WO2025017034A1 - Stable liquid antibody formulation for dupilumab - Google Patents
Stable liquid antibody formulation for dupilumab Download PDFInfo
- Publication number
- WO2025017034A1 WO2025017034A1 PCT/EP2024/070169 EP2024070169W WO2025017034A1 WO 2025017034 A1 WO2025017034 A1 WO 2025017034A1 EP 2024070169 W EP2024070169 W EP 2024070169W WO 2025017034 A1 WO2025017034 A1 WO 2025017034A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- concentration
- antibody formulation
- formulation
- liquid antibody
- liquid
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39591—Stabilisation, fragmentation
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/08—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
- A61K47/10—Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/16—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
- A61K47/18—Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
- A61K47/183—Amino acids, e.g. glycine, EDTA or aspartame
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/26—Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2866—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
-
- C—CHEMISTRY; METALLURGY
- C08—ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
- C08G—MACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
- C08G65/00—Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
- C08G65/02—Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
- C08G65/26—Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring from cyclic ethers and other compounds
- C08G65/2603—Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring from cyclic ethers and other compounds the other compounds containing oxygen
-
- C—CHEMISTRY; METALLURGY
- C08—ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
- C08L—COMPOSITIONS OF MACROMOLECULAR COMPOUNDS
- C08L71/00—Compositions of polyethers obtained by reactions forming an ether link in the main chain; Compositions of derivatives of such polymers
- C08L71/02—Polyalkylene oxides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/08—Solutions
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/21—Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
-
- C—CHEMISTRY; METALLURGY
- C08—ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
- C08G—MACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
- C08G2650/00—Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
- C08G2650/28—Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule characterised by the polymer type
- C08G2650/58—Ethylene oxide or propylene oxide copolymers, e.g. pluronics
Definitions
- the present invention relates to a liquid antibody formulation
- a liquid antibody formulation comprising an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0.
- the invention further relates to a method for the preparation thereof and use of the liquid antibody formulation in the treatment, prevention or amelioration of any disease or disorder associated with IL-4 activity, including diseases or disorders mediated by activation of IL-4RC, such as atopic dermatitis (AD), e.g. moderate to severe atopic dermatitis, atopic eczema, asthma, e.g.
- AD topic dermatitis
- atopic dermatitis e.g. moderate to severe atopic dermatitis
- atopic eczema e.g.
- CRSwNP chronic rhinosinusitis with nasal polyposis
- CRsNP chronic rhinosinusitis without nasal polyps
- EoE eosinophilic esophagitis
- PN prurigo nodularis
- COPD chronic obstructive pulmonary disease
- CSU chronic spontaneous urticaria
- CINDU chronic inducible cold urticaria
- AFRS allergic fungal rhinosinusitis
- Biopharmaceuticals such as therapeutic proteins (e.g., antibodies) have gained significant attention in recent years in human and animal medicine and up to date several therapeutic antibodies have been approved for use in the treatment of various diseases.
- therapeutic proteins e.g., antibodies
- subcutaneous injection of therapeutic antibodies usually requires formulations containing high concentrations of proteins, ranging from, for example, 150 mg/mL up to 200 mg/mL in solution.
- concentration of antibody that can be accommodated by a certain formulation.
- a further consideration when formulating therapeutic antibodies is that in liquid solution the antibodies are prone to degradation, aggregation or undesired chemical modifications such as formation of acidic or basic species. Accordingly, liquid antibody solutions have to be formulated properly to ensure the antibodies maintain their stability during storage and subsequent use, but also under stress conditions such as shaking or temperature variations which might occur upon shipment, as well as planned or unplanned freezing.
- liquid antibody formulations relate to the viscosity of the solution, but also to the visual quality of the formulation.
- a further problem of formulations of therapeutic proteins involves degradation of the therapeutic proteins resulting in fragmentation products which are also referred to as low molecular weight species.
- therapeutic proteins under storage conditions can form unfavorable acidic or basic variants, concomitant with a decrease of the main peak.
- formulations of therapeutic proteins should be adjusted to an osmolality of body fluid to avoid pain and tissue damage during application, targeting to be below 400 mOsmol/kg, preferably close to 300 mOsmol/kg.
- a liquid antibody formulation is, thus, required to contain an adequate concentration of antibody, to possess a suitable viscosity, to remain physically and chemically stable during storage and subsequent use, and to enable convenient administration to patients.
- Common art to generate a stable formulation of high concentrated proteins of up to 200 mg/mL is to add one excipient to the formulation to stabilize against physicochemical degradation, typically a sugar or sugar alcohol, and an additional viscosity reducing agent.
- One exemplary therapeutic antibody which is approved for subcutaneous application and formulated as a liquid formulation comprising a sugar and L-arginine as an additional viscosity reducing agent is the anti-IL-4Ra antibody dupilumab (CAS Registry Number 1190264-60-8).
- the recombinant, human, monoclonal IgG4 antibody dupilumab is specifically directed against the interleukin (IL)-4Ra subunit present at type I and II receptors on immune cells such as B or T cells and thus inhibits the reaction cascades triggered via interleukin-4 and interleukin- 13 (IL-4 and IL- 13).
- IL-4 and IL- 13 interleukin- 13
- Dupilumab thus has an antiphlogistic and immunosuppressive effect in diseases whose pathophysiology is based on overactive IL- 4 and IL- 13 signaling.
- a liquid pharmaceutical formulation comprising dupilumab is described in EP 2 624 865 BL
- An especially preferred liquid pharmaceutical formulation comprises (i) dupilumab at a concentration up to 200 mg/mL, (ii) acetate at a concentration of 12.5 mM ⁇ 2 mM; (iii) histidine at a concentration of 20 mM ⁇ 3 mM; (iv) sucrose at a concentration of 5% w/v ⁇ 0.75% w/v; (v) polysorbate 20 or polysorbate 80 at a concentration of 0.2% w/v ⁇ 0.03% w/v; and (vi) arginine at a concentration of 25 mM ⁇ 3.75 mM or at a concentration of 50 mM ⁇ 7.5 mM; wherein the formulation has a pH of 5.6 to 6.2.
- the anti-hIL-4Ra antibody i.e. dupilumab
- sucrose as a thermal stabilizer
- arginine as a viscosity enhancer
- US 2023/348532 Al which generally relates to methods for manufacturing high titer antibody products, e.g. dupilumab, discloses in a side aspect excipient buffer solutions to achieve a certain target concentration of the antibody such as 150 mg/mL or 175 mg/mL. However, no final drug formulations are described.
- a liquid pharmaceutical formulation comprising dupilumab in accordance with EP 2 624 865 Bl is currently marketed under the tradename Dupixent® as a solution for injection containing dupilumab at a concentration of 150 mg/mL for dosages of 100 mg and 300 mg or 175 mg/mL for dosages of 200 mg, and further comprising arginine hydrochloride at a concentration of 25 mM and 50 mM, respectively, histidine, histidine hydrochloride monohydrate, polysorbate 80 (E 433), sodium acetate trihydrate, acetic acid 99% (E 260), sucrose, and water for injection, wherein the formulation has a pH of about 5.9 (Strickley R.G. and Lambert W.J. A review of Formulations of Commercially Available Antibodies. J Pharm Sci. 2021 Jul;110(7):2590-2608.).
- Dupixent® as an approved medication satisfies the requirements of sufficiently low viscosity
- enhanced storage stability of a liquid formulation comprising dupilumab is desirable. In that respect, it is particularly desirable that a high proportion of dupilumab remains in its native state upon storage. Further, it is desirable that a single formulation fits all dosages and devices to be marketed.
- a further object of the present invention is the provision of a stable liquid antibody formulation, preferably a stable liquid dupilumab formulation, having advantageous, e.g. enhanced physical stability, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting sufficiently low aggregation propensity, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, having a sufficiently low proportion of high molecular weight species (HMWS), e.g. upon storage and/or under stress conditions.
- HMWS high molecular weight species
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, wherein the hydrodynamic radius of the antibody is maintained, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, wherein the inter- molecular attractive forces are advantageously low, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting sufficiently low fragmentation propensity, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, having a sufficiently low proportion of fragments/low molecular weight species (LMWS), e.g. upon storage and/or under stress conditions.
- LMWS fragments/low molecular weight species
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, having improved, e.g. enhanced chemical stability, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting an acceptable, i.e. stabilized, main peak of dupilumab, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting an acceptable, i.e. reduced, formation of acidic variants, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting an acceptable, i.e. reduced, formation of basic species, e.g. upon storage and/or under stress conditions.
- a further object of the present invention is the provision of a stable liquid formulation of a therapeutic antibody, preferably dupilumab, having acceptable solution viscosity and exhibiting an osmolality close to an osmolality of body fluid, i.e. near 300 mOsmol/kg.
- a further object of the present invention is the provision of a stable liquid formulation of a therapeutic antibody, preferably dupilumab, which is particularly suited for drug approval.
- Another object of the present invention is the provision of a liquid formulation of a therapeutic antibody, preferably dupilumab, meeting the requirements for marketing authorization as a biosimilar.
- a formulation containing as little excipients as possible e.g. the use of further excipients like stabilizers, e.g. sugars, sugar alcohols, other amino acids such as methionine or proline should be avoided.
