LYOPHILIZED ANTIBODY FORMULATION

Information

  • Patent Application
  • 20220273796
  • Publication Number
    20220273796
  • Date Filed
    July 16, 2020
    3 years ago
  • Date Published
    September 01, 2022
    a year ago
Abstract
The present invention relates to a stable pharmaceutical formulation comprising an antibody; preferably the pharmaceutical formulation is lyophilized.
Description
TECHNICAL FIELD

The present invention relates to a stable pharmaceutical formulation comprising an antibody; preferably the pharmaceutical formulation is lyophilized.


BACKGROUND

In the context of pharmaceutical product development, a pharmaceutical formulation refers to a mixture of an active pharmaceutical ingredient(s) and inactive ingredients, also known as excipients that are combined to produce a final drug product.


While the active pharmaceutical ingredient (API) has a pharmacological activity that contributes to the diagnosis, treatment, or prevention of a certain disease(s), the inactive ingredients have the role of contributing to product stability, biopharmaceutical profile, appearance and to patient acceptability (P. Furrer, European Pharmaceutical Review, Issue 2, 2013, Apr. 18, 2013). In particular the excipients, according to the International Pharmaceutical Excipients Council (IPEC, 1995) “are all substances contained in a dosage form other than the active substance or finished dosage form, which have been appropriately evaluated for safety and are included in a drug delivery system to either aid the processing of the drug delivery system during its manufacture, protect, support, enhance stability, bioavailability, or patient acceptability, assist in product identification, or enhance any other attributes of the overall safety and effectiveness of the drug delivery system during storage or use”. Excipients therefore include solvents, diluents, buffering agents, pH-adjusting agents, surfactants, preservatives, tonicity agents, stabilizing agents etc.


When developing a drug product, the choice of the pharmaceutical formulation (API(s) concentration, excipients, excipients concentration) depends on the properties of the API(s), on the drug product presentation, on the route of administration of the drug product and on the administration dosage. Among the factors driving such choice, there is the stability of the final drug product, for which integrity of the API and safety of the formulation for the patients must be assured.


For the production of therapeutic antibodies, the development of stable pharmaceutical formulations is necessary to obtain an effective and stable drug products, therefore optimizing antibody pharmaceutical formulations, and in parallel performing exhaustive stability tests, is a primary need of the pharmaceutical industry.







SUMMARY

The present invention relates to a stable pharmaceutical formulation comprising an anti-OX40 antagonist antibody or antibody fragment thereof, one or more stabilizing or tonicity agent and a buffer, wherein said pharmaceutical formulation is liquid or lyophilized.


In one aspect, the pharmaceutical formulation of the present invention comprises a buffer selected from the group comprising acetate, L-histidine, citrate and phosphate and present within said pharmaceutical formulation at concentration between about 1 mM and about 10 mM.


In another aspect, the pharmaceutical formulation of the present invention comprises one or more stabilizing or tonicity agent selected from the group comprising sodium acetate, sodium bicarbonate, sodium carbonate, sodium chloride, potassium acetate, potassium bicarbonate, potassium carbonate, potassium chloride, calcium chloride, sucrose, glutamate, mannitol, polyols, Polysorbate 20, Polysorbate 40, Polysorbate 80, Poloxamer, Poloxamer 188, Poloxamer 407, amino acids such as histidine, arginine, glycine, methionine, proline, lysine, glutamic acid, amines, cyclodextrins, β-cyclodextrins, polyvinylpyrrolidone, polyethylene glycol 400, sorbitol, trehalose and EDTA, and present within said pharmaceutical formulation at a percentage between about 0.001% and about 10%.


In a particular embodiment, the pharmaceutical formulation of the present invention comprises an anti-OX40 antagonist antibody or fragment thereof present within said pharmaceutical formulation at a concentration between about 20 mg/mL and about 80 mg/mL, L-histidine buffer present within said pharmaceutical formulation at a concentration between about 1 mM and 10 mM, sucrose present within said pharmaceutical formulation at a percentage between about 1% and about 10% and Polysorbate 80 present within said pharmaceutical formulation at a percentage between about 0.001% and about 0.1%, and said pharmaceutical formulation has pH between about 5.0 and about 7.0.


In another particular embodiment, the pharmaceutical formulation of the present invention comprises an anti-OX40 antagonist antibody or fragment thereof present within said pharmaceutical formulation at a concentration between about 40 mg/mL and about 60 mg/mL, L-histidine buffer present within said pharmaceutical formulation at a concentration between about 3 mM and 7 mM, sucrose present within said pharmaceutical formulation at a percentage between about 2% and about 6% and Polysorbate 80 present within said pharmaceutical formulation at a percentage between about 0.005% and about 0.05%, and said pharmaceutical formulation has pH between about 5.5 and about 6.5.


In a preferred embodiment, the pharmaceutical formulation of the present invention comprises an anti-OX40 antagonist antibody or fragment thereof present within said pharmaceutical formulation at a concentration of about 48 mg/m L, L-histidine buffer present within said pharmaceutical formulation at a concentration of about 5 mM, sucrose present within said pharmaceutical formulation at a percentage of about 4% and Polysorbate 80 present within said pharmaceutical formulation at a percentage of about 0.01%, and said pharmaceutical formulation has pH of about 6.0.


According to one aspect of the present invention, the pharmaceutical formulation disclosed herein is lyophilized and it is stable at about +5±3° C. for at least 24 months, preferably for at least 48 moths, at about +25±2° C. for at least 12 months, and at about +40±2° C. for at least 3 months.


According to another aspect of the present invention, the lyophilized formulation is reconstituted and comprises an anti-OX40 antagonist antibody or antibody fragment thereof at a concentration comprised between about 0.5 mg/mL and bout 200 mg/m L.


According to a further aspect of the present invention, the lyophilized formulation is reconstituted with water for injection to obtain a reconstituted formulation comprising an anti-OX40 antagonist antibody or antibody fragment thereof at a concentration comprised between about 100 mg/mL and bout 200 mg/mL.


In certain embodiments, the reconstituted formulation is further diluted with a dilution media selected from the group comprising saline solution, glucose solution, dextrose solution, to obtain a diluted formulation comprising an anti-OX40 antagonist antibody or antibody fragment thereof at a concentration comprised between about 0.5 mg/mL and bout 5 mg/m L.


According to certain aspects of the present invention, the pharmaceutical formulation disclosed herein is suitable for subcutaneous administration or for intravenous administration.


The present invention also relates to pharmaceutical formulation as disclosed herein for use in the treatment of an OX40-mediated disorder.


In a specific aspect, the OX40-mediated disorder is selected from the group comprising atopic dermatitis, rheumatoid arthritis, autoimmune uveitis, multiple sclerosis, lupus (such as systemic lupus erythematosus), ulcerative colitis, scleroderma and graft-versus-host disease (GVHD), scleroderma, hidradenitis, and ulcerative colitis.


The present invention also disclose a method of manufacturing said pharmaceutical formulation.


The present invention also disclose an article of manufacture comprising the pharmaceutical formulation.


As used herein, the following terms have the following meanings: “a”, “an”, and “the” as used herein refers to both singular and plural unless the context clearly dictates otherwise.


Unless otherwise defined, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Generally, nomenclatures utilized in connection with, and techniques of cell and tissue culture, molecular biology, and protein and oligo- or polynucleotide chemistry, laboratory procedures and techniques of analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well-known and commonly used in the art.


Temperatures above 0° C. are expressed in the present invention either by preceding the temperature value by “+” or not, e.g. 25° C. and +25° C. can be used interchangeably according to the present invention.


The pharmaceutical formulation of the invention may be a liquid formulation, a lyophilized formulation or a reconstituted formulation.


A “liquid” formulation is one that has been prepared in a liquid format. Such a formulation may be suitable for direct administration to a subject or, alternatively, can be packaged for storage either in a liquid form, in a frozen state or in a dried form (e.g. lyophilized) for later reconstitution into a liquid form or other form suitable for administration to a subject.


A “lyophilized” formulation is one that has been prepared by freeze-drying a liquid or pre-lyophilization formulation. Freeze-drying is performed by freezing the formulation and then subliming ice from the frozen content at a temperature suitable for primary drying. Under this condition the product temperature is below the collapse temperature of the formulation. A secondary drying stage may then be carried out, which produces a suitable lyophilized cake.


A “reconstituted” formulation is one that has been prepared by dissolving a lyophilized protein formulation in a diluent, such as a reconstitution media, such that the protein is dispersed in the reconstituted formulation. The reconstituted formulation should be suitable for administration (e.g. parenteral administration) to a subject to be treated with the protein of interest. Additionally, the reconstituted formulation can be further “diluted” if a diluent is added to be ready for administration, for instance via an infusion system. Suitable diluent may be selected from the group comprising saline solution, dextrose solution, glucose solution and their combination.


Suitable “reconstitution media” useful for the preparation of a reconstituted formulation include ones which are pharmaceutically acceptable (safe and non-toxic for administration to a human). Examples of suitable diluents include sterile water for injection (SWFI), bacteriostatic water for injection (BWFI). Water for injection (WFI), a pH buffered solution e.g. phosphate-buffered saline (PBS), sterile saline solution, Ringer's solution or dextrose solution and the buffer used to prepare the pharmaceutical formulation.


