succinate- buffer or acetate-buffer or histidine buffer or suitable mixture thereof.
11. The formulation as claimed in claim 10, wherein the buffer system is a succinate-buffer.
12. The formulation as claimed in claim 1, wherein the buffer is in the concentration of ImM to about 100 mM, preferably 5 mM to 50 mM, more preferably 10 mM to 20 mM.
13. The formulation as claimed in claim 1, wherein the amino acid is selected from arginine, glycine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophane, methionine, serine, proline, cysteine / cystine and suitable combination thereof.
14. The formulation as claimed in claim 13, wherein the stabilizer is arginine alone or in combination with other suitable amino acids.
15. The formulation as claimed in claim 1, wherein the cyclodextrin is selected from a-cyclodextrin, β-cyclodextrin, γ-cyclodextrin, their hydroxypropylated, hydroxyethylated, ethylated and methylated derivatives thereof, Sulfobutyl ether beta-cyclodextrin (SBE-beta-CD), a branched cyclodextrins, cyclodextrin polymers and suitable mixture thereof.
16. The formulation as claimed in claim 16, wherein stabilizer is hydroxypropylated cyclo beta-dextrin (ΗΡ-β-CD).
17. The formulation as claimed in claim 1, comprising additional stabilizers selected from sugars and polyols, including their suitable combination.
18. The formulation as claimed in claim 17, wherein the sugar is selected from glucose, fructose, galactose, mannose, sorbose, ribose, deoxyribose, sucrose, trehalose, lactose, maltose, raffinose and suitable mixtures thereof preferably trehalose or raffinose.
19. The formulation as claimed in claim 18, wherein the polybl is selected from mannitol, sorbitol, dextran, glycerol, arabitol, propylene glycol, polyethylene glycol and suitable combinations thereof.
20. The formulation as claimed in claim 19, wherein the stabilizer is sorbitol.
21. The formulation as claimed in claim 13, wherein the amino acid is present in the concentration of 1% to 10%.
22. The formulation as claimed in 15, wherein the cyclodextrin is in the concentration of 0.2% to 10 %.
23. The formulation as claimed in claim 1 which further comprises suitable excipients selected from surfactants, tonicity agent and suitable combination thereof.
24. The formulation as claimed in claim 23, wherein the surfactant is selected from polyoxyethylensorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers, alkylphenylpolyoxyethylene ethers, polyoxyethylene-polyoxypropylene copolymer and sodium dodecyl sulphate (SDS).
25. The formulation as claimed in claim 24, wherein the surfactant is selected from polysorbate 20 or polysorbate 80.
26. The formulation as claimed in claim 25, wherein the surfactant is 0.001 % to about 1 %.
27. The formulation as claimed in claim 26, wherein the tonicity agent is sodium chloride or potassium chloride.
28. The formulation as claimed in claim 23, wherein the tonicity agent is present in amount about 10 mM to about 150 mM,
29. A formulation as claimed in claim 1 comprising
a) 1 - 160 mg / mL of antibody,
b) 1-10% stabilizer
c) A buffer system with a pH of 4 to 8.
30. The formulation as claimed in claim 29, wherein the stabilizer is selected from sorbitol, raffinose, trehalose, arginine, lysine, hydroxypropylated cyclo beta- dextrin (ΗΡ-β-CD) and combination thereof.
31. The formulation as claimed in claim 29, wherein the buffer system is a succinate or acetate or histidine buffer system.
32. The formulation as claimed in claim 29, wherein the antibody is directed to
TNFa.
33. The formulation as claimed in claim 29, wherein the antibody is Adalimumab or antigen binding portion thereof.
34. The formulation as claimed in claim 29 wherein the antibody concentration is 50 mg / mL.
35. The formulation as claimed in claim 29 which further comprises a surfactant and/or tonicity agent.
36. The formulation as claimed in claim 30, wherein the surfactant is polysorbate 80 and the tonicity agent is sodium chloride.
37. A formulation claimed in claim 1 comprising
a) 50 mg / mL of antibody,
b) 10 mg / mL sorbitol,
c) 5 mg / mL arginine
d) 0.1 mg / mL polysorbate 80
e) 10 mM Succinate buffer of pH of 5.2
38. A lyophilized formulation comprising the composition as claimed in claim 1-37.
39. The formulation of claim 39 wherein the lyophilized powder is reconstituted with water.
40. The formulation as claimed in claim 1 is suitable for parenteral administration.
41. The formulation of claim 1 which is administered to a patient suffering from the disorder in which activity of TNFa is detrimental.
PCT/IN2013/000129 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies WO2013164837A1 (en)

Priority Applications (21)