- further excipients like stabilizers, e.g. sugars, sugar alcohols, other amino acids such as methionine or proline should be avoided.
- sucrose should be avoided.
- a further object of the present invention is the provision of a single stable liquid formulation of a therapeutic antibody, preferably dupilumab, which is suited for using the same concentration (e.g. 150 mg/mL or 175 mg/mL) in different antibody dosages (e.g. 100 mg, 200 mg or 300 mg) and/or in different application forms (e.g. a pre-filled syringe, autoinjector, pre-filled pen, pre-filled cartridge for on-body injector, vial and microinfuser).
- a therapeutic antibody preferably dupilumab
- a high concentration of L-arginine of from 150 to 300 mM in a liquid antibody formulation comprising an antibody, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0, not only allows to lower the viscosity of the formulation, but also minimizes the formation of aggregates, while, surprisingly, at the same time less degradation products (i.e. fragments) are formed and less undesired chemical modifications, such as charge variants, are observed, thus resulting in a liquid antibody formulation exhibiting acceptable viscosity and improved storage stability and, thus, extended shelf-life.
- EP 2 624 865 Bl discloses that high amounts of arginine may cause stability issues, and particularly suggests including arginine only at a concentration of 25 mM or at a concentration of 50 mM. This recommendation was later put into practice in the authorized product Dupixent®.
- a liquid antibody formulation comprising high concentrated dupilumab and further comprising arginine at a concentration beyond 100 mM might solve the above-mentioned problems, e.g. exhibit minimized formation of aggregates, and particularly reduced formation of degradation products (i.e. fragments) and charge variants, while having acceptable viscosity and enhanced stability upon long- term storage and use as well as further advantages set forth below.
- the present invention relates to a liquid antibody formulation
- a liquid antibody formulation comprising (a) an antibody, preferably dupilumab or an antibody having at least 90% sequence identity with dupilumab, (b) L-arginine at a concentration of from 150 to 300 mM, (c) a surfactant, and (d) optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0.
- hydrodynamic radius refers to the Stokes radius or effective hydrated radius in solution. Degradation products of samples can be either fragments with smaller radii than the radius of the monomer (e.g of a therapeutic protein such as dupilumab), or aggregates with higher radii. Maintenance of the hydrodynamic radius is a parameter demonstrating colloidal stability, a periodical determination by e.g. dynamic light scattering (DLS) of a sample might demonstrate particles aggregating over time by increasing the hydrodynamic radius of the particles.
- DLS dynamic light scattering
- the hydrodynamic radius of the antibody molecules preferably dupilumab
- the hydrodynamic radius of the antibody molecules is maintained in the liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM.
- Dupilumab is thus prevented from aggregation during storage and/or under stress conditions, leading to improved colloidal stability.
- colloidal stability generally relates to particle size change (e.g. aggregation or agglomeration) of particles in a dispersion, e.g. of antibody molecules in a liquid antibody formulation. If particles are not subject to size variation, the dispersion is considered as having colloidal stability. Colloidal stability can depend on several types of interactions such as van der Waals and electrostatic interactions, steric interactions (e.g. polymer adsorption), and hydrophobic effect, which renders it difficult to theoretically predict the colloidal stability of a dispersion.
- the ko value i.e. diffusion interaction parameter
- DLS dynamic light scattering
- the present invention it was surprisingly found that protein-protein attractive forces are reduced in the liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM, thus preventing dupilumab from aggregation during storage and/or under stress conditions and leading to improved colloidal stability.
- intermolecular forces are sufficiently repulsive for preventing dupilumab from aggregation during storage and/or under stress conditions and leading to improved colloidal stability.
- the liquid antibody formulation of the present invention exhibits enhanced physical stability, including stability against aggregation (i.e. reduced HMWS formation) and against degradation/fragmentation (i.e. reduced LMWS formation), and at the same time enhanced chemical stability including stabilization of the main peak of the antibody, lower increase of acidic and/or basic species.
- physical stability can refer to stability against aggregation (i.e. reduced HMWS formation) of a therapeutic antibody and against degradation/fragmentation (i.e. reduced LMWS formation) of a therapeutic antibody.
- HMWS high molecular weight species
- SE-HPLC size-exclusion chromatography
- LMWS low molecular weight species
- SE-HPLC size-exclusion chromatography
- Both, HMWS and LMWS, can be indicative of the content of impurities of the liquid antibody formulation.
- liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM exhibits advantageous low formation of HMWS and LMWS. Therefore, the liquid antibody formulation has advantageous high stability against aggregation and fragmentation/degradation and, thus desirable, e.g. enhanced physical stability, e.g. upon storage and/or under stress conditions.
- chemical stability can refer to stability against undesired chemical modifications such as the formation of acidic and/or basic species of a therapeutic antibody.
- Acidic or basic species which are also referred to as “charge variants”, are defined as the sum of the antibody (e.g. dupilumab) peaks that elute from a cation exchange (CEX-HPLC) column with earlier or later retention times than the main peak, respectively.
- charge variants are defined as the sum of the antibody (e.g. dupilumab) peaks that elute from a cation exchange (CEX-HPLC) column with earlier or later retention times than the main peak, respectively.
- CEX-HPLC cation exchange
- Both, acidic and basic species, are indicative of the content of impurities of the liquid antibody formulation, whereas the main peak is an indicator of the purity of the liquid antibody formulation.
- the change i.e. the “DELTA” or “A” between the main peak, the peak for the acidic species and the peak for the basic species, respectively, is determined in the formulation as prepared, i.e. at to, and at a later point in time.
- the three absolute DELTAs between (1) the main peak at to and the main peak at tiater point in time, and (2) the peak for the acid species at to and the peak for the acidic species at hater point in time, and (3) the peak for the basic species at to and the peak for the acid species at tiater point in time are calculated.
- the absolute DELTA relative area is 10%.
- the absolute DELTA relative area is 5%.
- Those three absolute DELTAs can be considered as an indicator for the stability of the antibody in a specific formulation.
- a liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM exhibits a reduced formation of acidic and basic species and, concomitantly has a stabilized main peak of the antibody, e.g. dupilumab, e.g. upon storage and/or under stress conditions. Therefore, the liquid antibody formulation has desirable, e.g. enhanced chemical stability.
- a liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM surprisingly exhibits improved physical stability and/or improved chemical stability.
- these desirable properties can be achieved while maintaining the remaining desirable properties of the formulation, e.g. maintaining the desirable level of osmolality and viscosity.
- EP 2 624 865 Bl provides data in Table 3 suggesting that inclusion of 5% arginine in a liquid antibody formulation comprising dupilumab might reduce formation of acidic species, but at the same time induces elevated levels of aggregation and enhanced formulation of basic species.
- EP 2624865 B 1 it could not have been expected that improved physical stability in combination with improved chemical stability might be achieved by including L- arginine in a liquid antibody formulation comprising dupilumab.
- the present invention provides for liquid formulations of high concentrated antibodies, particularly high concentrated dupilumab, exhibiting optimized stability and, hence, enhanced stability upon long-term storage and/or under stress conditions.
- the liquid antibody formulation of the present invention can exhibit high levels of stability.
- a liquid antibody formulation of the present invention is considered stable, if one of the following stability criteria (1) to (6) is fulfilled.
- Stability can be measured, for example, by determining the percentage of native antibody that remains in the formulation after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions.
- the percentage of native antibody can be determined, for example, by size exclusion chromatography, e.g., size exclusion high performance liquid chromatography (SE- HPLC).
- SE- HPLC size exclusion high performance liquid chromatography
- the percentage of native antibody can be determined, for example, by non-reducing capillary electrophoresis (nRCE-SDS or non-reducing cGE).
- stable means that the antibody within the liquid antibody formulation, preferably dupilumab, remains in its native form or retains an acceptable degree of chemical structure or biological function upon storage and/or under stress conditions.
- a formulation may be stable even though the antibody contained therein does not remain 100% native or maintain 100% of its chemical structure or biological function upon storage and/or under stress conditions.
- a liquid antibody formulation may be regarded as “stable”, if about 90%, about 95%, about 96%, about 97%, about 98% or about 99% of the antibody’s structure or function are maintained upon storage and/or under stress conditions may.
- a liquid antibody formulation may be regarded as “stable”, if about 90%, about 95%, about 96%, about 97%, about 98% or about 99% of the antibody are maintained in its native form upon storage and/or under stress conditions.
- At least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of the native form of the antibody, preferably dupilumab, can be detected in the formulation upon storage and/or under stress conditions.
- storage conditions may refer to at least 1 week, at least 2 weeks (i.e. at least 0.5 month), at least 3 weeks, at least 4 weeks, at least 1 month, at least
- the term “long-term storage” may refer to a storage period of at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, at least 18 months, at least 24 months, at least 36 months, or more.
- stress conditions may refer to storage of at least 1 week, at least 2 weeks (i.e. at least 0.5 month), at least 3 weeks, at least 4 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or at least 6 months at room temperature such as 25 °C ⁇ 2°C / 60% relative humidity (RH) ⁇ 5% RH or at 40°C ⁇ 2°C/75% RH ⁇ 5% RH.