The term “buffer” as used herein refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components. Examples of buffers that can control the pH in this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, amino acids, such as histidine (e.g. histidine-HCl), citrate, phosphate, other organic acid buffer, their salts and combinations of buffers. A buffer of this invention has a pH in the range from about 5.0 to about 7.0; preferably the buffer has a pH selected from the group comprising pH about 5, pH about 5.5, pH about 6, pH about 6.5 and pH about 7. Examples of buffers that can control the pH in this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, amino acids, such as histidine (e.g. L-histidine), citrate, phosphate, other organic acid buffer, their salts and combinations of buffers. In one embodiment of the present invention the buffer is present within the pharmaceutical formulation at concentration between about 0.1 mg/mL and about 2 mg/mL; preferably the buffer is present within the pharmaceutical formulation at concentration selected from the group comprising 0.1 mg/mL, 0.2 mg/mL, 0.4 mg/mL, 0.6 mg/mL, 0.8 mg/mL, 1 mg/mL, 1.2 mg/mL, 1.4 mg/mL, 1.6 mg/mL, 1.8 mg/mL and 2 mg/mL. The present invention also includes a buffer with a concentration at any intermediate value of the above said values. In a particular embodiment of the present invention, the buffer is L-Histidine, present within the pharmaceutical formulation at a concentration of about 0.8 mg/mL. In one embodiment of the present invention the buffer is present within the pharmaceutical formulation at concentration between about 0.1 mM and about 50 mM; preferably the buffer is present within the pharmaceutical formulation at concentration selected from the group comprising 0.1 mM, 0.5 mM, 1 mM, 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 30 mM, 35 mM, 40 mM, 45 mM and 50 mM. The present invention also includes a buffer with a concentration at any intermediate value of the above said values. In a particular embodiment of the present invention, the buffer is L-Histidine, present within the pharmaceutical formulation at a concentration of about 5 mM.


Preferably, an excipient is added to the formulation. Such excipients may typically include one or more surfactant, lyoprotectant, bulking agent, inorganic or organic salt, stabilizer, diluent, solubilizer, reducing agent, antioxidant, chelating agent, preservative, stabilizing or tonicity agents and the like.


Examples of stabilizing or tonicity agent include comprising sodium acetate, sodium bicarbonate, sodium carbonate, sodium chloride (NaCl), potassium acetate, potassium bicarbonate, potassium carbonate, potassium chloride, calcium chloride (CaCl2)) sugars such as sucrose, glucose and trehalose, polyols such as mannitol, maltitol, sorbitol, xylitol, erythritol, and isomalt, polyethylene glycol, such as PEG400, Ethylenediaminetetraacetic acid (EDTA), amino acids such as histidine (e.g. histidine-HCl), arginine (e.g. arginine hydrochloride) and glycine, methionine, proline, lysine (e.g. lysine-HCl), glutamic acid, glutamine, cysteine, amines, glutathione, cyclodextrin, such as such as Hydroxypropyl β-cyclodextrin (HPBCD), Hydroxypropyl-sulfobutyl β-cyclodextrin (HPSBCD), Sulfobutylether β-cyclodextrin (SBECD), β-cyclodextrin (BetaCD), α-cyclodextrin (Alpha CD) and γ-cyclodextrin (Gamm CD) and surfactants. Non limiting examples of a typical surfactant include: non-ionic surfactants (HLB 6 to 18) such as sorbitan fatty acid esters (e.g. sorbitan monocaprylate, sorbitan monolaurate, sorbitan monopalmitate), glycerine fatty acid esters (e.g. glycerine monocaprylate, glycerine monomyristate, glycerine monostearate), poly glycerine fatty acid esters (e.g. decaglyceryl monostearate, decaglyceryl distearate, decaglyceryl monolinoleate), polyoxyethylene sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monostearate, polyoxyethylene sorbitan monopalmitate, polyoxyethylene sorbitan trioleate, polyoxyethylene sorbitan tristearate), polyoxyethylene sorbitol fatty acid esters (e.g. polyoxyethylene sorbitol tetrastearate, polyoxyethylene sorbitol tetraoleate), polyoxyethylene glycerine fatty acid esters (e.g. polyoxyethylene glyceryl monostearate), polyethylene glycol fatty acid esters (e.g. polyethylene glycol distearate), polyoxyethylene alkyl ethers (e.g. polyoxyethylene lauryl ether), polyoxy ethylene polyoxypropylene alkyl ethers (e.g. polyoxyethylene polyoxypropylene glycol ether, polyoxyethylene polyoxypropylene propyl ether, polyoxyethylene polyoxypropylene cetyl ether), polyoxyethylene alkylphenyl ethers (e.g. polyoxyethylene nonylphenyl ether), polyoxyethylene hydrogenated castor oils (e.g. polyoxyethylene castor oil, polyoxyethylene hydrogenated castor oil), polyoxyethylene beeswax derivatives (e.g. polyoxyethylene sorbitol beeswax), polyoxyethylene lanolin derivatives (e.g. polyoxyethylene lanolin), and polyoxyethylene fatty acid amides (e.g. polyoxyethylene stearyl amide); anionic surfactants such as Cio-Cis alkyl sulfates salts (e.g. sodium cetyl sulfate, sodium lauryl sulfate, sodium oleyl sulfate), polyoxyethylene Cio-Cis alkyl ether sulfates salts with an average of 2-4 moles of ethylene oxide (e.g. sodium polyoxyethylene lauryl sulfate), and Cs-Cis alkyl sulfosuccmate ester salts (e.g. sodium lauryl sulfosuccmate ester); natural surfactants such as lecithin, glycerophospho lipid, sphingophospho lipids (e.g. sphingomyelin) and sucrose esters of C 12-C 18 fatty acids; Poloxamers such as Poloxamer 188, Poloxamer 407, Poloxamer 124, Poloxamer 237, Poloxamer 338; salts and combinations of the above cited components. Preferably, the surfactant is selected from polyoxyethylene sorbitan fatty acid esters. Particularly preferably the surfactant is Polysorbate 20, 21, 40, 60, 65, 80, 81 and 85, most preferably Polysorbate 80. Polysorbate 80 is also known by the brand name Tween 80™ (ICI Americas, Inc.).


Examples of solubilizers include amino acids such as proline or glycine, prophylene glycol, plasdone C and K povidones, cyclodextrins and plasdone K polymers. Example of reducing agents, antioxidants and/or chelating agents. Examples of a reducing agent include N-acetylcysteine, N-acetylhomocysteine, thioctic acid, thiodiglycol, thioethanolamine, thioglycerol, thiosorbitol, thioglycolic acid and a salt thereof, sodium thiosulfate, glutathione and a C1-C7 thioalkanoic acid. Examples of an antioxidant include amino acids such as methionine, erythorbic acid, dibutylhydroxytoluene, butylhydroxyanisole, alpha-tocopherol, tocopherol acetate, L-ascorbic acid and a salt thereof, L-ascorbic acid palmitate, L-ascorbic acid stearate, sodium bisulfite, sodium sulfite, triamyl gallate and propyl gallate. Examples of a chelating agent include disodium ethylenediammetetraacetate (EDTA), sodium pyrophosphate and sodium metaphosphate.


In certain embodiments of the present invention, the stable pharmaceutical formulation comprise one or more stabilizing or tonicity agent at a concentration between about 0.05 mg/mL and about 50 mg/mL, preferably at a concentration selected from the group comprising about 0.05 mg/mL, about 0.1 mg/mL, about 0.5 mg/mL, about 1 mg/mL, about 5 mg/mL, about 10 mg/mL, about 15 mg/mL, about 20 mg/mL, about 25 mg/mL, about 30 mg/mL, about 35 mg/mL, about 40 mg/mL, about 45 mg/mL, about 50 mg/m L. The present invention also includes one or more stabilizing or tonicity agent with a concentration at any intermediate value of the above said values. In a particular embodiment of the present invention, the one or more stabilizing or tonicity agents are sucrose present within the pharmaceutical formulation at a concentration of about 40 mg/mL and Polysorbate 80 present within the pharmaceutical formulation at a concentration of about 0.1 mg/mL. In certain embodiments of the present invention, the stable pharmaceutical formulation comprise one or more stabilizing or tonicity agent at a percentage between about 0.001% and about 10%, preferably at a concentration selected from the group comprising about 0.001%, about 0.005%, about 0.01%, about 0.05%, about 0.1%, about 0.5%, about 1%, about 2%, about 4%, about 5%, about 8%, about 10%. The present invention also includes one or more stabilizing or tonicity agent with a percentage at any intermediate value of the above said values. In a particular embodiment of the present invention, the one or more stabilizing or tonicity agents are sucrose present within the pharmaceutical formulation at a percentage of about 4% and Polysorbate 80 present within the pharmaceutical formulation at a percentage of about 0.01%.


Not all excipients in a liquid formulation are suitable for inclusion a lyophilized form of the formulation and therefore a number of changes may be made to the liquid formulation to make it suitable for freeze-drying.


A stabilizer may be added to the formulation to stabilise the protein in the lyophilised form. Examples of a stabiliser include creatinine, an amino acid selected from histidine, alanine, glutamic acid, glycine, leucine, phenylalanine, methionine, isoleucine, proline, aspartic acid, arginine, lysine and threonine, a carbohydrate selected from sucrose, trehalose, sorbitol, xylitol and mannose, surfactants selected from polyethylene glycol (PEG; e.g. PEG3350 or PEG4000) or polyoxyethylene sorbitan fatty acid esters (e.g. Polysorbate 20 or Polysorbate 80), or any combination thereof. In a preferred embodiment the stabiliser comprises a lyoprotectant, which may be selected from a non-reducing sugar (e.g. sucrose or trehalose) and/or a sugar alcohol (e.g. mannitol). Addition of a lyoprotectant helps to reduce the amount of degradation or aggregation of the protein upon lyophilisation. For instance, the lyophilized formulation is isotonic upon reconstitution; therefore the amount of lyoprotectant in the lyophilized formulation should be sufficient to achieve an isotonic reconstituted formulation. Alternatively, the reconstituted formulation may be hypertonic and therefore a greater amount of lyoprotectant is required in the lyophilized formulation.


A preservative may be added to the diluent to reduce bacterial action in the reconstituted formulation Examples of a preservative include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethyl-ammonium chlorides in which the alkyl groups are long-chain compounds), benzethonium chloride, aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol.