Application Number Priority Date Filing Date Title
EP21162387.1A EP3912639A1 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies
US14/383,533 US10093728B2 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of TNF-alpha antibodies
AU2013255413A AU2013255413C1 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of TNF-alpha antibodies
CA2866692A CA2866692A1 (en) 2012-03-07 2013-03-05 Formulations which prevent formation of antibody aggregates
KR1020167023263A KR20160105535A (en) 2012-03-07 2013-03-05 Pharmaceutical Formulations of TNF-Alpha Antibodies
MX2014010776A MX363700B (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies.
SG11201405475UA SG11201405475UA (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies
KR1020147026991A KR101730694B1 (en) 2012-03-07 2013-03-05 Pharmaceutical Formulations of TNF-Alpha Antibodies
JP2014560526A JP2015509526A (en) 2012-03-07 2013-03-05 Pharmaceutical formulation
EA201491644A EA201491644A1 (en) 2012-03-07 2013-03-05 PHARMACEUTICAL COMPOSITIONS
AP2014007922A AP2014007922A0 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of TNF-alpha antibodies
NZ629261A NZ629261A (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies
EP13742519.5A EP2822591B1 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies
BR112014021325A BR112014021325A2 (en) 2012-03-07 2013-03-05 LIQUID PHARMACEUTICAL FORMULATION AND LYOPHILIZED FORMULATION
EP18162422.2A EP3412310B1 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies
CN201380012986.8A CN104159614A (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of TNF-alpha antibodies
ZA2014/06414A ZA201406414B (en) 2012-03-07 2014-09-01 Pharamaceuticals formulations of tnf-alpha antibodies
PH12014501988A PH12014501988B1 (en) 2012-03-07 2014-09-05 Pharmaceutical formulations of tnf-alpha antibodies
HK15101176.7A HK1200709A1 (en) 2012-03-07 2015-02-04 Pharmaceutical formulations of tnf-alpha antibodies tnf-
US16/123,293 US20190071496A1 (en) 2012-03-07 2018-09-06 Pharmaceutical formulations of tnf-alpha antibodies
US17/890,866 US20230047111A1 (en) 2012-03-07 2022-08-18 Pharmaceutical formulations of tnf-alpha antibodies

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
IN610MU2012 2012-03-07
IN610/MUM/2012 2012-03-07
IN1606MU2012 2012-05-30
IN1606/MUM/2012 2012-05-30
IN3031/MUM/2012 2012-10-17
IN3031MU2012 2012-10-17

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/383,533 A-371-Of-International US10093728B2 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of TNF-alpha antibodies
US16/123,293 Continuation US20190071496A1 (en) 2012-03-07 2018-09-06 Pharmaceutical formulations of tnf-alpha antibodies

Publications (1)

Publication Number Publication Date
WO2013164837A1 true WO2013164837A1 (en) 2013-11-07

Family

ID=48901138

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2013/000129 WO2013164837A1 (en) 2012-03-07 2013-03-05 Pharmaceutical formulations of tnf-alpha antibodies

Country Status (19)

Country Link
US (3) US10093728B2 (en)
EP (3) EP3412310B1 (en)
JP (3) JP2015509526A (en)
KR (2) KR101730694B1 (en)
CN (1) CN104159614A (en)
AP (1) AP2014007922A0 (en)
AU (1) AU2013255413C1 (en)
BR (1) BR112014021325A2 (en)
CA (1) CA2866692A1 (en)
CL (1) CL2014002316A1 (en)
EA (1) EA201491644A1 (en)
HK (1) HK1200709A1 (en)
MX (2) MX363700B (en)
NZ (1) NZ629261A (en)
PH (1) PH12014501988B1 (en)
SG (2) SG10201609982PA (en)
TR (1) TR201810815T4 (en)
WO (1) WO2013164837A1 (en)
ZA (1) ZA201406414B (en)