- stress conditions may also refer to storage with mechanical stressing (e.g. orbital shaking or overhead rotation (OH)) overhead rotation for 24 h at 25°C/300 rpm.
- stress conditions may also refer to application of at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 freeze/thaw cycles of -80°C/room temperature such as +25 °C.
- the liquid antibody formulation of the invention is deemed stable, if at least 97% or at least 98% or at least 99% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 40°C, as determined by SE-HPLC.
- the delta of the percentage of native antibody between to and ti month at 4o°c is less than 3% or less than 2% or less than 1%, as determined by SE-HPLC.
- the liquid antibody formulation of the invention is deemed stable, if at least 98% or at least 99% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 25°C, or after 3 months of storage at 5°C, or after
- the delta of the percentage of native antibody between to and t3 months at 5°c or between to and t3 months at 25°c or between to and ts months at 5°c or between to and ts months at 25 °C or between to and to months at 5°c is less than 2% or less than 1%, as determined by SE-HPLC.
- the liquid antibody formulation of the invention is deemed stable, if at least 95% or at least 96% or at least 97% or at least 98% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 40°C, as determined by nRCE-SDS.
- the delta of the percentage of native antibody between to and ti month at 4o°c is less than 5% or less than 4% or less than 3% or less than 2%, as determined by nRCE-SDS.
- the liquid antibody formulation of the invention is deemed stable, if at least 97% or at least 98% or at least 99% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 25 °C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by nRCE- SDS.
- the delta of the percentage of native antibody between to and t3 months at 5°c or between to and t3 months at 25°c or between to and ts months at 5°c or between to and ts months at 25°c or between to and to months at 5°c is less than 3% or less than 2% or less than 1%, as determined by nRCE-SDS.
- At least 90% or at least 95% or at least 96% or at least 97% or at least 98% of the native form of the antibody, preferably dupilumab is recovered from the liquid antibody formulation after one month of storage at 40°C or after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by size exclusion chromatography.
- Stability can be measured by determining the percentage of antibody that forms aggregates (i.e. HMWS) within the liquid antibody formulation after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the proportion of aggregates that is formed.
- the percentage of aggregated antibody can be determined by, for example, size exclusion chromatography, e.g., size exclusion high performance liquid chromatography (SE-HPLC).
- stable as used herein in reference to the liquid antibody formulation, usually means that at most 5% of the antibody is in an aggregated form detected in the formulation upon storage and/or under stress conditions. Under certain circumstances, a liquid antibody formulation may be regarded as “stable”, if at most about 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of the antibody can be detected in an aggregated form in the formulation upon storage and/or under stress conditions.
- less than 3% or less than 2% of the antibody which is recovered from the liquid antibody formulation after one month of storage at 40°C or after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, is aggregated, as determined by SE-HPLC.
- Stability can be measured by determining the percentage of antibody that degrades to form fragments (i.e. LMWS) within the liquid antibody formulation after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the proportion of fragments that is formed.
- the percentage of fragmented antibody can be determined by, for example, by non-reducing capillary electrophoresis (nRCE-SDS or non-reducing cGE).
- stable as used herein in reference to the liquid antibody formulation, usually means that at most 5% of the antibody is in a fragmented form detected in the formulation upon storage and/or under stress conditions. Under certain circumstances, a liquid antibody formulation may be regarded as “stable”, if at most about 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of the antibody can be detected in a fragmented form in the formulation upon storage and/or under stress conditions.
- less than 5% or less than 4% or less than 3% or less than 2% of the antibody which is recovered from the liquid antibody formulation after one month of storage at 40°C or after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, is fragmented, as determined by nRCE-SDS. 4) High proportion of antibody in the main peak/main fraction
- Stability can be measured by determining the percentage of antibody remaining in the main fraction/main peak of antibody during ion exchange after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is proportional to the fraction of antibody in the main fraction/main peak.
- the percentage of antibody in the main fraction/main peak can be determined, for example, by ion exchange chromatography, e.g., cation exchange high performance liquid chromatography (CEX-HPLC) or CZE (capillary zone electrophoresis).
- stable means that the antibody, preferably dupilumab, when measured by an appropriate analytical method such as by capillary zone electrophoresis, demonstrates a loss of main peak area relative to the main peak area at to (i.e. Amain peak) of less than 20% or less than 15% or less than 14% or less than 13% or less than 12% or less than 11% or less than 10% or less than 9% or less than 8% or less than 7% or less than 6% or less than 5% after the liquid antibody formulation has undergone a stress condition and/or storage conditions.
- the liquid antibody formulation demonstrates a loss of main peak area relative to the main peak area at to (i.e. Amain peak) of less than 15% or less than 13% or less than 11% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C, as determined by CZE.
- the liquid antibody formulation demonstrates a loss of main peak area relative to the main peak area at to (i.e. Amain peak) of less than 5% or less than 4% or less than 3% or less than 2% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by CZE.
- Stability can be measured by determining the percentage of antibody that migrates in a more acidic fraction during capillary zone electrophoresis (“acidic form”, “acidic species”) than in the main fraction/main peak of antibody (“neutral conformation”) after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the fraction of antibody in the acidic form.
- the percentage of “acidified” or “deamidated” antibody, i.e. acidic species can be determined, for example, by capillary zone electrophoresis (CZE) or ion exchange chromatography, e.g., cation exchange high performance liquid chromatography (CEX-HPLC).
- stable means that the antibody, preferably dupilumab, when measured by an appropriate analytical method such as by capillary zone electrophoresis, demonstrates an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. A ac idic species peak) of less than 20% or less than 15% or less than 14% or less than 13% or less than 12% or less than 11% or less than 10% or less than 9% or less than 8% or less than 7% or less than 6% or less than 5% after the liquid antibody formulation has undergone a stress condition and/or storage conditions.
- the liquid antibody formulation demonstrates an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. A ac idic species pea k) of less than 15% or less than 13% or less than 11% or less than 10% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C, as determined by CZE.
- the liquid antibody formulation demonstrates an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. A ac idic species pea k) of less than 5% or less than 4% or less than 3% or less than 2% or less than 1% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5 °C, as determined by CZE.
- a ac idic species pea k an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. A ac idic species pea k) of less than 5% or less than 4% or less than 3% or less than 2% or less than 1% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after
- Stability can be measured by determining the percentage of antibody that migrates in a more basic fraction during ion exchange (“basic form”, “basic species”) than in the main fraction/main peak of antibody (“neutral conformation”) after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the fraction of antibody in the basic form.
- the percentage of “basified” antibody, i.e. basic species can be determined, for example, by ion exchange chromatography, e.g., cation exchange high performance liquid chromatography (CEX-HPLC) or capillary zone electrophoresis (CZE).
- stable means that the antibody, preferably dupilumab, when measured by an appropriate analytical method such as by capillary zone electrophoresis, demonstrates an increase of peak area of basic species relative to the peak area of basic species at to (i.e. Abasic species peak) of less than 5% or less than 4% or less than 3% or less than 2% or less than 1% or less than 0.9% or less than 0.8% or less than 0.7% or less than 0.6% or less than 0.5% after the liquid antibody formulation has undergone a stress condition and/or storage conditions.
- the liquid antibody formulation demonstrates an increase of peak area of basic species relative to the peak area of basic species at to (i.e. Abasic species peak) of less than 3%, less than 2% or less than 1% or less than 0.8% or less than 0.5% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C, as determined by CZE.
- the liquid antibody formulation demonstrates an increase of peak area of basic species relative to the peak area of basic species at to (i.e. Abasic species peak) of less than 2% or less than 1% or less than 0.8% or less than 0.5% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by CZE.
- a liquid antibody formulation may be considered stable if, after 6 or more months of storage at about 5°C to about 25°C, the change in OD405 of the formulation is less than about 0.05 (e.g., 0.04, 0.03, 0.02, 0.01, or less) from the OD405 of the formulation at time zero.
- DSC differential scanning calorimetry
- Stability may also be assessed by measuring the biological activity or binding affinity of the antibody to its target.
- a formulation may be regarded as stable if, after storage at 5°C etc. for a defined period of time of at least six months (e.g., 6 to 12 months), the antibody contained within the formulation, preferably dupilumab, binds to IL-4Ra with an affinity that is at least 90%, 95%, or more of the binding affinity of the antibody prior to said storage. Binding affinity may be determined by e.g., ELISA or plasmon resonance.
- Biological activity may be determined by an IL-4Ra activity assay, such as e.g., contacting a cell that expresses IL-4Ra with the formulation comprising the anti IL- 4Ra antibody.
- the binding of the antibody to such a cell may be measured directly, such as e.g., via FACS analysis.
- the downstream activity of the IL-4Ra system may be measured in the presence of the antibody, preferably dupilumab, and an IL-4Ra agonist, and compared to the activity of the IL-4Ra system in the absence of antibody.
- the IL-4Ra may be endogenous to the cell.
- the IL-4Ra may be ectopically expressed in the cell.
- the liquid antibody formulation of the present invention further generally exhibits desirable low solution viscosity and, thus, can be suitable to be administered by subcutaneous injection e.g. using thin needles for injection which is associated with reduced pain during injection.