The lyophilized formulation may be reconstituted with a diluent. Diluents for use in reconstitution of the lyophilized formulation include sterile water, bacteriostatic water for injection (BWFI), water for injection (WFI) a pH buffered solution such as phosphate buffered saline, sterile saline solution, Ringer's solution or dextrose solution.


The pharmaceutical formulation according to the present invention also comprises an anti-OX40 antagonist antibody or an antibody fragment thereof.


The term “antibody” as referred to herein includes whole antibodies and any antigen binding fragments or single chains thereof. An “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding fragment thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) with are hypervariable in sequence and/or involved in antigen recognition and/or usually form structurally defined loops, interspersed with regions that are more conserved, termed framework regions (FR or FW). Each VH and VL is composed of three CDRs and four FWs, arranged from amino-terminus to carboxy-terminus in the following order: FW1, CDR1, FW2, CDR2, FW3, CDR3, FW4. The amino acid sequences of FW1, FW2, FW3, and FW4 all together constitute the “non-CDR region” or “non-extended CDR region” of VH or VL as referred to herein.


The term “heavy chain variable framework region” as referred herein may comprise one or more (e.g., one, two, three and/or four) heavy chain framework region sequences (e.g., framework 1 (FW1), framework 2 (FW2), framework 3 (FW3) and/or framework 4 (FW4)). Preferably the heavy chain variable region framework comprises FW1, FW2 and/or FW3, more preferably FW1, FW2 and FW3. The term “light chain variable framework region” as referred herein may comprise one or more (e.g., one, two, three and/or four) light chain framework region sequences (e.g., framework 1 (FW1), framework 2 (FW2), framework 3 (FW3) and/or framework 4 (FW4)). Preferably the light chain variable region framework comprises FW1, FW2 and/or FW3, more preferably FW1, FW2 and FW3.


The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the First component (C1q) of the classical complement system.


Antibodies are grouped into classes, also referred to as isotypes, as determined genetically by the constant region. Human constant light chains are classified as kappa (CK) and lambda (CX) light chains. Heavy chains are classified as mu (μ), delta (δ), gamma (γ), alpha (α), or epsilon (ε), and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Thus, “isotype” as used herein is meant any of the classes and/or subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions. The known human immunoglobulin isotypes are IgG1 (IGHG1), IgG2 (IGHG2), IgG3 (IGHG3), IgG4 (IGHG4), IgA1 (IGHA1), IgA2 (IGHA2), IgM (IGHM), IgD (IGHD), and IgE (IGHE). The so-called human immunoglobulin pseudo-gamma IGHGP gene represents an additional human immunoglobulin heavy constant region gene which has been sequenced but does not encode a protein due to an altered switch region (Bensmana M et al., (1988) Nucleic Acids Res. 16(7): 3108). In spite of having an altered switch region, the human immunoglobulin pseudo-gamma IGHGP gene has open reading frames for all heavy constant domains (CH1-CH3) and hinge. All open reading frames for its heavy constant domains encode protein domains which align well with all human immunoglobulin constant domains with the predicted structural features. This additional pseudo-gamma isotype is referred herein as IgGP or IGHGP. Other pseudo immunoglobulin genes have been reported such as the human immunoglobulin heavy constant domain epsilon PI and P2 pseudo-genes (IGHEP1 and IGHEP2). The IgG class is the most commonly used for therapeutic purposes. In humans this class comprises subclasses IgG1, IgG2, IgG3 and IgG4. In mice this class comprises subclasses IgG1, IgG2a, IgG2b, IgG2c and IgG3.


The terms “antagonistic antibody” or “antagonist antibody” are used herein equivalently and include an antibody that is capable of inhibiting and/or neutralising the biological signaling activity of OX40, for example by blocking binding or substantially reducing binding of OX40 to OX40 ligand and thus inhibiting or reducing the signalisation pathway triggered by OX40 and/or inhibiting or reducing an OX40-mediated cell response like lymphocyte proliferation, cytokine expression, or lymphocyte survival.


The term “anti-OX40 antagonist antibody or fragment thereof” is used herein to indicate antibodies or antibody fragments thereof that bind to OX40 e.g. human OX40, and are capable of inhibiting and/or neutralising the biological signalling activity of OX40, for example by blocking binding or substantially reducing binding of OX40 to OX40 ligand and thus inhibiting or reducing the signalisation pathway triggered by OX40 and/or inhibiting or reducing an OX40-mediated cell response like lymphocyte proliferation, cytokine expression, or lymphocyte survival.


Antibody fragments include, but are not limited to, (i) the Fab fragment consisting of VL, VH, CL and CHI domains, including Fab′ and Fab′-SH, (ii) the Fd fragment consisting of the VH and CHI domains, (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward E S et al., (1989) Nature, 341: 544-546) which consists of a single variable, (v) F(ab′)2 fragments, a bivalent fragment comprising two linked Fab fragments (vi) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site (Bird R E et al, (1988) Science 242: 423-426; Huston J S et al, (1988) Proc. Natl. Acad. Sci. USA, 85: 5879-83), (vii) bispecific single chain Fv dimers (PCT/US92/09965), (viii) “diabodies” or “triabodies”, multivalent or multispecific fragments constructed by gene fusion (Tomlinson I & Hollinger P (2000) Methods Enzymol. 326: 461-79; WO94/13804; Holliger P et al, (1993) Proc. Natl. Acad. Sci. USA, 90: 6444-48) and (ix) scFv genetically fused to the same or a different antibody (Coloma M J & Morrison S L (1997) Nature Biotechnology, 15(2): 159-163).


In a particular embodiment of the present invention, the stable pharmaceutical formulation comprises an antibody or antibody fragment thereof present within said pharmaceutical formulation at a concentration between about 5 mg/mL and about 100 mg/mL, preferably between about 20 mg/mL and about 80 mg/mL, more preferably between about 40 mg/mL and about 60 mg/mL. In particular, the stable pharmaceutical formulation of the present invention comprises an antibody or antibody fragment at a concentration selected from the group comprising about 5 mg/mL, about 10 mg/mL, about 15 mg/mL, about 20 mg/mL, about 25 mg/mL, about 30 mg/mL, about 35 mg/mL, about 40 mg/mL, about 45 mg/mL, about 48 mg/mL, about 50 mg/mL, about 55 mg/mL, about 60 mg/mL, about 65 mg/mL, about 70 mg/mL, about 75 mg/mL, about 80 mg/mL, about 85 mg/mL, about 90 mg/mL, about 95 mg/mL, about 100 mg/mL. The present invention also includes an antibody or antibody fragment thereof with a concentration at any intermediate value of the above said values. In a particular embodiment of the present invention, the antibody or antibody fragment thereof is present within the pharmaceutical formulation at a concentration of about 48 mg/m L.


In one aspect of the present invention the formulation according to the present invention comprises an anti-OX40 antagonist antibody or fragment thereof, a buffer and one or more stability or tonicity agents. In a particular aspect the pharmaceutical formulation comprises histidine buffer and/or sucrose and/or Polysorbate 80. In a more particular aspect the pharmaceutical formulation of the present invention comprises Histidine buffer at a concentration between about 0.1 mM and about 50 mM, preferably between about 1 mM and about 10 mM, more preferably at a concentration of about 5 mM; Polysorbate 80 at a concentration between about 0.0001% and 10%, preferably between about 0.005% and about 1%, preferably between about 0.001% and about 1%, more preferably at a concentration of about 0.01% and sucrose at a concentration between about 0.1% and 10%, preferably between about 1% and about 8%, more preferably at a concentration of about 4%. In a specific aspect of the present application, the anti-OX40 antagonist antibody or fragment thereof is present within the pharmaceutical formulation at a concentration between about 1 mg/mL and about 100 mg/mL, preferably at a concentration of about 50 mg/mL, more preferably at a concentration of about 48 mg/mL. The present invention also discloses concentrations of the anti-OX40 antagonist antibody or fragment thereof, of the buffer and of the stability or tonicity agents at any value intermediate to the above cited values. According to one aspect of the present invention the pharmaceutical formulation has a pH between about 5 and 7, preferably of about 6.


In a certain embodiment, the present invention discloses a pharmaceutical formulation wherein said anti-OX40 antagonist antibody or fragment thereof is present within said pharmaceutical formulation at a concentration between about 20 mg/mL and about 80 mg/mL, L-histidine buffer is present within said pharmaceutical formulation at a concentration between about 1 mM and 10 mM, sucrose is present within said pharmaceutical formulation at a percentage between about 1% and about 10% and Polysorbate 80 present within said pharmaceutical formulation at a percentage between about 0.001% and about 0.1%, and wherein said pharmaceutical formulation has pH between about 5.0 and about 7.0.


In a certain embodiment, the present invention discloses a pharmaceutical formulation wherein said anti-OX40 antagonist antibody or fragment thereof is present within said pharmaceutical formulation at a concentration between about 40 mg/mL and about 60 mg/mL, L-histidine buffer is present within said pharmaceutical formulation at a concentration between about 3 mM and 7 mM, sucrose is present within said pharmaceutical formulation at a percentage between about 2% and about 6% and Polysorbate 80 present within said pharmaceutical formulation at a percentage between about 0.005% and about 0.05%, and wherein said pharmaceutical formulation has pH between about 5.5 and about 6.5.