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140086930A1 (en) * 2002-08-16 2014-03-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating tnf-alpha associated disorders
EP2863951A1 (en) * 2012-06-12 2015-04-29 Boehringer Ingelheim International GmbH Pharmaceutical formulation for a therapeutic antibody
WO2015090162A1 (en) * 2013-12-16 2015-06-25 浙江海正药业股份有限公司 Pharmaceutical composition comprising adalimumab
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
WO2015151115A1 (en) * 2014-04-02 2015-10-08 Intas Pharmaceuticals Limited Liquid pharmaceutical composition of adalimumab
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
EP2946767A1 (en) * 2014-05-23 2015-11-25 Ares Trading S.A. Liquid pharmaceutical composition
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2016066688A1 (en) * 2014-10-28 2016-05-06 Richter Gedeon Nyrt. Pharmaceutical anti-tnf-alpha antibody formulation
US9340612B2 (en) 2001-03-19 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
WO2016089946A1 (en) * 2014-12-02 2016-06-09 Biogen Ma Inc. Methods and compositions for controlling antibody aggregation
EP3053572A1 (en) * 2015-02-06 2016-08-10 Ares Trading S.A. Liquid pharmaceutical composition
WO2016162819A1 (en) 2015-04-07 2016-10-13 Lupin Limited Stable aqueous pharmaceutical composition of anti-tnf alpha antibody
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
CN106456538A (en) * 2014-05-23 2017-02-22 阿雷斯贸易股份有限公司 Liquid pharmaceutical composition
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
JP2017516847A (en) * 2014-05-23 2017-06-22 アレス トレーディング ソシエテ アノニム Liquid pharmaceutical composition
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
WO2017117311A1 (en) * 2015-12-30 2017-07-06 Genentech, Inc. Formulations with reduced degradation of polysorbate
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
WO2018004260A1 (en) * 2016-06-30 2018-01-04 (주)셀트리온 Stable liquid pharmaceutical preparation
EP3372241A1 (en) * 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
EP3383435A4 (en) * 2015-11-30 2019-07-10 Medimmune, LLC Optimized ratios of amino acids and sugars as amorphous stabilizing compounds in pharmaceutical compositions containing high concentrations of protein-based therapeutic agents
CN110536698A (en) * 2016-12-30 2019-12-03 拜奥卡德联合股份公司 The aqueous pharmaceutical composition of recombinant monoclonal anti-TNF alpha antibodies
EP3563867A4 (en) * 2016-12-30 2020-12-16 Joint Stock Company "Biocad" Aqueous pharmaceutical composition of a recombinant monoclonal antibody to tnf alfa
US10940185B2 (en) 2016-12-28 2021-03-09 Jcr Pharmaceuticals Co., Ltd. Lyophilized preparation
US11021543B2 (en) 2015-06-24 2021-06-01 Janssen Biotech, Inc. Immune modulation and treatment of solid tumors with antibodies that specifically bind CD38
US11071782B2 (en) 2016-04-20 2021-07-27 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11236146B2 (en) * 2016-10-28 2022-02-01 Celltrion Inc. Stable pharmaceutical formulation
WO2022111547A1 (en) * 2020-11-24 2022-06-02 The University Of Hong Kong Inhaled powder formulations for respiratory delivery of antibodies
US11534402B2 (en) 2017-03-06 2022-12-27 Arecor Limited Liquid pharmaceutical composition
US11566079B2 (en) 2015-11-03 2023-01-31 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11608357B2 (en) 2018-08-28 2023-03-21 Arecor Limited Stabilized antibody protein solutions
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US11713355B2 (en) 2014-02-28 2023-08-01 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG10201609982PA (en) * 2012-03-07 2017-01-27 Cadila Healthcare Ltd Pharmaceutical formulations of tnf-alpha anitbodies
UA117466C2 (en) * 2012-12-13 2018-08-10 Мерк Шарп Енд Доме Корп. SOLUTION FORMULATIONS OF ENGINEERED ANTI-IL-23p19 ANTIBODIES
US9821059B2 (en) * 2014-10-17 2017-11-21 Alteogen Inc. Composition for stabilizing protein and pharmaceutical formulation comprising the same
JP6738608B2 (en) * 2016-01-22 2020-08-12 田中貴金属工業株式会社 Chromatographic media
GB201604124D0 (en) * 2016-03-10 2016-04-27 Ucb Biopharma Sprl Pharmaceutical formulation
FR3054444A1 (en) * 2016-07-29 2018-02-02 Laboratoire Francais Du Fractionnement Et Des Biotechnologies ORAL COMPOSITION OF ANTI-TNF ALPHA ANTIBODY
WO2018063963A1 (en) * 2016-09-28 2018-04-05 Board Of Regents, The University Of Texas System Antibody and protein therapeutic formulations and uses thereof
WO2018131893A1 (en) 2017-01-11 2018-07-19 ㈜셀트리온 Stable liquid formula
BR112019019162A2 (en) * 2017-03-16 2020-04-14 Lg Chemical Ltd liquid formulation of an anti-tnf-a antibody and method for preparing the liquid formulation
CN108686205B (en) * 2017-04-07 2021-12-10 海正生物制药有限公司 Lyophilized preparation of infliximab
CN108686204A (en) * 2017-04-07 2018-10-23 浙江海正药业股份有限公司 Include the infliximab composition of histidine buffer system
WO2018204374A1 (en) 2017-05-02 2018-11-08 Merck Sharp & Dohme Corp. Formulations of anti-lag3 antibodies and co-formulations of anti-lag3 antibodies and anti-pd-1 antibodies
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
CA3071930A1 (en) * 2017-08-08 2019-02-14 Csl Behring Ag Hemopexin formulations
CN110151988A (en) * 2018-02-11 2019-08-23 百奥泰生物制药股份有限公司 A kind of human antibody preparation of targeted therapy TNF-α related disease
CN110302377B (en) * 2018-02-11 2020-04-17 百奥泰生物制药股份有限公司 Human antibody preparation for targeted therapy of TNF- α related diseases
AU2020225202B2 (en) 2019-02-18 2023-10-26 Eli Lilly And Company Therapeutic antibody formulation
US20220042977A1 (en) * 2020-08-04 2022-02-10 ProStabilis, Inc. Protein Solubility Screening Kits and Their Use
KR20230054395A (en) * 2020-08-20 2023-04-24 모멘타 파머슈티컬스 인코포레이티드 sialylated glycoproteins
AU2022348349A1 (en) * 2021-09-16 2024-05-02 Aprogen Inc. PHARMACEUTICAL COMPOSITION CONTAINING ANTI-TNFα ANTIBODY