- a liquid antibody formulation having a “sufficiently low viscosity” or a “suitable viscosity” or an “acceptable viscosity” will exhibit an absolute viscosity of less than 15 mPa-s, or less than 13 mPa-s, or from 10 to 13 mPa-s or from 10 to 12.5 mPa-s, wherein the shear viscosity is measured by rotational rheometry at 20°C.
- a liquid antibody formulation containing an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, optionally a buffer, and a surfactant, and having a pH of from 5.0 to 7.0 exhibits sufficiently low viscosity and sufficiently high stability without requiring a further viscosity reducing agent or a further stabilizing agent.
- an antibody preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM
- a buffer optionally a buffer, and a surfactant, and having a pH of from 5.0 to 7.0
- the liquid antibody formulation of the present invention comprises an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant and optionally a buffer, and has a pH of from 5.0 to 7.0, wherein the formulation does not comprise sucrose and/or treahlose and/or methionine and/or proline, especially the formulation does not comprise sucrose.
- liquid antibody formulation of the present invention generally has an osmolality that is physiologically compatible.
- a “physiologically compatible” or “physiologically isotonic” osmolality refers to an osmolality of body fluid of about 300 mOsmol/kg, which allows to avoid pain and tissue damage during application.
- the liquid antibody formulation has an osmolality of 200 to 400 mOsm/kg, preferably 250 to 350 mOsmol/kg, preferably 270 to 330 mOsm/kg, more preferably 290 ⁇ 20 mOsm/kg.
- the visual appearance of the liquid antibody formulation of the present invention met the acceptance criteria, being slightly yellowish, opalescent and more viscous than water.
- the visual appearance was assessed using an inspection light box equipped with non-flickering fluorescent lamps and a black and a white background plate (Portable Inspection Hood MIH-PORT, Bosch). The samples were evaluated in their storage container without magnification to assess sample appearance in respect to visible particles, color and clarity.
- the liquid antibody formulation of the present invention requires less excipients for obtaining a stable formulation.
- the liquid antibody formulation of the present invention does not require a sugar.
- the liquid antibody formulation of the present invention does not require a sugar alcohol.
- the liquid antibody formulation of the present invention does not require amino acids other than L- arginine, i.e. the liquid antibody formulation of the present invention does not require amino acids such as methionine or proline.
- a surfactant selected from the group consisting of polyoxyethylene(20)sorbitan monolaurate, polyoxy- ethylene(20)sorbitan mono
- a particularly stable liquid antibody formulation preferably a particularly stable liquid dupilumab formulation is obtained.
- Said stable liquid antibody formulation, preferably dupilumab formulation is particularly suitable for drug approval.
- Concerning suitability for drug approval it is particularly advantageous that the liquid antibody formulation, preferably the liquid dupilumab formulation, is particularly stable and requires only a minimum number of excipients.
- the liquid antibody formulation comprises (a) an antibody.
- antibody refers to monoclonal or polyclonal antibodies.
- the term “antibody” includes but is not limited to recombinant antibodies that are generated by recombinant technologies as known in the art.
- the term “antibody” includes antibodies of any species, in particular of mammalian species; such as human antibodies of any isotype, including IgAl, lgA2, IgD, IgGl, lgG2a, lgG2b, lgG3, lgG4, IgE and IgM, as well as modified variants thereof; non-human primate antibodies, e.g.
- antibody also refers to “chimeric” antibodies in which a first portion of at least one heavy and/or light chain antibody sequence is from a first species and a second portion of the heavy and/or light chain antibody sequence is from a second species.
- antibody further includes antigen-binding portions or antigenbinding fragments of an antibody.
- antigen-binding fragment also refers to an antibody that comprises at least one heavy or light chain immunoglobulin domain as known in the art and binds to one or more antigen(s).
- antibody fragments that can be used as biologies include Fab, Fab', F(ab')2, and Fv and scFv fragments; as well as diabodies, triabodies, tetrabodies, minibodies, domain antibodies, single-chain antibodies, bispecific, trispecific, tetraspecific or multispecific antibodies formed from antibody fragments or antibodies, including but not limited to Fab-Fv constructs.
- the antibody is a vNAR, a camelid antibody, a VHH antibody, or an antigenbinding portion thereof.
- Antibody fragments as defined above are known in the art.
- the antibody is an IgGl, IgG2, IgG3 or IgG4 antibody. In some embodiments, the antibody is an IgG4 antibody. In some embodiments the antibody is a human antibody, a humanized antibody, or a chimeric antibody.
- the antibody is an anti-interleukin-4 receptor alpha (anti-IL-4Ra) antibody.
- anti-IL-4Ra anti-interleukin-4 receptor alpha
- the antibody is dupilumab, i.e. dupilumab having CAS Registry Number 1190264-60-8.
- the antibody can be an antibody having at least 90% sequence identity with dupilumab having CAS Registry Number 1190264-60-8.
- the antibody included in the liquid antibody formulation of the present invention is dupilumab.
- the liquid antibody formulation comprises the antibody, preferably dupilumab, at a concentration of from 150 to 200 mg/mL, preferably at a concentration of 150 mg/mL or 175 mg/mL.
- liquid antibody formulations of the present invention are particularly suitable for parenteral application including intradermal, subcutaneous, intramuscular, intraosseous, intraperitoneal, and intravenous administration. Therefore, the liquid antibody formulations of the present invention can be formulated as dosage forms, particularly as dosage forms for parenteral application, e.g. dosage forms for intradermal, subcutaneous, intramuscular, intraosseous, intraperitoneal, and intravenous administration.
- the dosage form containing the liquid antibody formulation of the present invention.
- the dosage form comprises an application device (e.g. as further described below) said device containing the antibody formulation of the present invention.
- the dosage form contains the antibody formulation, wherein the antibody, preferably dupilumab, is present in the dosage range from 50 mg to 500 mg, in particular at dosages of 100 mg or 200 mg or 300 mg.
- the dosage form contains the antibody formulation, wherein the antibody, preferably dupilumab, is present at a concentration of 150 mg/mL and at dosages of 100 mg or 200 mg or 300 mg.
- the dosage form contains the antibody formulation, wherein the antibody, preferably dupilumab, is present at a concentration of 175 mg/mL and at dosages of 100 mg or 200 mg or 300 mg.
- the liquid antibody formulation comprises (b) the amino acid L-arginine (also referred to as “arginine” herein) at a concentration of from 150 mM to 300 mM.
- amino acid refers to one of the 20 known amino acids including alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, L-histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine. All amino acids except for glycine are stereoisomers (mirror images of their structure). These are labelled L-amino acids and D-amino acids to distinguish. Whenever any of the amino acids are mentioned herein, it is meant as L-amino acid.
- L-arginine is meant and so on.
- an individual amino acid is mentioned, it encompasses its derivatives.
- L-arginine is used, L-arginine HC1 is encompassed.
- the liquid antibody formulation comprises L-arginine at a concentration of from 150 mM, 155 mM, 160 mM, 165 mM, 170 mM up to 300 mM, 295 mM, 290 mM, 285 mM, 280 mM, 275 mM, 270 mM, 265 mM, 260 mM, 255 mM, 250 mM, 245 mM, 240 mM, 235 mM, 230 mM.
- the liquid antibody formulation comprises L-arginine at a concentration from 150 mM to 290 mM, from 150 mM to 280 mM, from 150 mM to 270 mM, from 150 mM to 260 mM, from 150 mM to 250 mM, from 150 mM to 240 mM, from 150 mM to 230 mM, from 150 mM to 220 mM, from 150 mM to 210 mM, from 150 mM to 200 mM.
- the liquid antibody formulation comprises L-arginine at a concentration from 160 mM to 300 mM, from 170 mM to 300 mM, from 180 mM to 300 mM, from 190 mM to 300 mM, from 200 mM to 300 mM, from 210 mM to 300 mM, from 220 mM to 300 mM, from 230 mM to 300 mM, from 240 mM to 300 mM, from 250 mM to 300 mM, from 260 mM to 300 mM, from 270 mM to 300 mM, from 280 mM to 300 mM, from 290 mM to 300 mM.
- the liquid antibody formulation of the present invention comprises L-arginine at a concentration of from 150 to 250 mM, preferably at a concentration of from 150 to 230 mM, preferably at a concentration of from 170 to 250 mM, preferably at a concentration of from 170 to 230 mM, preferably at a concentration of 170 mM.
- the liquid antibody formulation of the present invention comprises L-arginine at a concentration of 170 mM. In a preferred embodiment, the liquid antibody formulation of the present invention comprises L-arginine at a concentration of 230 mM
- the liquid antibody formulation of the present invention further comprises (c) a surfactant.
- the surfactant comprises preferably a non-ionic surfactant.
- non-ionic surfactant means a surfactant that contains neither positively nor negatively charged functional groups. In contrast to anionic and cationic surfactants, non- ionic surfactants do not ionize in solution.
- the non-ionic surfactant can be a poloxamer or a polysorbate.
- Poloxamers are non-ionic triblock copolymers composed of a central hydrophobic chain of poly(propyleneoxide) flanked by two hydrophilic chains of polyethylene oxide).