In one aspect the pharmaceutical formulation of the present invention is lyophilized. In one embodiment the lyophilizate comprises histidine at an amount between about 0.1 mg and about 50 mg, preferably between about 1 mg and about 10 mg, more preferably at an amount of about 4 mg or at an amount of about 3.1 mg, Polysorbate 80 at an amount between about 0.01 mg and 10 mg, preferably between about 0.1 mg and 1 mg, more preferably at an amount of about 0.5 mg or at an amount of about 0.4 mg and sucrose at an amount between about 100 mg and 300 mg, preferably between about 150 mg and about 250 mg, more preferably at an amount of about 200 mg or at an amount of about 160 mg. In a specific aspect of the present application, the anti-OX40 antagonist antibody or fragment thereof is present within the pharmaceutical formulation at an amount between about 100 mg and about 350 mg, preferably between about 150 mg and about 250 mg, more preferably at an amount of about 240 mg or at an amount of about 192 mg. The present invention also discloses concentrations of the anti-OX40 antagonist antibody or fragment thereof, of the buffer and of the stability or tonicity agents at any value intermediate to the above cited values.


According to one aspect of the present invention, a reconstitution medium such as water for injection (WFI) is added to the lyophilized formulation such that the antibody concentration in the reconstituted formulation is present in an amount of between about 0.1 mg/ml and about 250 mg/ml, preferably between about 0.5 mg/ml and about 200 mg/ml. In certain aspects of the present invention the antibody concentration in the reconstituted formulation between about 100 mg/ml and about 200 mg/ml, more preferably the antibody concentration in the reconstituted formulation is selected from the group comprising about 100 mg/ml, about 120 mg/mL, about 150 mg/mL, about 170 mg/mL, about 200 mg/mL; more preferably the antibody concentration in the reconstituted formulation is about 150 mg/mL. The present invention also includes antibody concentrations at any intermediate value of the above said values.


In other particular aspects of the present invention, the reconstituted formulation is further diluted with a dilution media selected from the group comprising saline solution, glucose solution, dextrose solution, and their combinations to obtain a diluted formulation comprising said antibody or antibody fragment thereof at a concentration below 200 mg/mL, or below 150 mg/mL, below 100 mg/mL, below 50 mg/mL, below 10 mg/mL, below 5 mg/mL; preferably comprised between about 0.1 mg/mL and bout 150 mg/mL; more preferably between about 0.5 mg/mL and bout 5 mg/mL. In certain aspects, the antibody concentration in the diluted formulation is selected from the group comprising about 0.5 mg/ml, about 1 mg/mL, about 1.2 mg/mL, about 1.5 mg/mL, about 1.8 mg/mL, about 2 mg/mL, about 2.2 mg/mL, about 2.5 mg/mL, about 2.8 mg/mL, about 3 mg/mL about 3.2 mg/mL, about 3.5 mg/mL, about 3.8 mg/mL, about 4 mg/mL about 4.2 mg/mL, about 4.5 mg/mL, about 4.8 mg/mL, about 5 mg/mL; more preferably the antibody concentration in the reconstituted formulation is about 2.2 mg/m L. The present invention also includes antibody concentrations at any intermediate value of the above said values.


The pharmaceutical formulation according to the present invention is stable. A “stable” formulation is one in which the protein therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed for example in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones A (1993) Adv Drug Delivery Rev, 10: 29-90. Stability can be measured at a selected temperature for a selected time period.


Analytical tests useful to determine said stability include but are not limited to: monitoring of the visual appearance as a significant change in the appearance of sample may indicate product degradation and/or microbial contamination leading to safety risk for the patients; sub-visible particles analysis, as the presence of higher sub-visible particles in parental solutions may lead to immunogenic responses; protein content measurement (e.g. by measuring absorbance at 280 nm wavelength (A280) by UV-VIS Spectroscopy or by SoloVPE) as any significant variation from its target concentration would not provide effective dose to patients; pH measurement as changes in pH may be indicative of degradation of buffering agents and lead to protein instability; size variants monitoring (e.g. by SE-HPLC and/or by cGE reduced and non-reduced) as changes in monomeric content toward aggregates (higher size than monomer) or fragments (smaller size than monomer) is an indication of its degradation; charge variants monitoring (e.g. by clEF) as changes in content of charged variants is an indication of its degradation; antibody potency measurement (e.g. by ELISA) as any significant change of binding property of the antibody toward its target would indicate antibody degradation. Additionally, the amino acid sequence as well as post-translational modifications (i.e. deamidation, oxidation, glycation, N-terminal variants, C-terminal variants and glycosylation site occupancy) can be verified, for instance by peptide mapping. Other characteristics of the formulation can be monitored, such as osmolarity and viscosity, as well as the protein thermal stability for instance by nano-format of Differential Scanning Fluorimetry (DSF).


In one aspect of the present invention, the pharmaceutical formulation is stable at about +5±3° C. for at least 48 months, at about +25±2° C. for at least 12 months, and at about +40±2° C. for at least 3 months.


In a particular embodiment, the anti-OX40 antibody or fragment thereof is a humanized monoclonal anti-OX40 antagonist antibody or fragment thereof.


The term “humanized antibody” or “humanized anti-OX40 antibody” as used herein includes antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences as well as within the CDR sequences derived from the germline of another mammalian species.


More specifically, the anti-OX40 antagonist antibody or fragment is GBR830 (CAS Registry Number 2126777-87-3).


In a certain embodiment, the pharmaceutical formulation of the present invention can be used in the treatment of patients suffering of an OX40-mediated disorders.


As used herein, the term “OX40-mediated disorder” includes conditions such as allergy, asthma, COPD, rheumatoid arthritis, psoriasis and diseases associated with autoimmunity and inflammation. In particular, according to the present invention, exemplary OX40 mediated disorders include infections (viral, bacterial, fungal and parasitic), endotoxic shock associated with infection, arthritis, rheumatoid arthritis, asthma, chronic obstructive pulmonary disease (COPD), pelvic inflammatory disease, Alzheimer's Disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Peyronie's Disease, coeliac disease, gallbladder disease, Pilonidal disease, peritonitis, psoriasis, vasculitis, surgical adhesions, stroke, Type I Diabetes, lyme disease, arthritis, meningoencephalitis, autoimmune uveitis, immune mediated inflammatory disorders of the central and peripheral nervous system such as multiple sclerosis, lupus (such as systemic lupus erythematosus) and Guillain-Barr syndrome, Atopic dermatitis, autoimmune hepatitis, fibrosing alveolitis, Grave's disease, IgA nephropathy, idiopathic thrombocytopenic purpura, Meniere's disease, pemphigus, primary biliary cirrhosis, sarcoidosis, scleroderma, Wegener's granulomatosis, pancreatitis, trauma (surgery), graft-versus-host disease (GVHD), transplant rejection, cardiovascular disease including ischaemic diseases such as myocardial infarction as well as atherosclerosis, intravascular coagulation, bone resorption, osteoporosis, osteoarthritis, periodontitis, hypochlorhydia, hidradenitis and neuromyelitis optica.


Other exemplary OX40 mediated disorder include infections (viral, bacterial, fungal and parasitic), endotoxic shock associated with infection, arthritis, rheumatoid arthritis, asthma, bronchitis, influenza, respiratory syncytial virus, pneumonia, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), cryptogenic fibrosing alveolitis (CFA), idiopathic fibrosing interstitial pneumonia, emphysema, pelvic inflammatory disease, Alzheimer's Disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Peyronie's Disease, coeliac disease, gallbladder disease, Pilonidal disease, peritonitis, psoriasis, vasculitis, surgical adhesions, stroke, Type I Diabetes, lyme disease, arthritis, meningoencephalitis, autoimmune uveitis, immune mediated inflammatory disorders of the central and peripheral nervous system such as multiple sclerosis, lupus (such as systemic lupus erythematosus) and Guillain-Barr syndrome, Atopic dermatitis, autoimmune hepatitis, fibrosing alveolitis, Grave's disease, IgA nephropathy, idiopathic thrombocytopenic purpura, Meniere's disease, pemphigus, primary biliary cirrhosis, sarcoidosis, scleroderma, Wegener's granulomatosis, pancreatitis, trauma (surgery), graft-versus-host disease (GVHD), transplant rejection, cardiovascular disease including ischaemic diseases such as myocardial infarction as well as atherosclerosis, intravascular coagulation, bone resorption, osteoporosis, osteoarthritis, periodontitis, hypochlorhydia, hidradenitis and neuromyelitis optica.


In accordance to a preferred aspect of the present invention, the pharmaceutical formulation of the present invention is used for the treatment or prevention of an OX40-mediated disorder selected from the group comprising atopic dermatitis, rheumatoid arthritis, autoimmune uveitis, multiple sclerosis, lupus (such as systemic lupus erythematosus), ulcerative colitis, scleroderma and graft-versus-host disease (GVHD), scleroderma, hidradenitis, and ulcerative colitis.


The present invention provides a pharmaceutical formulation for use in the treatment of an OX40-mediated disorder selected from the group comprising atopic dermatitis, rheumatoid arthritis, autoimmune uveitis, multiple sclerosis, lupus (such as systemic lupus erythematosus), ulcerative colitis, scleroderma and graft-versus-host disease (GVHD), scleroderma, hidradenitis, and ulcerative colitis.


The present invention also provides a method for treating an OX40-mediated disorder using the pharmaceutical formulation of the present invention, wherein the OX40-mediated disorder is selected from the group comprising atopic dermatitis, rheumatoid arthritis, autoimmune uveitis, multiple sclerosis, lupus (such as systemic lupus erythematosus) and graft-versus-host disease (GVHD), scleroderma, hidradenitis, and ulcerative colitis.


As used herein, the term “subject” includes any human or nonhuman animal. The term “nonhuman animal” includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. Preferably the subject is human.


A “patient” for the purposes of the present invention includes both humans and other animals, preferably mammals and most preferably humans. Thus the antibodies of the present invention have both human therapy and veterinary applications. The term “treatment” or “treating” in the present invention is meant to include therapeutic treatment, as well as prophylactic, or suppressive measures for a disease or disorder. Thus, for example, successful administration of an antibody prior to onset of the disease results in treatment of the disease. As another example, successful administration of an antibody after clinical manifestation of the disease to combat the symptoms of the disease comprises treatment of the disease.