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004016286A2 (en) * 2002-08-16 2004-02-26 Abbott Biotechnology Ltd. Pharmaceutical anti-tnf-alpha antibody formulation
WO2010077422A2 (en) * 2008-10-29 2010-07-08 Wyeth Llc Formulations of single domain antigen binding molecules

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US609382A (en) 1898-08-16 Cotton-chopper
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
CN103275221B (en) 1996-02-09 2016-08-17 艾伯维生物技术有限公司 People's antibody in conjunction with human TNF alpha
EP0852951A1 (en) * 1996-11-19 1998-07-15 Roche Diagnostics GmbH Stable lyophilized monoclonal or polyclonal antibodies containing pharmaceuticals
DE60235013D1 (en) * 2001-07-25 2010-02-25 Facet Biotech Corp STABLE LYOPHILIZED PHARMACEUTICAL FORMULATION OF THE IGG ANTIBODY DACLIZUMAB
JP4583762B2 (en) * 2002-02-27 2010-11-17 イミュネックス・コーポレーション Polypeptide preparation
AU2003293543A1 (en) 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
EP1988922A4 (en) * 2006-02-03 2010-06-02 Medimmune Llc Protein formulations
EP2043711A4 (en) * 2006-06-30 2017-08-30 AbbVie Biotechnology Ltd Automatic injection device
RU2009141592A (en) * 2007-04-11 2011-05-20 Алькон Рисерч, Лтд. (Us) USE OF TNFα INHIBITOR IN COMBINATION WITH ANTIHISTAMINE TREATMENT FOR TREATMENT OF ALLERGIC RHINITIS AND ALLERGIC CONJUNCTIVITIS
PE20091174A1 (en) 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd LIQUID FORMULATION WITH HIGH CONCENTRATION OF ANTIBODY CONTENT
CA2742988A1 (en) * 2008-11-17 2010-05-20 Genentech, Inc. Method and formulation for reducing aggregation of a macromolecule under physiological conditions
CN102458469B (en) 2009-05-04 2014-12-24 艾伯维生物技术有限公司 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
EP3409289B1 (en) 2010-02-26 2020-09-30 Novo Nordisk A/S Stable antibody containing compositions
HRP20220405T1 (en) * 2010-08-19 2022-05-27 Zoetis Belgium S.A. Anti-ngf antibodies and their use
EP2471554A1 (en) 2010-12-28 2012-07-04 Hexal AG Pharmaceutical formulation comprising a biopharmaceutical drug
GB201112429D0 (en) 2011-07-19 2011-08-31 Glaxo Group Ltd Antigen-binding proteins with increased FcRn binding
SG10201609982PA (en) * 2012-03-07 2017-01-27 Cadila Healthcare Ltd Pharmaceutical formulations of tnf-alpha anitbodies
EP2863951A1 (en) * 2012-06-12 2015-04-29 Boehringer Ingelheim International GmbH Pharmaceutical formulation for a therapeutic antibody
ES2784861T3 (en) 2012-09-07 2020-10-01 Coherus Biosciences Inc Stable aqueous formulations of adalimumab

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004016286A2 (en) * 2002-08-16 2004-02-26 Abbott Biotechnology Ltd. Pharmaceutical anti-tnf-alpha antibody formulation
WO2010077422A2 (en) * 2008-10-29 2010-07-08 Wyeth Llc Formulations of single domain antigen binding molecules