- the length of the polymer blocks can be customized, leading to different poloxamers with slightly different properties.
- the non-ionic surfactant can be Pluronic F127 (poloxamer 407), Pluronic F123 (poloxamer 403), Pluronic F-68 (poloxamer 188), Pluronic P123, Pluronic P85, or other polyethylene oxide-polypropylene oxide (EO-PO) block copolymers of greater than 3,000-4,000 MW or combinations thereof.
- poloxamer 188 as defined according to Fiedler Lexikon der Hilfsstoffe fur Pharmazie, Kosmetik und angrenzende füre, 5 th edition, volume 2, 2002), i.e. a polyoxyethylene-polyoxypropylene-block polymer which is also referred to as poloxamer 188.
- Polysorbates are composed of ethylene oxide and sorbitan fatty acid esters. These emulsifiers are much more widely known as polysorbates, e.g., polysorbate 20, 60, and 80. Polysorbate 20, 60, and 80 utilize laurate, stearate, and oleate, respectively, for the fatty acid portion of the molecule.
- the liquid antibody formulation comprises a polysorbate. In some embodiments, the liquid antibody formulation comprises polyoxyethylene(20)sorbitan monolaurate (i.e. polysorbate 20). In some embodiments, the liquid antibody formulation comprises polyoxyethylene(20)sorbitan monostearate (i.e. polysorbate 60). In some embodiments, the liquid antibody formulation comprises polyoxyethylene(20)sorbitan monooleate (i.e. polysorbate 80).
- the concentration of surfactant can be reduced, while maintaining the properties required for obtaining marketing authorization as a biosimilar.
- the liquid antibody formulation comprises a polysorbate at a concentration of from 0.1 to 4.0 mg/mL, from 0.2 to 3.5 mg/mL, from 0.5 to 3.0 mg/mL, from 1.0 to 2.5 mg/mL, from 1.5 to 2.0 mg/mL, such as 0.2 mg/mL or 2.0 mg/mL.
- the liquid antibody formulation comprises a polysorbate at a concentration of from 1.0 to 3.0 mg/mL.
- the liquid antibody formulation comprises a polysorbate at a concentration of from 1.5 to 2.5 mg/mL.
- the liquid antibody formulation comprises a polysorbate at a concentration of 0.2 mg/mL or 2.0 mg/mL.
- the liquid antibody formulation comprises polysorbate 20 at a concentration of from 0.1 to 4.0 mg/mL, from 0.2 to 3.5 mg/mL, from 0.5 to 3.0 mg/mL, from 1.0 to 2.5 mg/mL, from 1.5 to 2.0 mg/mL, such as 0.2 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of 0.2 mg/mL or 2.0 mg/mL.
- the liquid antibody formulation comprises polysorbate 80 at a concentration of from 0.1 to 4.0 mg/mL, from 0.2 to 3.5 mg/mL, from 0.5 to 3.0 mg/mL, from 1.0 to 2.5 mg/mL, from 1.5 to 2.0 mg/mL, such as 0.2 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a concentration of 0.2 mg/mL or 2.0 mg/mL.
- the liquid antibody formulation comprises a poloxamer at a concentration of from 0.1 to 4.0 mg/mL, from 0.5 to 3.5 mg/mL, from 1.0 to 3.0 mg/mL, from 1.5 to 2.5 mg/mL, such as 0.5 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of 0.5 mg/mL or 2.0 mg/mL.
- the liquid antibody formulation comprises poloxamer 188 at a concentration of 0.1 to 4.0 mg/mL, from 0.5 to 3.5 mg/mL, from 1.0 to 3.0 mg/mL, from 1.5 to 2.5 mg/mL, such as 0.5 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of 0.5 mg/mL or 2.0 mg/mL.
- the surfactant included in the liquid antibody formulation of the present invention is polyoxyethylene(20)sorbitan monolaurate, also referred to as polysorbate 20 or polyoxyethylene(20)sorbitan monooleate, also referred to as polysorbate 80, or a polyoxyethylene-polyoxypropylene-block polymer, preferably a polyoxy ethylene-polyoxypropylene-block polymer of the following formula (I) wherein x is an integer from 5 to 146, y is an integer from 18 to 60, and z is an integer from 5 to 146.
- formula (I) wherein x is an integer from 5 to 146, y is an integer from 18 to 60, and z is an integer from 5 to 146.
- the surfactant is polyoxyethylene(20)sorbitan monooleate, also referred to as polysorbate 80.
- the liquid antibody formulation comprises the surfactant at a concentration of from 0.1 to 4.0 mg/mL, preferably at a concentration of 0.2 mg/mL or 0.5 mg/mL or 2.0 mg/mL.
- the surfactant is polyoxyethylene(20)sorbitan monooleate (i.e. polysorbate 80) at a concentration of 0.2 mg/mL or 2.0 mg/mL.
- the surfactant is polyoxyethylene(20)sorbitan monooleate (i.e. polysorbate 80) at a concentration of 2.0 mg/mL.
- the liquid antibody formulation of the present invention optionally comprises (d) a buffer.
- buffer refers to a substance that allows a solution to maintain its pH or only marginally change its pH after addition of acidic or basic substances.
- the buffer is a single buffer. In some embodiments, the buffer may be a salt buffer or an amino acid buffer.
- the amino acid buffer is alanine, aspartic acid, glutamic acid, L- histidine, lysine, cysteine, tyrosine, or serine.
- component (b) L-arginine is not considered a buffer.
- a L-histidine buffer is used, i.e. L-histidine/L-histidine HC1. Said buffer is also referred to as “histidine” or “L-histidine” herein.
- the concentration of the amino acid buffer in the liquid antibody formulation may range from 5 mM to 20 mM. In some embodiments, the concentration of the amino acid buffer in the liquid antibody formulation is from 5 mM to 15 mM, such as 10 mM.
- the concentration of L-histidine in the liquid antibody formulation may range from 5 to 20 mM, or 5 mM to 15 mM, such as 10 mM.
- the concentration of L-histidine in the liquid antibody formulation is 10 mM.
- a salt buffer is used.
- the salt buffer system is citrate; adipate, bicarbonate; phosphate, succinate, or tartrate.
- a succinate buffer is used i.e. succinic acid/succinate.
- Said buffer is also referred to as “succinate” herein.
- the liquid antibody formulation does not comprise an acetate buffer.
- the concentration of the salt buffer in the liquid antibody formulation may range from 5 mM to 20 mM. In some embodiments, the concentration of the salt buffer in the liquid antibody formulation is from 5 mM to 15 mM, such as 10 mM.
- the concentration of succinate in the liquid antibody formulation may range from 2 mM to 25 mM, 5 mM to 20 mM or 5 mM to 10 mM. In some embodiments, the concentration of succinate in the liquid antibody formulation is 10 mM.
- a single buffer is used. In some embodiments a single buffer is used, which is an amino acid buffer. In some embodiments a single buffer is used, which is a salt buffer. In some embodiments the liquid antibody formulation comprises L-histidine as the buffer and comprises no other buffer. In some embodiments the liquid antibody formulation comprises succinate as the buffer and comprises no other buffer. In a preferred embodiment, the liquid antibody formulation comprises a buffer. Preferably, the buffer is L-histidine/L-histidine HC1 or succinic acid/succinate.
- the liquid antibody formulation comprises the buffer, preferably the L-histidine/L-histidine HC1 or succinic acid/succinate buffer at a concentration of from 5 to 20 mM, preferably at a concentration of 10 mM.
- the liquid antibody formulation comprises as the buffer succinic acid/succinate buffer at a concentration of from 5 to 20 mM, preferably at a concentration of 10 mM.
- the liquid antibody formulation of the present invention does not comprise an acetate buffer.
- the liquid antibody formulation of the present invention may further comprise (e) L-glycine.
- the liquid antibody formulation comprises L-glycine at a concentration of from 20 to 150 mM.
- the concentration of L-glycine in the liquid antibody formulation is from 20 to 150 mM, from 25 mM to 150 mM, from 30 mM to 150 mM, from 35 mM to 150 mM, from 40 mM to 150 mM, from 45 mM to 150 mM, from 50 mM to 150 mM, from 55 mM to 150 mM, from 60 mM to 150 mM, from 70 mM to 150 mM, from 75 mM to 150 mM, from 80 mM to 150 mM, from 90 mM to 150 mM, from 95 mM to 150 mM, from 100 mM to 150 mM.
- the liquid antibody formulation of the present invention may further comprise (f) a sugar alcohol.
- Sugars and sugar alcohols are used as common stabilizers in liquid antibody formulations.
- “Sugar”, as used herein, refers to monosaccharides, disaccharide sugars or polysaccharide sugars. Disaccharides form when two monosaccharides undergo a dehydration reaction (or a condensation reaction or dehydration synthesis). During this process, one monosaccharide's hydroxyl group combines with another monosaccharide's hydrogen, releasing a water molecule and forming a covalent bond. This is called a glycosidic bond. Glycosidic bonds (or glycosidic linkages) can be alpha or beta type.
- “Sugar alcohol”, as used herein, refers to organic compounds, typically derived from sugars, containing one hydroxyl group (-OH) attached to each carbon atom.