“Treatment” and “treating” also encompasses administration of an antibody after the appearance of the disease in order to eradicate the disease. Successful administration of an antibody after onset and after clinical symptoms have developed, with possible abatement of clinical symptoms and perhaps amelioration of the disease, comprises treatment of the disease. Those “in need of treatment” include mammals already having the disease or disorder, as well as those prone to having the disease or disorder, including those in which the disease or disorder is to be prevented.


The antibody or of the present invention can be administered via one or more routes of administration using one or more of a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. Preferred routes of administration include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. More preferred routes of administration are intravenous or subcutaneous. The phrase “parenteral administration” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Alternatively, an antibody of the invention can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasally, orally, vaginally, rectally, sublingually or topically.


According to a specific aspect of the present invention, the pharmaceutical formulation of the present invention is suitable for subcutaneous administration, or via intravenous infusion. In a particular aspect of the present invention, the pharmaceutical formulation of the present invention suitable for subcutaneous administration comprises the anti-OX40 antibody or fragment thereof at a concentration of about 150 mg/mL. In another particular aspect of the present invention, the pharmaceutical formulation of the present invention suitable for intravenous administration comprises the anti-OX40 antibody or fragment thereof at a concentration of about 2.2 mg/m L.


The antibody of the present invention can be administered at a single or multiple doses. The term “dose” or “dosage” as used in the present invention are interchangeable and indicates an amount of drug substance administered per body weight of a subject or a total dose administered to a subject irrespective to their body weight.


In one embodiment, a therapeutically effective amount of the pharmaceutical formulation according to the present invention is administrated to a patient in need thereof.


Administration is preferably in a “therapeutically effective amount”, this being sufficient to show benefit to a subject. Such benefit may be at least amelioration of at least one symptom. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of medical doctors. Appropriate doses of antibody are well known in the art (Ledermann J A et al., (1999) Int J Cancer 47: 659-664; Bagshawe K D et al., (1991) Antibody, Immunoconjugates and Radiopharmaceuticals, 4: 915-922). The precise dose will depend upon a number of factors, including the size and location of the area to be treated, body weight of the subject, the precise nature of the antibody (e.g. whole antibody or fragment) and any additional therapeutic agents administered before, at the time of or after administration of the antibody.


The present invention also relates to a method of manufacturing the pharmaceutical formulation described herein.


The present invention also relates to a method to test the pharmaceutical formulation disclosed herein to any assay for stability determination.


The present invention also relates to an article of manufacture comprising the pharmaceutical formulation.


EXAMPLE
Example 1

The stability of a high concentration drug products (HC DP) lyophilizate in vial GLEc013 composed of 240 mg of Antibody 1, 4 mg L-Histidine, 200 mg Sucrose and 0.5 mg Polysorbate 80 has been tested, based on the results in Table 1 to 3 it can be concluded that Antibody 1 is stable:


1) at +5±3° C. for 48 months


2) at +25±2° C. for 12 months


3) at +40±2° C. for 3 months.









TABLE 1





Stability at Long Term Stability Condition of +5 ± 3° C.

















Stability
Specifi-
Time Interval (months)














Tests
cation
T0
T1 M
T2 M
T3 M
T6 M
T9 M





Cake
Loose
Uniform,
Uniform,
Uniform,
Uniform,
Uniform,
Uniform,


Appearance
powder to
flat, not
flat,
flat,
flat, not
flat, not
flat, not



well-
adherent
adherent to
adherent to
adherent to
adherent to
adherent to



formed
to vial
most part of
most part of
vial surface.
vial surface,
vial surface,



cake,
surface,
the vial
the vial
No visible
cracked
not cracked



absence of
cracked in
surface.
surface.
cracks



foreign
some parts
Cracked in
Cracked in



body, no
of the
some parts
some parts



evidence of
surface.
of the
of the



meltback

surface.
surface.


Reconstitution
≤30 mins
15 min
31 min
6 min
14 min
10 min
10 min


time


Karl Fisher
≤4%
 0.3
NA
NA
NA
NA
NA


pH
pH at T0 ±0.2
 5.9
 6.0
 6.0
 6.0
 6.0
 6.0



≥5.7 and ≤6.1


Physical
Clear to
Clear and
Clear and
Clear and
Clear and
Clear and
Clear and


Appearance
slightly
slightly
slightly
brownish
slightly
brownish
slightly


of liquid
opalescent,
brownish
brownish

brownish

brownish



colorless to



(slightly)



brownish/



yellowish


A280
≥120 mg/ml
156 
153 
158 
163 
159 
153 



and ≤180 mg/ml


SE-HPLC
% monomer
99
99
99
99
99
99



IgG peak ≥95%


cGE (NR)
IgG peak ≥90%
92
92
92
92
92
93



Assigned
 7
 6
 6
 6
 7
 5



peaks ≤8%



Total other
 2
 2
 2
 2
 2
 1



peaks ≤3%


cGE (R)
Similar
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Heavy chain
65
64
63
63
64
63



peak ≥60%



Light chain
35
35
36
35
34
35



peak ≥30%



Total other
 1
 1
 1
 2
 3
 3



peaks ≤3%


cIEF/iCE3
Similar
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std)
Ref. Std.
Ref. Std.



Acidic
44
44
45
44
48
44



peaks: ≥14%



and ≤59%



Main
39
39
38
39
36
39



peak: ≥33%



and ≤54%



Basic
17
17
17
17
16
17



peaks: ≥7%



and ≤35%



pl main
 8.3
 8.3
 8.3
 8.3
 8.3
 8.2



peak: ≥8.1



and ≤8.5


CEX-HPLC
Similar
Similar
NA
NA
NA
Similar
NA



profile
profile



profile



to that
to that



to that



of the
of the



of the



Ref. Std.
Ref. Std.



Ref. Std.


Binding
≥50 and ≤150%
82
142 
122 
123 
127 
97


ELISA
of Ref. Std.



EC50


ELISA Ki
Report result
76
NA
NA
NA
114 
NA


(competition)
as % IC50 of



Ref. Std.


Subvisible
≤6000
11
NA
NA
NA
NA
NA


particles
particles of


(SVPs)
10 μm in



size or



larger per



container



≤600
 0
NA
NA
NA
NA
NA



particles of



25 μm in



size or



larger per



container


Container
Report
NA
NA
NA
NA
NA
NA


Closure
result


Integrity












Stability




Indicating

Time Interval (months)













Tests
Specification
T12 M
T18 M
T24 M
T36 M
T48 M





Cake
Loose powder
Uniform,
Partially
Well-formed
Well-formed
Loose powder


Appearance
to well-
concave,
collapsed
cake, no
cake, no
cake, no



formed cake,
not adherent
cake, absence
evidence of
evidence of
evidence of



absence of
to vial
of foreign
meltback,
meltback,
meltback,



foreign
surface,
body, no
absence of
absence of
absence of



body, no
cracked
evidence of
foreign body
foreign body
foreign body



evidence of

meltback



meltback


Reconstitution
≤30 mins
5 min
5 min
16 min
18 min
14 min


time


Karl Fisher
≤4%
 1
NA
 1
 0
 1


pH
pH at T0 ±0.2
 6.0
 6.0
 6.0
 6.0
 6.0



≥5.7 and ≤6.1


Physical
Clear to
Clear and
Clear and
Clear and
Slightly
Slightly


Appearance
slightly
brownish
brownish
brownish
opalescent,
opalescent,


of liquid
opalescent,



brownish
brownish



colorless to



(slightly)



brownish/



yellowish


A280
≥120 mg/ml
148 
157 
150 
165 
146 



and ≤180 mg/ml


SE-HPLC
% monomer
99
99
99
98
98



IgG peak ≥95%


cGE (NR)
IgG peak ≥90%
92
92
91
90
90



Assigned
 6
 7
 7
 7
 8



peaks ≤8%



Total other
 2
 2
 2
 3
 2



peaks ≤3%


cGE (R)
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Heavy chain
63
64
64
63
65



peak ≥60%



Light chain
35
35
35
36
34



peak ≥30%



Total other
 2
 1
 1
 1
 1



peaks ≤3%


cIEF/iCE3
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Acidic
42
44
45
46
44



peaks: ≥14%



and ≤59%



Main
40
39
38
37
38



peak: ≥33%



and ≤54%



Basic
18
18
17
18
18



peaks: ≥7%



and ≤35%



pl main
 8.3
 8.3
 8.3
 8.4
 8.3



peak: ≥8.1



and ≤8.5


CEX-HPLC
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.


Binding ELISA
≥50 and ≤150%
111 
99
97
83
113 



of Ref. Std.



EC50


ELISA Ki
Report result
130 
91
82
84
99


(competition)
as % IC50



of Ref. Std.


Subvisible
≤6000
403 
NA
98
43
81


particles
particles of


(SVPs)
10 μm in



size or



larger per



container



≤600
46
NA
26
22
48



particles of



25 μm in



size or



larger per



container


Container
Report
Pass
NA
Pass
NA
NA


Closure
result


Integrity
















TABLE 2







Stability at Accelerated Stability Condition of +25 ± 2° C.









Stability




Indicating

Time Interval (months)















Tests
Specification
T0
T1 M
T2 M
T3 M
T6 M
T9 M
T12 M





Cake
Loose powder
Uniform,
Uniform,
Uniform,
Uniform,
Uniform,
Uniform,
Uniform,


Appearance
to well-
flat, not
flat, adherent
flat, adherent
flat, not
flat, not
flat, not
concave,



formed cake,
adherent
to most part
to most part
adherent to
adherent to
adherent to
not adherent



absence of
to vial
of the vial
of the vial
vial surface.
vial surface,
vial surface,
to vial



foreign body,
surface,
surface.
surface.
No visible
cracked
cracked in
surface,



no evidence
cracked in
Cracked in
Cracked in
cracks.

some parts of
cracked



of meltback
some parts
some parts
some parts


the surface.




of the
of the
of the




surface.