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MAITY HARIPADA ET AL: "Effects of arginine on photostability and thermal stability of IgG1 monoclonal antibodies", CURRENT PHARMACEUTICAL BIOTECHNOLOGY, BENTHAM SCIENCE PUBLISHERS, NL, vol. 10, no. 8, 1 December 2009 (2009-12-01), pages 761 - 766, XP009148288, ISSN: 1389-2010 *
MARK CORNELL MANNING ET AL: "Stability of Protein Pharmaceuticals: An Update", PHARMACEUTICAL RESEARCH, KLUWER ACADEMIC PUBLISHERS-PLENUM PUBLISHERS, NL, vol. 27, no. 4, 9 February 2010 (2010-02-09), pages 544 - 575, XP019793935, ISSN: 1573-904X, DOI: 10.1007/S11095-009-0045-6 *
ROBERT J FALCONER ET AL: "Stabilization of a monoclonal antibody during purification and formulation by addition of basic amino acid excipients", JOURNAL OF CHEMICAL TECHNOLOGY & BIOTECHNOLOGY, vol. 86, no. 7, 7 July 2011 (2011-07-07), pages 942 - 948, XP055076799, ISSN: 0268-2575, DOI: 10.1002/jctb.2657 *
TIM J KAMERZELL ET AL: "Protein excipient interactions: Mechanisms and biophysical characterization applied to protein formulation development", ADVANCED DRUG DELIVERY REVIEWS, ELSEVIER, vol. 63, no. 13, 26 July 2011 (2011-07-26), pages 1118 - 1159, XP028320148, ISSN: 0169-409X, [retrieved on 20110729], DOI: 10.1016/J.ADDR.2011.07.006 *