- Sugar alcohols have the general formula HOCH2(CHOH) n CH2OH.
- sugars have two fewer hydrogen atoms, for example H0CH2(CH0H) n CH0 or H0CH2(CH0H) n IC(0)CH20H.
- the sugar alcohols differ in chain length. Most have five- or six-carbon chains, because they are derived from pentoses (five-carbon sugars) and hexoses (six-carbon sugars), respectively. They have one -OH group attached to each carbon.
- sugar alcohols do not - although they can be dehydrated to give cyclic ethers (e.g., sorbitol can be dehydrated to isosorbide).
- a sugar is not necessary for a liquid antibody formulation.
- a sugar such as trehalose that comprises two monomer units is not necessary for a liquid antibody formulation. Since trehalose and sucrose are both non-reducing sugars commonly used as a stabilizer in polypeptide formulations, and since it is known that trehalose may even be better as compared to sucrose for polypeptide formulations, it was surprisingly found that sugars, in general are not required for a liquid antibody formulation.
- the liquid antibody formulation does not comprise a sugar comprising two monomer units. In some embodiments, the liquid antibody formulation does not comprise a sugar. In some embodiments, the liquid antibody formulation does not comprise a sugar and does not comprise a sugar alcohol.
- the liquid antibody formulation does not comprise a sugar comprising two monomer units but comprises a sugar alcohol. In some embodiments, the liquid antibody formulation does not comprise a sugar but comprises a sugar alcohol.
- the liquid antibody formulation does not comprise a sugar comprising two monomer units and does not comprise a sugar alcohol. In some embodiments, the liquid antibody formulation does not comprise a sugar and does not comprise a sugar alcohol.
- the liquid antibody formulation does not comprise a sugar but comprises a sugar alcohol.
- the sugar alcohol is present at a concentration of from 50 mM to 250 mM. In some embodiments, the sugar alcohol is present at a concentration of from 50 mM to 250 mM, from 50 mM to 200 mM, from 50 mM to 150 mM, from 50 mM to 130 mM. In some embodiments, the sugar alcohol is present at a concentration of from 50 mM to 150 mM. In some embodiments, the sugar alcohol is present at a concentration of 60 mM or 120 mM.
- the sugar alcohol is mannitol. In some embodiments, mannitol is present at a concentration of from 50 mM to 250 mM. In some embodiments, mannitol is present at a concentration of from 50 mM to 250 mM, from 50 mM to 200 mM, from 50 mM to 150 Mm, from 50 mM to 130 mM. In some embodiments, mannitol is present at a concentration of from 50 mM to 150 mM. In some embodiments, mannitol is present at a concentration of 60 mM or 120 mM.
- the sugar alcohol is sorbitol. In some embodiments, sorbitol is present at a concentration of from 50 mM to 250 mM. In some embodiments, sorbitol is present at a concentration of from 50 mM to 250 mM, from 50 mM to 200 mM, from 50 mM to 150 MM, from 50 mM to 130 mM. In some embodiments, sorbitol is present at a concentration of from 50 mM to 150 mM. In some embodiments, sorbitol is present at a concentration of 60 mM or 120 mM.
- the liquid antibody formulation does not comprise a sugar.
- the liquid antibody formulation does not comprise a sugar comprising more than one monomer unit.
- the liquid antibody formulation does not comprise a sugar alcohol.
- the peak area of HMWS in %, as measured by size exclusion chromatography is lower as compared to a liquid antibody formulation of the same pH or the same pH range comprising, (a) the same antibody, preferably dupilumab, at the same concentration or at the same concentration range, (b) L-arginine, (c) the same surfactant and (d) the same buffer at the same concentration range, but additionally comprising a sugar, e.g. a sugar comprising more than one monomer unit, and/or a sugar alcohol.
- a sugar e.g. a sugar comprising more than one monomer unit, and/or a sugar alcohol.
- the liquid antibody formulation comprises (f) a sugar alcohol.
- the sugar alcohol is mannitol or sorbitol.
- the liquid antibody formulation comprises mannitol as a sugar alcohol at a concentration from 50 to 250 mM such as 60 or 120 mM.
- the liquid antibody formulation comprises sorbitol as a sugar alcohol at a concentration from 50 to 250 mM such as 60 or 120 mM.
- the liquid antibody formulation of the present invention may further comprise a salt.
- the liquid antibody formulation comprises a salt at a concentration of from 10 mM to 100 mM, such as 20 mM to 80 mM, or 30 mM to 50 mM.
- the salt can be selected from a group consisting of sodium chloride (NaCl), potassium chloride (KCL), monosodium dihydrogen phosphate (NaH ⁇ PC ), disodium hydrogen phosphate (ISfeHPCU), and potassium dihydrogen phosphate (KH2PO4).
- the liquid antibody formulation comprises sodium chloride.
- the liquid antibody formulation comprises sodium chloride at a concentration range from 10 mM to 100 mM such as 20 mM to 80 mM, or 30 mM to 50 mM.
- the pH of the liquid antibody formulation is from 5.0 to 7.0, preferably from 5.5 to 6.2 such as 5.9. In a preferred embodiment, the pH of the liquid antibody formulation is about 5.9.
- liquid antibody formulation of the present invention may further comprise HC1 or NaOH.
- the liquid antibody formulation of the present invention comprises 0.05 to 0.2 N HC1, more preferably 0.1 N HC1.
- the liquid antibody formulation of the present invention may comprise one or more further excipient(s) and/or additive(s).
- excipient and/or additive
- additive are used interchangeably herein and refer to any non-therapeutic agent added to the formulation to provide a desired consistency, viscosity or stabilizing effect.
- Stabilizer refers to an excipient and/or an additive that protects and stabilizes a protein in the composition in dry form or in the absence of water. Further, a stabilizer also protects the protein from degradation under storage conditions. For example, a stabilizer prevents denaturation or aggregation by preserving the tertiary or quaternary structure of said protein.
- the stabilizer can be a thermal stabilizer.
- the stabilizer e.g. thermal stabilizer can be included at a concentration of from about 0.9% ⁇ 0.135% w/v to about 10% ⁇ 1.5% w/v.
- the thermal stabilizer is a sugar.
- the sugar is selected from the group consisting of sucrose, mannitol and trehalose.
- the thermal stabilizer is sucrose at a concentration of about 5% 6 0.75% w/v.
- the liquid antibody formulation of the present invention does not comprise a sugar, preferably the formulation does not comprise sucrose and/or trehalose.
- the liquid antibody formulation of the present invention does not comprise a sugar and/or a sugar alcohol and/or methionine and/or proline and/or an acetate buffer, preferably the formulation does not comprise sucrose and/or trehalose, more preferably the formulation does not comprise sucrose.
- Exemplary preferred liquid antibody formulations according to the invention comprise 150 mg/mL or 175 mg/mL of dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80. In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM histidine, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM histidine, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM histidine, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.2 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 20.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.5 mg/mL pol oxamer 188.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL pol oxamer 188.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80. In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM histidine, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM histidine, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM histidine, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.2 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 20.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.5 mg/mL pol oxamer 188.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL pol oxamer 188.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
- the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80 and does not comprise sucrose.
- the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80. In a particularly preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80 and does not comprise sucrose.
- All of the above formulations preferably have a pH of 5.9.
- the liquid antibody formulation of the invention can be provided as a dosage form, comprising an application device, said device containing the liquid formulation.
- the application device comprises a container.
- Preferred embodiments for respective devices or containers are a pre-filled syringe, an autoinjector, a pre-filled pen, a pre-filled cartridge for on -body injector, a glass vial, and a microinfuser, wherein a pre-filled syringe or an autoinjector is preferred.
- a preferred container size of the pre-filled syringe or the autoinjector is 2.25 ml.
- An alternatively preferred container size of the pre-filled syringe or the autoinjector is 1 ml.
- the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 100 mg antibody, preferably dupilumab, preferably at a concentration of 150 mg/mL.
- the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 100 mg antibody, preferably dupilumab, preferably at a concentration of 175 mg/mL.
- the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 200 mg antibody, preferably dupilumab, preferably at a concentration of 150 mg/mL.
- the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 200 mg antibody, preferably dupilumab, preferably at a concentration of 175 mg/mL.
- the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 300 mg antibody, preferably dupilumab, preferably at a concentration of 150 mg/mL.
- the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 300 mg antibody, preferably dupilumab, preferably at a concentration of 175 mg/mL.
- each of the devices can be filled with the same liquid antibody formulation according to the invention.
- a single formulation can be used in a set of dosage forms.
- the currently marketed Dupixent® formulations comprise different amounts of the excipients for different dupilumab concentration of 150 mg/mL and 175 mg/mL.
- US 2023/348532 Al suggests different excipient buffer solutions for a target concentration of 150 mg/mL and for a target FDS concentration of 175 mg/mL.
- the set comprise a plurality of dosage forms according to the present invention.
- the set comprises at least three dosage forms containing the same antibody formulation in different volumes.
- different dosages can be achieved, e.g. (e.g. 100 mg, 200 mg and 300 mg of antibody, preferably dupilumab).