Reconstitution
≤30 mins
15 min
21 mi
5 min
13 min
26 min
8 min
7 min


time


Karl Fisher
≤4%
 0.3
NA
NA
NA
NA
NA
1


pH
pH at T0 ±0.2
 5.9
 6.0
 6.0
 6.0
 6.0
 6.0
 6.0



(≥5.7 and ≤6.1)


Appearance
Clear to
Clear and
Clear and
Clear and
Clear and
Slightly
Clear and
Clear and


(liquid)
slightly
slightly
slightly
brownish
slightly
opalescent
slightly
brownish



opalescent,
brownish
brownish

brownish
and
brownish



colorless to




brownish



(slightly)



brownish/



yellowish


A280
≥120 mg/ml
156 
155 
168 
163 
162 
149 
143 



and ≤180 mg/ml


SE-HPLC
% monomer
99
99
98
99
99
98
98



IgG peak ≥95%


cGE (NR)
IgG peak ≥90%
92
92
92
92
92
93
92



Assigned
 7
 6
 7
 7
 6
 6
 6



peaks ≤8%



Total other
 2
 2
 2
 2
 2
 2
 2



peaks ≤3%


cGE (R)
Similar
Similar
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Heavy chain
65
65
63
63
64
63
64



peak ≥60%



Light chain
35
35
36
36
34
35
34



peak ≥30%



Total other
 1
 1
 1
 2
 2
 3
 2



peaks ≤3%


cIEF/iCE3
Similar
Similar
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Acidic
44
43
45
44
48
45
43



peaks: ≥14%



and ≤59%



Main
39
40
38
38
36
36
38



peak: ≥33%



and ≤54%



Basic
17
18
17
18
16
19
19



peaks: ≥7%



and ≤35%



pl main
 8.3
 8.4
 8.3
 8.3
 8.3
 8.3
 8.3



peak: ≥8.1



and ≤8.5


CEX-HPLC
Similar
Similar
NA
NA
NA
Similar
NA
Similar



profile
profile



profile

profile



to that
to that



to that

to that



of the
of the



of the

of the



Ref. Std.
Ref. Std.



Ref. Std.

Ref. Std.


Binding
≥50 and ≤150%
82
112 
121 
119 
88
69
111 


ELISA
of Ref. Std.



EC50


ELISA Ki
Report result
76
NA
NA
NA
110 
NA
102 


(competition)
as % IC50



of Ref. Std.


Particulate
≤6000
11
NA
NA
NA
NA
NA
13


content
particles of


(Subvisible)
10 μm in



size or



larger per



container



≤600
 0
NA
NA
NA
NA
NA
 0



particles of



25 μm in



size or



larger per



container
















TABLE 3







Stability at Stress Stability Condition of +40 ± 2° C.









Stability




Indicating

Time Interval (months)












Tests
Specification
T0
T1 M
T2 M
T3 M





Cake
Loose powder to well-
Uniform,
Uniform, flat,
Uniform, flat,
Uniform, flat,


Appearance
formed cake, absence
flat, not
adherent to
adherent to
not adherent



of foreign body, no
adherent
most part of
most part of
to vial



evidence of meltback
to vial
the vial
the vial
surface.




surface,
surface.
surface.
Cracked




cracked in
Cracked in
Cracked in





some parts
some parts
some parts





of the
of the
of the





surface.
surface.
surface.



Reconstitution
≤30 mins
15 min
23 min
6 min
13 min


time







Karl Fisher
≤4%
0.3%
NA
NA
 1%


pH
pH at T0 ± 0.2
5.9
6.0
6.0
6.0



(≥5.7 and ≤6.1)






Appearance
Clear to slightly
Clear and
Clear and
Clear and
Clear and


(liquid)
opalescent, colorless
slightly
slightly
brownish
slightly



to (slightly)
brownish
brownish

brownish



brownish/yellowish






A280
≥120 mg/ml and ≤180
156
155
162
163



mg/ml






SE-HPLC
% monomer IgG peak ≥
99
98
97
97



95%






cGE (NR)
IgG peak ≥ 90%
92
92
92
91



Assigned peaks ≤ 8%
7
6
6
7



Total other peaks ≤ 3%
2
2
2
2


cGE (R)
Similar profile to that
Similar
Similar
Similar
Similar



of the Ref. Std.
profile to
profile to
profile to
profile to




that of the
that of the
that of the
that of the




Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Heavy chain peak ≥
65
64
63
62



60%







Light chain peak ≥ 30%
35
35
36
36



Total other peaks ≤ 3%
1
1
1
2


cIEF/iCE3
Similar profile to that
Similar
Similar
Similar
Similar



of the Ref. Std.
profile to
profile to
profile to
profile to




that of the
that of the
that of the
that of the




Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Acidic peaks: ≥14%
44
43
45
44



and ≤59%







Main peak: ≥33%
39
38
36
37



and ≤54%







Basic peaks: ≥7% and
17
19
18
19



≤35%







pl main peak: ≥8.1
8.3
8.3
8.3
8.3



and ≤8.5






CEX-HPLC
Similar profile to that
Similar
NA
NA
Similar



of the Ref. Std.
profile to


profile to




that of the


that of the




Ref. Std.


Ref. Std.


Binding ELISA
≥50 and ≤150% of
 82%
120
106
109



Ref. Std. EC50






ELISA Ki
Report result as % IC50
76
NA
NA
95%


(competition)
of Ref. Std.






Particulate
≤6000 particles of
11
NA
NA
0


content
10 μm in size or larger






(Subvisible)
per container







≤600 particles of
0
NA
NA
0



25 μm in size or larger







per container









Example 2

Next the stability of a second presentation of the high concentration drug products (HC DP) lyophilizate in vial GLEc013 composed of 192 mg of Antibody 1, 3.1 mg L-Histidine, 160 mg Sucrose and 0.4 mg Polysorbate 80 was investigated. Based on the results in Table 4 to 6 it can be concluded that Antibody 1 is stable for:

    • 1) 24 months when stored at +5±3° C.
    • 2) 12 months when stored at +25±2° C./60%±5% RH.
    • 3) 3 months when stored at +40±2° C./75%±5% RH.









TABLE 4







Stability at Stability Condition of +5 ± 3° C.









Time interval (months)















Stability

Initial








Tests
Specification
T0
T3
T6
T9
T12
T18
T24





Physical
Clear to
Clear and
Clear and
Clear and
Clear and
Clear and
Clear and
Clear and


appearance
slightly
slightly
slightly
slightly
slightly
slightly
slightly
slightly


of liquid
opalescent,
yellowish
yellowish
yellowish
yellowish
yellowish
brownish.
brownish.



colorless to
liquid.
liquid.
liquid.
liquid.
liquid.
Practically
Practically



brownish/
Practically
Practically
Practically
Practically
Practically
free of
free of



yellowish,
free of
free of
free of
free of
free of
visual
visual



practically
visual
visual
visual
visual
visual
particulates
particulates



free of
particulates.
particulates.
particulates.
particulates.
particulates.



particulates


pH
pH at T0 ±0.2
 6.2
 6.3
 6.2
 6.2
 6.2
 6.2
 6.2



(≥6.0 and ≤6.4)


Cake
Loose powder
Uniform,
Well-formed
Well-formed
Well-formed
Well-formed
Well-formed
Well-formed


appearance
to well-
flat,
cake,
cake,
cake,
cake,
cake,
cake,



formed cake,
adherent
absence
absence
absence
absence
absence
absence



absence of
to the vial
of foreign
of foreign
of foreign
of foreign
of foreign
of foreign



foreign body,
surface,
body, no
body, no
body, no
body, no
body, no
body, no



no evidence
some cracks
evidence of
evidence of
evidence of
evidence of
evidence of
evidence of



of meltback

meltback
meltback
melt back
melt back
melt back
melt back



(2)


Reconstitution
≤30 mins
4 min 10 sec
6 min 30 sec
15 min 25 sec
12 min 36 sec
7 min 32 sec
17 min 27 sec
11 min 40 sec


time


Karl Fisher
≤4%
 1
NA
NA
NA
 1
NA
 1


A280
Concentration
151 
145 
145 
144 
141 
148 
145 



at T0 ±10%



(≥136 mg/mL and



≤166 mg/mL)


SE-HPLC
% monomer
99
99
98
99
98
98
99



IgG peak ≥95%


CGE (non-
IgG peak ≥90%
93
92
93
92
93
93
93


reduced)



Assigned
 5
 6
 5
 6
 6
 4
 5



peaks ≤8%



Total other
 2
 1
 2
 2
 2
 2
 2



peaks ≤3%


CGE
Similar
Similar
Similar
Similar
Similar
Similar
Similar
Similar


(reduced)
profile
profile
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Heavy chain
64
64
65
64
64
64
63



peak ≥60%



Light chain
35
35
35
35
35
35
36



peak ≥30%



Total other
 1
 1
 1
 1
 1
 1
 1



peaks ≤3%


cIEF/iCE3
Similar
Similar
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Acidic
40
33
33
35
34
33
33



peaks: ≥14%



and ≤59%



Main
41
45
45
44
44
45
45



peak: ≥33%



and ≤54%



Basic
20
22
22
21
22
22
23



peaks: ≥7%



and ≤35%



pl main
 8.3
 8.3
 8.3
 8.3
 8.3
 8.3
 8.2



peak: ≥8.1



and ≤8.5


HPLC-CEX
Similar
Similar
NA
Similar
NA
Similar
Similar
Similar



profile
profile

profile

profile
profile
profile



to that
to that

to that

to that
to that
to that



of the
of the

of the

of the
of the
of the



Ref. Std.
Ref. Std.

Ref. Std.

Ref. Std.
Ref. Std.
Ref. Std.