Cited By (167)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9340612B2 (en) 2001-03-19 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9738714B2 (en) 2002-08-16 2017-08-22 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8795670B2 (en) * 2002-08-16 2014-08-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US20140127222A1 (en) * 2002-08-16 2014-05-08 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating tnf-alpha associated disorders
US9750808B2 (en) 2002-08-16 2017-09-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8802101B2 (en) * 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8802100B2 (en) * 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8802102B2 (en) * 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8911741B2 (en) 2002-08-16 2014-12-16 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8916157B2 (en) 2002-08-16 2014-12-23 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8916158B2 (en) 2002-08-16 2014-12-23 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8932591B2 (en) 2002-08-16 2015-01-13 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8940305B2 (en) 2002-08-16 2015-01-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US20140086929A1 (en) * 2002-08-16 2014-03-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating tnf-alpha associated disorders
US9327032B2 (en) 2002-08-16 2016-05-03 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9302011B2 (en) 2002-08-16 2016-04-05 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-α associated disorders
US9732152B2 (en) 2002-08-16 2017-08-15 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9295725B2 (en) 2002-08-16 2016-03-29 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
US9289497B2 (en) 2002-08-16 2016-03-22 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US20140086930A1 (en) * 2002-08-16 2014-03-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating tnf-alpha associated disorders
US9272041B2 (en) 2002-08-16 2016-03-01 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US20140086931A1 (en) * 2002-08-16 2014-03-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating tnf-alpha associated disorders
US9114166B2 (en) 2002-08-16 2015-08-25 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9220781B2 (en) 2002-08-16 2015-12-29 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9272042B2 (en) 2002-08-16 2016-03-01 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9365645B1 (en) 2011-04-27 2016-06-14 Abbvie, Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9359434B2 (en) 2012-04-20 2016-06-07 Abbvie, Inc. Cell culture methods to reduce acidic species
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9346879B2 (en) 2012-04-20 2016-05-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
EP2863951A1 (en) * 2012-06-12 2015-04-29 Boehringer Ingelheim International GmbH Pharmaceutical formulation for a therapeutic antibody
JP2015519382A (en) * 2012-06-12 2015-07-09 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Pharmaceutical formulations for therapeutic antibodies
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9290568B2 (en) 2012-09-02 2016-03-22 Abbvie, Inc. Methods to control protein heterogeneity
US9789185B2 (en) 2012-09-07 2017-10-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10207000B2 (en) 2012-09-07 2019-02-19 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10799585B2 (en) 2012-09-07 2020-10-13 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10786566B2 (en) 2012-09-07 2020-09-29 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10780163B2 (en) 2012-09-07 2020-09-22 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10772959B2 (en) 2012-09-07 2020-09-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10772960B2 (en) 2012-09-07 2020-09-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9782480B2 (en) 2012-09-07 2017-10-10 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10722579B2 (en) 2012-09-07 2020-07-28 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716854B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9346880B2 (en) 2012-09-07 2016-05-24 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716852B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716853B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10688183B2 (en) 2012-09-07 2020-06-23 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9770507B2 (en) 2012-09-07 2017-09-26 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10286071B2 (en) 2012-09-07 2019-05-14 Coherus Biosciences, Inc. Syringe containing stable aqueous formulations of adalimumab
US9340611B2 (en) 2012-09-07 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9682145B2 (en) 2012-09-07 2017-06-20 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10286072B2 (en) 2012-09-07 2019-05-14 Coherus Biosciences, Inc. Methods of manufacturing stable aqueous formulations of adalimumab
US9782479B2 (en) 2012-09-07 2017-10-10 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10195275B2 (en) 2012-09-07 2019-02-05 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10159733B2 (en) 2012-09-07 2018-12-25 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9707293B2 (en) 2012-09-07 2017-07-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9757454B2 (en) 2012-09-07 2017-09-12 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10159732B2 (en) 2012-09-07 2018-12-25 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9724414B2 (en) 2012-09-07 2017-08-08 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9724415B2 (en) 2012-09-07 2017-08-08 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9731009B2 (en) 2012-09-07 2017-08-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9731008B2 (en) 2012-09-07 2017-08-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10155039B2 (en) 2012-09-07 2018-12-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9737600B2 (en) 2012-09-07 2017-08-22 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9382317B2 (en) 2012-09-07 2016-07-05 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9861695B2 (en) 2012-09-07 2018-01-09 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9808525B2 (en) 2012-09-07 2017-11-07 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9200069B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
RU2664736C2 (en) * 2013-12-16 2018-08-22 Чжэцзян Хисунь Фармасьютикал Ко., Лтд Pharmaceutical composition containing adalimumab
WO2015090162A1 (en) * 2013-12-16 2015-06-25 浙江海正药业股份有限公司 Pharmaceutical composition comprising adalimumab
US11713355B2 (en) 2014-02-28 2023-08-01 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
WO2015151115A1 (en) * 2014-04-02 2015-10-08 Intas Pharmaceuticals Limited Liquid pharmaceutical composition of adalimumab
US10688187B2 (en) 2014-04-02 2020-06-23 Intas Pharmaceuticals Ltd. Liquid pharmaceutical composition of adalimumab
US10772961B2 (en) 2014-05-23 2020-09-15 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
CN106456538A (en) * 2014-05-23 2017-02-22 阿雷斯贸易股份有限公司 Liquid pharmaceutical composition
US11752208B2 (en) 2014-05-23 2023-09-12 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11752209B2 (en) 2014-05-23 2023-09-12 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