- the set preferably comprises dosage forms containing at least two different devices selected from pre-filled syringe, autoinjector, pre-filled pen, pre-filled cartridge for on-body injector, glass vial and microinfuser.
- a preferred set comprises the following plurality of dosage forms: a pre-filled syringe containing a dosage of 100 mg antibody; a pre-filled syringe containing a dosage of 200 mg antibody; a pre-filled syringe containing a dosage of 300 mg antibody; an autoinjector containing a dosage of 100 mg antibody; an autoinjector containing a dosage of 200 mg antibody; an autoinjector containing a dosage of 300 mg antibody.
- the set of dosage forms according to the present invention preferably contains the same formulation. For the set of dosage forms different dosages are preferably achieved by using different amounts of formulation, but not different concentrations.
- the present invention further relates to a method of preparing the liquid antibody formulation disclosed herein comprising mixing (a) an antibody, preferably dupilumab, (b) L-arginine at a concentration of from 150 to 300 mM, and (c) a surfactant, and optionally (d) a buffer and/or optionally one or more further excipient(s).
- the present invention further relates to the use of arginine at a concentration of from 110 to 320 mM for stabilization of an antibody, preferably dupilumab, in a liquid antibody formulation.
- the present invention further relates to the use of arginine at a concentration of from 150 to 250 mM, preferably at a concentration of from 150 to 230 mM, preferably at a concentration of from 170 to 250 mM, preferably at a concentration of from 170 to 230 mM, preferably at a concentration of 170 mM for stabilization of dupilumab in a liquid antibody formulation.
- the present invention further relates to the liquid antibody formulation disclosed herein for use in the treatment of allergic bronchopulmonary aspergillosis, allergic fungal rhinosinusitis, allergic rhinitis, allergic rhinoconjunctivitis (grass pollen allergy), allergies food milk, alopecia areata, aspirin-exacerbated respiratory disease, asthma, allergic asthma, atopic dermatitis (AD; moderate to severe atopic dermatitis/dermatitis/eczema/skin diseases/genetic skin diseases/inbom genetic diseases/eczematous skin diseases/hypersensitivity/immediate hypersensitivity/immune system diseases), atopic keratoconjunctivitis, bullous pemphigoid, cholinergic urticaria, chronic obstructive pulmonary disease (COPD), chronic rhinosinusitis with nasal polyps (CRSwNP), chronic rhinosinusitis without nasal polyps (CRsNP; sinusitis/chronic sinusitis/sinus disorder
- the present invention further relates to the liquid antibody formulation disclosed herein for use in the treatment of atopic dermatitis (AD), e.g. moderate to severe atopic dermatitis, atopic eczema, asthma, e.g.
- AD atopic dermatitis
- atopic dermatitis e.g. moderate to severe atopic dermatitis, atopic eczema, asthma, e.g.
- CRSwNP chronic rhinosinusitis with nasal polyposis
- CRsNP chronic rhinosinusitis without nasal polyps
- EoE eosinophilic esophagitis
- PN prurigo nodularis
- COPD chronic obstructive pulmonary disease
- CSU chronic spontaneous urticaria
- CINDU chronic inducible cold urticaria
- AFRS allergic fungal rhinosinusitis
- the concentration of dupilumab was determined by absorption spectroscopy at 280 nm with correction for any aggregated particles at 320 nm using a Nanophotometer N120 from Implen.
- Formulated drug substance (DS) was diluted with the respective placebo solution and measured against the respective placebo solution. Protein concentrations were calculated using the absorption coefficient of 1.412 L*g' 1 *cm' 1 . Concentrations were reported based on the mean of three replicate measurements.
- CZE capillary zone electrophoresis
- dupilumab 5 pg of formulated dupilumab (diluted to 1 mg/mL with mobile phase 50 mmol/L sodium phosphate, 300 mmol/L sodium chloride, pH 6.8) were separated on a TSKgel® UP- SW3000 SEC column (4.6 x 150 mm, 2 pm, from Tosoh Bioscience) using a ThermoFisher Ultimate 3000 HPLC system with an isocratic flow of 0.35 mL/min (mobile phase: 50 mmol/L sodium phosphate, 300 mmol/L sodium chloride, pH 6.8) to analyze presence of high molecular weight species of dupilumab (HMWS) during a run time of 8 min. Data were recorded at 280 nm by a UV detector. Chromatogram profiles were compared and evaluated for HMWS and main peak relative area. All measurements were performed as single measurement.
- Non-reducing capillary electrophoresis (nRCE-SDS or non-reducing cGE) was performed to quantify LMWS in dupilumab formulations, using a Perkin Elmer Labchip GXII Touch Protein Characterization System, a HT Protein Express Chip (760499, Perkin Elmer) and Protein Express assay reagent kit (CLS960008, Perkin Elmer).
- the sample preparation was performed according to the manufacturer general instruction as described in the Kit handbook (PN CLS140159, Rev. F) and following:
- Formulated dupilumab samples were diluted with water to 1 mg/mL and 10 pl thereof were further diluted with 35 pl of non-reducing denaturation solution.
- Non-reducing conditions were established with stocks of alkylating reagent, namely 200 n- Ethylmaleimide (NEM, cat#E1271, from Sigma Aldrich) added at 1 :20 (v/v) ratio to the sample buffer provided with the kit.
- Denaturation solution was supplemented with lithium dodecyl sulfate (LDS, #L4632, from Sigma Aldrich) to a final concentration of 1%. Sample denaturation was performed at 75 °C for 10 min in a Bio-Rad real-time PCR cycler without shaking.
- Electrophoretic separation was performed using the LabChip GXII Touch HT software, running the preset P200 Antibody Analysis method (Perkin Elmer).
- Raw data were exported and analyzed using Chromeleon software suite, version 7.2.10 from Thermo Fisher.
- the peak results were evaluated as normalized relative peak area (RFU). Normalization was achieved by division of the peak integral (RFU*min) by the peak migration time. Electropherogram profiles as well as purity (main peak) and impurity (LMWS, HMWS) content was monitored.
- Determination of pH was performed with 150 pl aliquots of the samples and either calibrated Ultra Micro ISM pH-electrode or Micro Pro-ISM pH-electrode (both Mettler Toledo) connected to a SevenExcellence pH-Meter (Mettler Toledo) at room temperature. Samples were equilibrated at room temperature for at least 30 min before measurement.
- Measurements were performed at 20°C on a Kinexus ultra plus rheometer and using a cone-plated geometry (diameter: 40 mm; cone angle: 0.5°; sample volume 160 pl). The samples were equilibrated for 2 minutes at 20°C prior to each measurement. After ramping up from 0.1 to 1000 s’ 1 , the dynamic viscosity was measured for one minute at constant shear rate of 1000 s’ 1 . All samples were analyzed as single measurements.
- Osmolality was determined by freezing point depression on an osmometer (Osmomat 030 D-RS from Gonotec, Germany. Calibration was performed using sodium chloride standards (500 and 850 mOmol/kg) and purified water. Samples were measured diluted at 50 mg/mL dupilumab concentration (50 pl sample volume) and recalculated. Dilution was performed by using purified water. Determination of hydrodynamic radius by DLS
- the hydrodynamic radius of samples containing dupilumab was determined by dynamic light scattering (DLS) at 25°C, using a DynaPro Plate Reader from Wyatt Technology (Santa Barbara, California, USA). Degradation products of samples can be either fragments with smaller radii than the radius of the monomer of dupilumab, or aggregates with higher radii. Samples were diluted to 2 mg/mL (diluted with 0.02 pm filtered water). Analysis was performed using the software Dynamics from Wyatt (version 8.1.2.144). Two replicates of each sample were transferred to a 384 well plate and measured twice. 35 acquisitions of 1 second each were performed. The radii for each population (x-axis) and the D50 value of all particles measured (y-axis) were plotted in a log-log scatter plot.
- ko value is a measure of the balance between repulsive and attractive intermolecular forces.
- a positive ko value reveals repulsive forces between the proteins in solutions and hence a reduced tendency for aggregation as the surface net charge of the proteins keeps them separated, negative values for ko are related to attractive net forces between the molecules.
- Nano-DSF measurements were performed on a NanoTemper Prometheus NT.plex in 24- channel glass capillaries with the PR.ThermControl software (v2.3.1). Conformational stability of dupilumab was determined by intrinsic fluorescence measurement at 330 nm and 350 nm during thermal unfolding with a l°C/min temperature ramp from maximum 25°C to 95°C. Samples were analyzed with PR.Stability Analysis software (vl.l) and compared regarding the onset and midpoint of protein unfolding, TON and T m , respectively. TON and inflection points were determined manually by identifying the transition and inflection points respectively on the curves with the PR.ThermControl software. Samples were measured diluted to 10 mg/mL in duplicates.
- Micron-sized protein aggregates and particles are important quality attributes of therapeutic protein formulations due to their risk of enhancing an immunogenic response.
- pharmacopoeias require quantification of SvP larger than 10 pm and 25 pm.
- the pharmacopeial acceptance criteria for parenteral administration in small volume measured by light obscuration are: NMT 6000 particles > 10 pm per container and NMT 600 particles > 25 pm per container.
- SvP ⁇ 10 pm must be monitored, however, specific acceptance criteria are not defined.