Report
18
NA
21
NA
20
19
18



% acidic



Report
47
NA
49
NA
50
49
48



% main



Report
35
NA
30
NA
30
32
31



% basic


Binding
≥50% and ≤150%
123 
75
80
104 
102 
80
105 


ELISA
of Ref. Std.



EC50


ELISA Ki
Report result
89
NA
100 
NA
94
117 
76


(competition)
as % IC50



of Ref. Std.


Subvisible
Report 2 μm
NA
NA
NA
NA
600 
NA
319 


particles
in size or



larger per



container



Report 5 μm
NA
NA
NA
NA
55
NA
119 



in size or



larger per



container



≤6000
27
NA
NA
NA
11
NA
84



particles of



10 μm in size



or larger per



container



≤600
 1
NA
NA
NA
 0
NA
33



particles of



25 μm in size



or larger per



container


Container
Report
NA
NA
NA
NA
Pass
NA
Pass


closure
results


integrity


Sterility
No Growth
No growth
NA
NA
NA
NA
NA
NA



at 14 days
at 14 days
















TABLE 5







Stability at Stability Condition of +25 ± 2° C./60% ± 5% RH








Stability
Time Interval (months)













Tests
Specification
Initial T0
T3
T6
T9
T12





Physical
Clear to
Clear and
Clear and
Clear and
Clear and
Clear and


appearance
slightly
slightly
slightly
slightly
slightly
slightly


of liquid
opalescent,
yellowish
yellowish
yellowish
yellowish
yellowish



colorless to
liquid.
liquid.
liquid.
liquid.
liquid.



brownish/
Practically
Practically
Practically
Practically
Practically



yellowish,
free of
free of
free of
free of
free of



practically
visual
visual
visual
visual
visual



free of
particulates.
particulates.
particulates.
particulates.
particulates.



particulates


pH
pH at T0 ± 0.2
 6.2
 6.2
 6.2
 6.2
 6.2



(≥6.0 and ≤6.4)


Cake
Loose powder
Uniform, flat,
Uniform cake,
Uniform cake,
Well-formed
Well-formed


appearance
to well-
adherent to
absence of
absence of
cake, absence
cake, absence



formed cake,
the vial
foreign
foreign
of foreign
of foreign



absence of
surface,
body, no
body, no
body, no
body, no



foreign body,
some cracks
evidence of
evidence
evidence
evidence



no evidence

meltback,
of melt-
of meltback
of meltback



of meltback (2)

cracked.
back


Reconstitution
≤30 mins
4 min 10 sec
7 min 22 sec
16 min 11 sec
13 min 22sec
8 min 46 sec


time


Karl Fisher
≤4%
 1
NA
NA
NA
 1


A280
Concentration at
151 
144 
147 
142 
144 



T0 ±10%



(≥136 mg/mL



and ≤166 mg/mL)


SE-HPLC
% monomer IgG
99
96
98
98
97



peak ≥95%


CGE (non-
IgG peak ≥90%
93
93
94
92
93


reduced)



Assigned
 5
 5
 5
 6
 6



peaks ≤8%



Total other
 2
 1
 2
 2
 2



peaks ≤3%


CGE
Similar
Similar
Similar
Similar
Similar
Similar


(reduced)
profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Heavy chain
64
64
64
64
63



peak ≥60%



Light chain
35
35
35
35
35



peak ≥30%



Total other
 1
 1
 1
 1
 1



peaks ≤3%


cIEF/iCE3
Similar
Similar
Similar
Similar
Similar
Similar



profile
profile
profile
profile
profile
profile



to that
to that
to that
to that
to that
to that



of the
of the
of the
of the
of the
of the



Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.
Ref. Std.



Acidic
40
33
34
34
34



peaks: ≥14%



and ≤59%



Main
41
42
43
43
42



peak: ≥33%



and ≤54%



Basic
20
25
23
23
24



peaks: ≥7%



and ≤35%



pl main
 8.3
 8.3
 8.3
 8.3
 8.3



peak: ≥8.1



and ≤8.5


HPLC-CEX
Similar
Similar
NA
Similar
NA
Similar



profile
profile

profile

profile



to that
to that

to that

to that



of the
of the

of the

of the



Ref. Std.
Ref. Std.

Ref. Std.

Ref. Std.



Report %
18
NA
21
NA
20



acidic



Report %
47
NA
49
NA
49



main



Report %
35
NA
30
NA
31



basic


Binding
≥50% and ≤150
123 
73
89
102 
87


ELISA
% of Ref. Std.



EC50


ELISA Ki
Report result
89
NA
112 
NA
123 


(competition)
as % IC50



of Ref. Std.


Sub-visible
Report 2 μm
NA
NA
NA
NA
825 


particles
in size or



larger per



container



Report 5 μm
NA
NA
NA
NA
70



in size or



larger per



container



≤6000
27
NA
NA
NA
28



particles



of 10 μm



in size or



larger per



container



≤600
 1
NA
NA
NA
 0



particles



of 25 μm



in size or



larger per



container
















TABLE 6







Stability at Stability Condition of +40 ± 2° C./75% ± 5% RH









Stability




Indicating

Time Interval (months)










Tests
Specification
Initial T0
T3





Physical
Clear to slightly opalescent,
Clear and slightly
Clear and slightly


appearance of
colorless to
yellowish liquid.
yellowish liquid.


liquid
brownish/yellowish,
Practically free of visual
Practically free of visual



practically free of
particulates.
particulates.



particulates




pH
pH at T0 ± 0.2 (≥6.0 and ≤
6.2
6.2



6.4)




Cake
Loose powder to well-formed
Uniform, flat, adherent
Uniform cake, absence


appearance
cake, absence of foreign
to the vial surface, some
of foreign body, no



body, no evidence of melt-
cracks
evidence of melt-back.



back (2)




Reconstitution
≤30 mins
4 min 10 sec
7 min 04 sec


time





Karl Fisher
≤4%
1
1


A280
Concentration at T0 ± 10% (≥
151
150



136 mg/mL and ≤166





mg/mL)




SE-HPLC
% monomer IgG peak ≥ 95%
99
98


CGE (non-
IgG peak ≥ 90%
93
93


reduced)
Assigned peaks ≤ 8%
5
6



Total other peaks ≤ 3%
2
1


CGE (reduced)
Similar profile to that of the
Similar profile to that of
Similar profile to that of



Ref. Std.
the Ref. Std.
the Ref. Std.



Heavy chain peak ≥ 60%
64
63



Light chain peak ≥ 30%
35
34



Total other peaks ≤ 3%
1
1


cIEF/iCE3
Similar profile to that of the
Similar profile to that of
Similar profile to that of



Ref. Std.
the Ref. Std.
the Ref. Std.



Acidic peaks: ≥ 14% and ≤
40
33



59%





Main peak: ≥33% and ≤54%
41
44



Basic peaks: ≥7% and ≤35%
20
23



pl main peak: ≥8.1 and ≤8.5
8.3
8.3


HPLC-CEX
Similar profile to that of the
Similar profile to that of
Similar profile to that of



Ref. Std.
the Ref. Std.
the Ref. Std.



Report % acidic
18
30



Report % main
47
47



Report % basic
35
23


Binding ELISA
≥50% and ≤150% of Ref. Std.
123
93



EC50




ELISA Ki
Report result as % IC50 of Ref.
89
88


(Competition)
Std.




Sub-visible
Report 2 μm in size or larger
NA
149


particles
per container





Report 5 μm in size or larger
NA
0



per container





≤6000 particles of 10 μm in
27
0



size or larger per container





≤600 particles of 25 μm in
1
0



size or larger per container











Example 3

The comparability of Antibody 1 with syringes and infusion bags, used in the clinics for subcutaneous (SC) injection and for intravenous (IV) infusion, respectively, was also investigated with regards to protein stability and adsorption over time at different temperatures and relevant protein concentrations.


SC Injection

The lyophilized drug product with each vials is composed of 240 mg Antibody 1, 4 mg Histidine, 200 mg sucrose and 0.5 mg polysorbate 80 (as in Example 1), the stability of Antibody 1 was reconstituted with water for injection (WFI) to target an antibody concentration of about 150 mg/ml.


Stability studies were performed in fully filled 2 ml syringes, in fully filled 1 mL syringes and in half filled 1 mL syringes, stored at 5±3° C. for 48 hours or at 25±2° C. for 24 hours. The results of the stability studies are shown in Table 7. The reconstituted drug product was found to be stable for 24 h at 25±2° C. and 48 h at 5±3° C. when stored in a syringe.









TABLE 7





Stability Antibody 1 formulation for SC administration at 5 ± 3° C. and 25 ± 2° C.



















Concentration and Incubation time




Target antibody concentration: 150 mg/ml




















2 ml fill in
1 ml fill in


Stability





2 ml syringe
1 ml syringe











Indicating
Specifica-
48 h at
24 h at
48 h at












Tests
tion
T0
5 ± 3° C.
25 ± 2° C.
5 ± 3° C.