EP3145487B1 (en) 2014-05-23 2018-08-22 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
JP2021107399A (en) * 2014-05-23 2021-07-29 フレゼニウス カビ ドイチュラント ゲゼルシャフト ミット ベシュレンクテル ハフツング Liquid pharmaceutical composition
EP3939566A1 (en) * 2014-05-23 2022-01-19 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
EP3403646B1 (en) 2014-05-23 2022-09-28 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
CN109248315A (en) * 2014-05-23 2019-01-22 费森尤斯卡比德国有限公司 Composition of liquid medicine
EP3148510B1 (en) 2014-05-23 2018-06-27 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
EP3741358A1 (en) * 2014-05-23 2020-11-25 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
JP2017516848A (en) * 2014-05-23 2017-06-22 アレス トレーディング ソシエテ アノニム Liquid pharmaceutical composition
EP3476386A1 (en) * 2014-05-23 2019-05-01 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
JP2017516847A (en) * 2014-05-23 2017-06-22 アレス トレーディング ソシエテ アノニム Liquid pharmaceutical composition
CN106535935A (en) * 2014-05-23 2017-03-22 阿雷斯贸易股份有限公司 Liquid pharmaceutical composition
CN111939257A (en) * 2014-05-23 2020-11-17 费森尤斯卡比德国有限公司 Liquid pharmaceutical composition
US10426832B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10426833B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10493152B2 (en) 2014-05-23 2019-12-03 Fresenius Kabi Deutschland Gmbh Adalimumab formulations
US11712471B2 (en) 2014-05-23 2023-08-01 Fresenius Kabi Deustschland GmbH Liquid pharmaceutical composition
AU2020286276B2 (en) * 2014-05-23 2023-06-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
EP2946767A1 (en) * 2014-05-23 2015-11-25 Ares Trading S.A. Liquid pharmaceutical composition
JP2020033361A (en) * 2014-05-23 2020-03-05 フレゼニウス カビ ドイチュラント ゲゼルシャフト ミット ベシュレンクテル ハフツング Liquid pharmaceutical composition
JP2022003091A (en) * 2014-05-23 2022-01-11 フレゼニウス カビ ドイチュラント ゲゼルシャフト ミット ベシュレンクテル ハフツング Liquid pharmaceutical composition
US10729769B2 (en) 2014-05-23 2020-08-04 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11707524B2 (en) 2014-05-23 2023-07-25 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
WO2015177059A1 (en) * 2014-05-23 2015-11-26 Ares Trading S.A. Liquid pharmaceutical composition
DE202015009619U1 (en) 2014-10-28 2018-08-29 Richter Gedeon Nyrt. Pharmaceutical anti-TNFα antibody formulation
EP3428189A1 (en) 2014-10-28 2019-01-16 Richter Gedeon Nyrt. Pharmaceutical anti-tnf-alpha antibody formulation
DE202015009016U1 (en) 2014-10-28 2016-06-24 Gedeon Nyrt. Richter Pharmaceutical anti-TNFα antibody formulation
DE202015009509U9 (en) 2014-10-28 2018-05-30 Gedeon Nyrt. Richter Pharmaceutical anti-TNFα antibody formulation
WO2016066688A1 (en) * 2014-10-28 2016-05-06 Richter Gedeon Nyrt. Pharmaceutical anti-tnf-alpha antibody formulation
US20220016244A1 (en) * 2014-10-28 2022-01-20 Richter Gedeon Nyrt. Pharmaceutical anti-tnf-alpha antibody formulation
DE202015009509U1 (en) 2014-10-28 2018-01-18 Gedeon Nyrt. Richter Pharmaceutical anti-TNFα antibody formulation
US20170252437A1 (en) * 2014-10-28 2017-09-07 Richter Gedeon Nyrt. Pharmaceutical anti-tnf-alpha antibody formulation
WO2016089946A1 (en) * 2014-12-02 2016-06-09 Biogen Ma Inc. Methods and compositions for controlling antibody aggregation
EP3053573A1 (en) * 2015-02-06 2016-08-10 Ares Trading S.A. Liquid pharmaceutical composition
WO2016124588A1 (en) * 2015-02-06 2016-08-11 Ares Trading S.A. Liquid pharmaceutical composition
EP3053572A1 (en) * 2015-02-06 2016-08-10 Ares Trading S.A. Liquid pharmaceutical composition
WO2016162819A1 (en) 2015-04-07 2016-10-13 Lupin Limited Stable aqueous pharmaceutical composition of anti-tnf alpha antibody
US11021543B2 (en) 2015-06-24 2021-06-01 Janssen Biotech, Inc. Immune modulation and treatment of solid tumors with antibodies that specifically bind CD38
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11566079B2 (en) 2015-11-03 2023-01-31 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11708419B2 (en) 2015-11-03 2023-07-25 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11732051B2 (en) 2015-11-03 2023-08-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11708420B2 (en) 2015-11-03 2023-07-25 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
EP3383435A4 (en) * 2015-11-30 2019-07-10 Medimmune, LLC Optimized ratios of amino acids and sugars as amorphous stabilizing compounds in pharmaceutical compositions containing high concentrations of protein-based therapeutic agents
US10525137B2 (en) 2015-12-30 2020-01-07 Genentech, Inc. Formulations with reduced degradation of polysorbate
WO2017117311A1 (en) * 2015-12-30 2017-07-06 Genentech, Inc. Formulations with reduced degradation of polysorbate
US20200215195A1 (en) * 2015-12-30 2020-07-09 Genentech, Inc. Formulations with reduced degradation of polysorbate
IL259646B2 (en) * 2015-12-30 2023-06-01 Genentech Inc Formulations with reduced degradation of polysorbate
AU2016380988B2 (en) * 2015-12-30 2022-07-21 Genentech, Inc. Formulations with reduced degradation of polysorbate
US10933141B2 (en) 2015-12-30 2021-03-02 Genentech, Inc. Formulations with reduced degradation of polysorbate
US11576971B2 (en) 2016-04-20 2023-02-14 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11071782B2 (en) 2016-04-20 2021-07-27 Coherus Biosciences, Inc. Method of filling a container with no headspace
AU2017287743B2 (en) * 2016-06-30 2020-01-30 Celltrion Inc. Stable liquid pharmaceutical preparation
CN109310628A (en) * 2016-06-30 2019-02-05 赛特瑞恩股份有限公司 Stable liquid pharmaceutical formulation
US11951207B2 (en) 2016-06-30 2024-04-09 Celltrion Inc. Stable liquid pharmaceutical preparation
EP4338752A3 (en) * 2016-06-30 2024-04-03 Celltrion, Inc. Stable liquid pharmaceutical preparation
AU2020201249B2 (en) * 2016-06-30 2021-07-15 Celltrion, Inc. Stable liquid pharmaceutical preparation
WO2018004260A1 (en) * 2016-06-30 2018-01-04 (주)셀트리온 Stable liquid pharmaceutical preparation
IL263630B2 (en) * 2016-06-30 2023-06-01 Celltrion Inc Stable liquid pharmaceutical preparation
AU2017287743C1 (en) * 2016-06-30 2020-10-01 Celltrion Inc. Stable liquid pharmaceutical preparation
US11236146B2 (en) * 2016-10-28 2022-02-01 Celltrion Inc. Stable pharmaceutical formulation
US10940185B2 (en) 2016-12-28 2021-03-09 Jcr Pharmaceuticals Co., Ltd. Lyophilized preparation
CN110536698A (en) * 2016-12-30 2019-12-03 拜奥卡德联合股份公司 The aqueous pharmaceutical composition of recombinant monoclonal anti-TNF alpha antibodies
CN110536698B (en) * 2016-12-30 2024-02-13 拜奥卡德联合股份公司 Aqueous pharmaceutical compositions of recombinant monoclonal anti-TNFα antibodies
EP3563867A4 (en) * 2016-12-30 2020-12-16 Joint Stock Company "Biocad" Aqueous pharmaceutical composition of a recombinant monoclonal antibody to tnf alfa
US11534402B2 (en) 2017-03-06 2022-12-27 Arecor Limited Liquid pharmaceutical composition
WO2018162503A1 (en) * 2017-03-06 2018-09-13 Ares Trading S.A. Liquid pharmaceutical composition
EP3372241A1 (en) * 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
US11534403B2 (en) 2017-03-06 2022-12-27 Arecor Limited Liquid pharmaceutical composition
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US11608357B2 (en) 2018-08-28 2023-03-21 Arecor Limited Stabilized antibody protein solutions
WO2022111547A1 (en) * 2020-11-24 2022-06-02 The University Of Hong Kong Inhaled powder formulations for respiratory delivery of antibodies

Also Published As

Publication number Publication date
JP2015509526A (en) 2015-03-30
PH12014501988A1 (en) 2014-11-24
ZA201406414B (en) 2016-08-31
EP2822591B1 (en) 2018-05-02
EA201491644A1 (en) 2014-12-30
SG10201609982PA (en) 2017-01-27
JP2018162295A (en) 2018-10-18
EP3412310B1 (en) 2022-09-07
BR112014021325A2 (en) 2017-08-22
EP3912639A1 (en) 2021-11-24
KR20160105535A (en) 2016-09-06
MX363700B (en) 2019-03-29
HK1200709A1 (en) 2015-08-14
MX2019003716A (en) 2019-09-04
EP2822591A1 (en) 2015-01-14
EP3412310A1 (en) 2018-12-12
US20190071496A1 (en) 2019-03-07
PH12014501988B1 (en) 2014-11-24
NZ629261A (en) 2017-08-25
JP2017019855A (en) 2017-01-26
MX2014010776A (en) 2014-10-14
US10093728B2 (en) 2018-10-09
KR101730694B1 (en) 2017-04-27
SG11201405475UA (en) 2014-10-30
AU2013255413C1 (en) 2016-03-24
CL2014002316A1 (en) 2015-02-27
KR20140134689A (en) 2014-11-24
AP2014007922A0 (en) 2014-09-30
JP6416841B2 (en) 2018-10-31
CA2866692A1 (en) 2013-07-11
US20230047111A1 (en) 2023-02-16
CN104159614A (en) 2014-11-19
TR201810815T4 (en) 2018-08-27
AU2013255413B2 (en) 2015-11-05
US20150071936A1 (en) 2015-03-12
AU2013255413A1 (en) 2014-09-25

Similar Documents

Publication Publication Date Title
US20230047111A1 (en) Pharmaceutical formulations of tnf-alpha antibodies
US20240058263A1 (en) Formulations of antibody
TWI738632B (en) Stable protein solution formulation containing high concentration of an anti-vegf antibody
TWI548424B (en) Stable liquid formulation of etanercept
US20120128687A1 (en) Novel antibody formulation
TW201424748A (en) High concentration antibody and protein formulations
JP7208302B2 (en) Pharmaceutical composition containing anti-human TSLP receptor antibody
JP7089121B2 (en) Protein solution preparation containing high concentration of anti-VEGF antibody
JP6445169B2 (en) Stable benzyl alcohol-free aqueous solution containing α-type interferon
OA17126A (en) Pharmaceutical formulations of TNF-alpha antibodies
JP2023553106A (en) Improved lyophilized formulation
CN114652825A (en) Stable antibody preparation, preparation method and application thereof
CN117838854A (en) Stabilizer composition of antibody medicine and pharmaceutical composition
TW201825109A (en) Stable pharmaceutical formulation

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2013742519

Country of ref document: EP

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13742519

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 234395

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: P935/2014

Country of ref document: AE

WWE Wipo information: entry into national phase

Ref document number: A201409733

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 2014560526

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14383533

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2866692

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/010776

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 20147026991

Country of ref document: KR

Kind code of ref document: A

Ref document number: 2013255413

Country of ref document: AU

Date of ref document: 20130305

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201491644

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014021325

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014021325

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140828