Physical
Clear to
Clear and
Clear and
Clear and
Clear and


appearance
slightly
colorless
colorless
colorless
colorless


of liquid
opalescent



liquid,



colorless to



slightly



yellowish



liquid


pH
Report
 5.972
5.967
 5.965
5.963



results


A280
T0 +/−20%
11.125
8.592
10.553
8.457



(mg/ml)















9.367
9.700
9.171
9.417















SE-HPLC
% monomer ≥98%
99%
99%
99%
 99%


iCE
Comparable
Conform
Conform
Conform
Conform



to reference



standard


Binding
100 ± 50%
85%
85%
89%
111%


ELISA
of the



reference



standard


cGE (R)
Comparable
Conform
Conform
Conform
Conform



to reference



standard


cGE (NR)
Comparable
Conform
Conform
Conform
Conform



to reference



standard


Sub-visible
Not more
Number of
Number of
Number of
Number of


particles
than 6000
particles/
particles/
particles/
particles/



particles/
container ≥10
container ≥10
container ≥10
container ≥10



container
μm: 80.00;
μm: 141.42;
μm: 75.72;
μm: 47.86;



of size
Number of
Number of
Number of
Number of



equal to or
particles/
particles/
particles/
particles/



greater
container ≥25
container ≥25
container ≥25
container ≥25



than 10 μm
μm: 0.00
μm: 0.00
μm: 0.00
μm: 0.00



and not



more than



600



particles/



container



of size



equal to or



greater



than 25 μm*
















Concentration and Incubation time





Target antibody concentration: 150 mg/ml














1 ml fill in
0.5 ml fill in



Stability

1 ml syringe
1 ml syringe













Indicating
Specifica-
24 h at
48 h at
24 h at



Tests
tion
25 ± 2° C.
5 ± 3° C.
25 ± 2° C.







Physical
Clear to
Clear and
Clear and
Clear and



appearance
slightly
colorless
colorless
colorless



of liquid
opalescent




liquid,




colorless to




slightly




yellowish




liquid



pH
Report
5.976
6.004
6.008




results



A280
T0 +/− 20%
8.753
8.961
8.672




(mg/ml)



SE-HPLC
% monomer ≥98%
99%
99%
99%



iCE
Comparable
Conform
Conform
Conform




to reference




standard



Binding
100 ± 50%
81%
71%
95%



ELISA
of the




reference




standard



cGE (R)
Comparable
Conform
Conform
Conform




to reference




standard



cGE (NR)
Comparable
Conform
Conform
Conform




to reference




standard



Sub-visible
Not more
Number of
Number of
Number of



particles
than 6000
particles/
particles/
particles/




particles/
container ≥10
container ≥10
container ≥10




container
μm: 32.85;
μm: 18.22;
μm: 32.86;




of size
Number of
Number of
Number of




equal to or
particles/
particles/
particles/




greater
container ≥25
container ≥25
container ≥25




than 10 μm
μm: 0.00
μm: 0.36
μm: 0.36




and not




more than




600




particles/




container




of size




equal to or




greater




than 25 μm*











IV infusion


The lyophilized drug product with each vials is composed of 240 mg Antibody 1, 4 mg Histidine, 200 mg sucrose and 0.5 mg polysorbate 80 (as in Example 1), the stability of Antibody 1 was reconstituted with water for injection (WFI) to target an antibody concentration of about 150 mg/ml, and further dilution in 500 ml 0.9% NaCl infusion bags for IV infusion, to target an antibody concentration of 2.2 mg/ml.


The antibody stability was studied at 5±3° C. for 48 hours and at 25±2° C. for 24 hours. The results of the stability studies are shown in Table 8. Antibody 1 was found to be stable for 24 h at 25±2° C. and 48 h at 5±3° C. when diluted in saline bags.









TABLE 8







Stability Antibody 1 formulation for IV administration at 5 ± 3° C. and 25 ± 2° C.











Concentration and Incubation time




Target concentration: 2.2 mg/ml












Stability

T0
T0




Indicating

(corresponding
(corresponding
48 h at
24 h at


Tests
Specification
to 5 ± 3° C.)
to 25 ± 2° C.)
5 ± 3° C.
25 ± 2° C.





Physical
Clear to slightly
Clear and
Clear and
Clear and
Clear and


appearance
opalescent liquid,
colorless
colorless
colorless
colorless


of liquid
colorless to slightly







yellowish liquid






pH
Report results*
6.226
6.336
6.086
6.110


A280
T0 +/− 20% (mg/ml)
1.884
1.882
1.863
1.857


SE-HPLC
% monomer ≥ 98%
99%
99%
 99%
 99%


iCE
Comparable to
Conform
Conform
Conform
Conform



reference standard






Binding
100 ± 50% of the
98%
NA
106%
112%


ELISA
reference standard






cGE (R)
Comparable to
Conform
Conform
Conform
Conform



reference standard






cGE (NR)
Comparable to
Conform
Conform
Conform
Conform



reference standard






Sub-visible
≤25 particles/mL of
Particles/mL
Particles/mL
Particles/mL
Particles/mL


particles
size equal to or
for ≥ 25 μm:
for ≥ 25 μm:
for ≥ 25 μm:
for ≥ 25 μm:



greater than ≥ 10 μm
48.67
44.83
10
5.34



& ≤ 3 particles/mL of
Particles/mL
Particles/mL
Particles/mL
Particles/mL



size equal to or
for ≥ 10 μm:
for ≥ 10 μm:
for ≥ 10 μm:
for ≥ 10 μm:



greater than ≥ 25 μm
0.67
0.50
0
0.33








Claims
  • 1. A stable pharmaceutical formulation comprising an anti-OX40 antagonist antibody or antibody fragment thereof, one or more stabilizing or tonicity agent and a buffer, wherein said pharmaceutical formulation is liquid or lyophilized.
  • 2. The pharmaceutical formulation of any one of the preceding claims, wherein said buffer is selected from the group comprising acetate, L-histidine, citrate and phosphate and wherein said buffer is present within said pharmaceutical formulation at concentration between about 1 mM and about 10 mM.
  • 3. The pharmaceutical formulation of any one of the preceding claims, wherein said one or more stabilizing or tonicity agent is selected from the group comprising sodium acetate, sodium bicarbonate, sodium carbonate, sodium chloride, potassium acetate, potassium bicarbonate, potassium carbonate, potassium chloride, calcium chloride, sucrose, glutamate, mannitol, polyols, Polysorbate 20, Polysorbate 40, Polysorbate 80, Poloxamer, Poloxamer 188, Poloxamer 407, amino acids such as histidine, arginine, glycine, methionine, proline, lysine, glutamic acid, amines, cyclodextrins, β-cyclodextrins, polyvinylpyrrolidone, polyethylene glycol 400, sorbitol, trehalose and EDTA, and wherein said one or more stabilizing or tonicity agent is present within said pharmaceutical formulation at a percentage between about 0.001% and about 10%.
  • 4. The pharmaceutical formulation of any one of the preceding claims wherein said anti-OX40 antagonist antibody or fragment thereof is present within said pharmaceutical formulation at a concentration between about 20 mg/mL and about 80 mg/m L, L-histidine buffer is present within said pharmaceutical formulation at a concentration between about 1 mM and 10 mM, sucrose is present within said pharmaceutical formulation at a percentage between about 1% and about 10% and Polysorbate 80 present within said pharmaceutical formulation at a percentage between about 0.001% and about 0.1%, and wherein said pharmaceutical formulation has pH between about 5.0 and about 7.0.
  • 5. The pharmaceutical formulation of any one of the preceding claims wherein said anti-OX40 antagonist antibody or fragment thereof is present within said pharmaceutical formulation at a concentration between about 40 mg/mL and about 60 mg/m L, L-histidine buffer is present within said pharmaceutical formulation at a concentration between about 3 mM and 7 mM, sucrose is present within said pharmaceutical formulation at a percentage between about 2% and about 6% and Polysorbate 80 present within said pharmaceutical formulation at a percentage between about 0.005% and about 0.05%, and wherein said pharmaceutical formulation has pH between about 5.5 and about 6.5.
  • 6. The pharmaceutical formulation of any one of the preceding claims wherein said anti-OX40 antagonist antibody or fragment thereof is present within said pharmaceutical formulation at a concentration of about 48 mg/m L, L-histidine buffer is present within said pharmaceutical formulation at a concentration of about 5 mM, sucrose is present within said pharmaceutical formulation at a percentage of about 4% and Polysorbate 80 present within said pharmaceutical formulation at a percentage of about 0.01%, and wherein said pharmaceutical formulation has pH of about 6.0.
  • 7. The pharmaceutical formulation of any one of the preceding claims, wherein said pharmaceutical formulation is lyophilized and it is stable at about +5±3° C. for at least 24 months, preferably for at least 48 moths, at about +25±2° C. for at least 12 months, and at about +40±2° C. for at least 3 months.
  • 8. The pharmaceutical formulation of claim 8, wherein said lyophilized formulation is reconstituted and comprises said antibody or antibody fragment thereof at a concentration comprised between about 0.5 mg/mL and bout 200 mg/m L.
  • 9. The pharmaceutical formulation of claim 7 or 8, wherein said lyophilized formulation is reconstituted with water for injection to obtain a reconstituted formulation comprising said antibody or antibody fragment thereof at a concentration comprised between about 100 mg/mL and bout 200 mg/m L.
  • 10. The pharmaceutical formulation of claim 9, wherein said reconstituted formulation is further diluted with a dilution media selected from the group comprising saline solution, glucose solution, dextrose solution, to obtain a diluted formulation comprising said antibody or antibody fragment thereof at a concentration comprised between about 0.5 mg/mL and bout 5 mg/m L.
  • 11. The pharmaceutical formulation of any one of claims 1 to 10, wherein said pharmaceutical formulation is suitable for subcutaneous administration or for intravenous administration.
  • 12. The pharmaceutical formulation of any one of the preceding claims for use in the treatment of an OX40-mediated disorder.
  • 13. The pharmaceutical formulation of any one of the preceding claims for use in the treatment of an OX40-mediated disorder selected from the group comprising atopic dermatitis, rheumatoid arthritis, autoimmune uveitis, multiple sclerosis, lupus (such as systemic lupus erythematosus), ulcerative colitis, scleroderma and graft-versus-host disease (GVHD), scleroderma, hidradenitis, and ulcerative colitis.
  • 14. A method of manufacturing the pharmaceutical formulation of any one of the preceding claims.
  • 15. An article of manufacture comprising the pharmaceutical formulation of any one of the preceding claims.
Priority Claims (1)
Number Date Country Kind
19187367.8 Jul 2019 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2020/070137 7/16/2020 WO