- formulations were analyzed by flow imaging microscopy using a FlowCam® 8100 system (ANASYSTA). This method captures SvP more efficiently compared to light obscuration since it is more sensitive at detecting particles with refractive indices similar to the solvent.
- ANASYSTA FlowCam® 8100 system
- Reversed phase high performance liquid chromatography was performed to quantify oxidized species in dupilumab formulations.
- dupilumab samples were diluted to a final concentration of approx. 0.4 g/L during the IdeS digestion and reduction. IdeS digestion and reduction was proceeded as follows:
- the measurement was performed using a Waters Aquity UPLC H-class system and Empower 3 software, also from Waters.
- Example 2 Liquid formulations of dupilumab selected for stability study
- Table 1 The formulations of Table 1 are tested according to Table 2 in a 12-month long-term stability at target storage condition 2°C to 8°C (i.e. 5°C ⁇ 3°C) as well as accelerated conditions at 25°C/60% RH for up to 6 months and at 40°C/75% RH for up to 1 month. In addition, samples will undergo mechanical stressing as well as five freeze/thaw cycles.
- Protein stability was determined by size exclusion chromatography (SE-HPLC) for the presence of high molecular weight species (HMWS) and by non-reducing SDS-cGE (nRCE-SDS) for the presence of low molecular weight species (LMWS).
- CZE was used to detect modifications leading to charge heterogeneities, i.e. to detect acidic and basic species. Oxidation was monitored by RP-HPLC chromatography after IdeS digestion. DLS was analyzed, also to calculate ko values for characterizing protein-protein interactions.
- Melting temperature (Tm) and aggregation temperature (Tagg) were analyzed by nano-differential scanning fluorometry (nano-DSF).
- samples were analyzed for visual appearance, dynamic viscosity, turbidity, sub-visible particle content and particle size. Osmolality of the samples was determined. The protein concentration of the samples was determined by UV-VIS spectroscopy. Also, pH was monitored during the stability study.
- Osmolality measurements were performed for all formulations of Table 1. Osmolality for all formulations is within the range of 260 to 390 mOsm/kg.
- Table 4 ko value of formulations prepared within this study along with adjusted correlation coefficient R 2 The ko values are observed with high confidence explaining >85% of the data.
- Table 5 HMWS and main peak of dupilumab as A re i.area [%] in the formulations prepared within this study
- Table 6 LMWS and main peak of dupilumab as A re i.area [%] in the formulations prepared within this study
- CZE was performed to determine the main peak, acidic species/acidic variants (AV) and basic species/basic variants (BV) of dupilumab in the formulations of Table 1, the results are provided in Table 7.
- Table 7 Acidic variants, main peak and basic variants of dupilumab as A re i.area [%] in the formulations prepared within this study
- Formulation WP6 01 corresponds to the marketed formulation of Dupixent® comprising 175 mg/mL dupilumab.
- Target concentration (175 mg/mL ⁇ 10%) post upconcentration was achieved for all formulations.
- Osmolality measurements were performed for all formulations of Table 8. Osmolality for all formulations was within the range of 290 to 360 mOsm/kg.
- Viscosity Table 9 Viscosity measured at constant shear rate.
- Formulations WP6 01 and WP6 03 containing sucrose tend to exhibit increased viscosity.
- the syringeability of formulations WP6 01 and WP6 03 might not be optimal.
- the ko values are observed with high confidence explaining > 89% of the data.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Animal Behavior & Ethology (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Oil, Petroleum & Natural Gas (AREA)
- General Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Polymers & Plastics (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Dermatology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
The present invention relates to a liquid antibody formulation comprising an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0. The invention further relates to a method for the preparation thereof and use of the liquid antibody formulation in the treatment, prevention or amelioration of any disease or disorder associated with IL-4 activity, including diseases or disorders mediated by activation of IL-4RC, such as atopic dermatitis (AD), e.g. moderate to severe atopic dermatitis, atopic eczema, asthma, e.g. moderate to severe eosinophilic or oral steroid dependent asthma, chronic rhinosinusitis with nasal polyposis (CRSwNP), chronic rhinosinusitis without nasal polyps (CRsNP), eosinophilic esophagitis (EoE), prurigo nodularis (PN), skin infections, chronic obstructive pulmonary disease (COPD), chronic spontaneous urticaria (CSU), chronic inducible cold urticaria (CINDU), allergic fungal rhinosinusitis (AFRS).
Description
STABLE LIQUID ANTIBODY FORMULATION FOR DUPILUMAB
The present invention relates to a liquid antibody formulation comprising an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0. The invention further relates to a method for the preparation thereof and use of the liquid antibody formulation in the treatment, prevention or amelioration of any disease or disorder associated with IL-4 activity, including diseases or disorders mediated by activation of IL-4RC, such as atopic dermatitis (AD), e.g. moderate to severe atopic dermatitis, atopic eczema, asthma, e.g. moderate to severe eosinophilic or oral steroid dependent asthma, chronic rhinosinusitis with nasal polyposis (CRSwNP), chronic rhinosinusitis without nasal polyps (CRsNP), eosinophilic esophagitis (EoE), prurigo nodularis (PN), skin infections, chronic obstructive pulmonary disease (COPD), chronic spontaneous urticaria (CSU), chronic inducible cold urticaria (CINDU), allergic fungal rhinosinusitis (AFRS).
Biopharmaceuticals such as therapeutic proteins (e.g., antibodies) have gained significant attention in recent years in human and animal medicine and up to date several therapeutic antibodies have been approved for use in the treatment of various diseases.
Many of these molecules have been administered intravenously. However, there is an increasing need for self-inj ection at home for which the more convenient subcutaneous injection would be preferred as said route of administration is particularly easy to perform, is not very painful, has few complications and can also be performed by the patients themselves, which allows the patients to receive their medication independent from medical professionals, thus not requiring an appointment with a doctor nor hospitalization.
As large subcutaneous injection volumes are considered to be associated with injection pain and adverse events at the injection site, the subcutaneous route of administration of therapeutic proteins poses a volume restriction. Due to the limited injection volume, subcutaneous injection of therapeutic antibodies, usually requires formulations containing high concentrations of proteins, ranging from, for example, 150 mg/mL up to 200 mg/mL in solution. One important consideration is therefore the concentration of antibody that can be accommodated by a certain formulation.
A further consideration when formulating therapeutic antibodies is that in liquid solution the antibodies are prone to degradation, aggregation or undesired chemical modifications such as formation of acidic or basic species. Accordingly, liquid antibody solutions have to be formulated properly to ensure the antibodies maintain their stability during storage and subsequent use, but also under stress conditions such as shaking or temperature variations which might occur upon shipment, as well as planned or unplanned freezing.
Additional requirements of liquid antibody formulations relate to the viscosity of the solution, but also to the visual quality of the formulation.
Especially for liquid formulations containing high concentrated therapeutic proteins, sufficiently low viscosity and physico-chemical stability including long-term aggregation stability are major issues.
Thus, for high concentrated solutions of therapeutic proteins high viscosity of the solution is a major hurdle to overcome as high solution viscosity can impact both manufacturing and injection administration capabilities of the formulation. In contrast thereto, a low viscosity allows to use thinner needles for injection and less pain during injection. Also, for some drug manufacturing processes like ultra-/diafiltration and sterile filtration, a low viscosity is a prerequisite.
Another major problem to solve for high concentrated formulations of therapeutic proteins is the higher tendency to aggregation during storage. Protein aggregation can result in an increase in high molecular weight species (as detected by size exclusion chromatography). These aggregate levels in drug substance and final drug formulation are a key factor when assessing critical quality attributes of the molecule since aggregation might impact biological activity of the biopharmaceutical. Accordingly, due to the high immunogenicity effects of protein aggregates, their control is an important safety aspect for therapeutic drug products.
A further problem of formulations of therapeutic proteins involves degradation of the therapeutic proteins resulting in fragmentation products which are also referred to as low molecular weight species.
Apart from issues relating to physical stability such as aggregation and fragmentation of therapeutic proteins, the chemical stability of proteins under storage conditions is critical to maintain effectivity and safety of the therapeutic proteins. In particular, therapeutic
proteins under storage conditions can form unfavorable acidic or basic variants, concomitant with a decrease of the main peak.
Furthermore, formulations of therapeutic proteins, especially if applied by subcutaneous injection, should be adjusted to an osmolality of body fluid to avoid pain and tissue damage during application, targeting to be below 400 mOsmol/kg, preferably close to 300 mOsmol/kg.
A liquid antibody formulation is, thus, required to contain an adequate concentration of antibody, to possess a suitable viscosity, to remain physically and chemically stable during storage and subsequent use, and to enable convenient administration to patients.
In view of the above, the formulation of therapeutic antibodies is a challenging task in the biopharmaceutical field.
To arrive at the above-described desirable properties, different kinds of excipients are typically used in the formulation, but also the amounts and proportions of the excipients relative to one another have to be chosen carefully.
Common art to generate a stable formulation of high concentrated proteins of up to 200 mg/mL is to add one excipient to the formulation to stabilize against physicochemical degradation, typically a sugar or sugar alcohol, and an additional viscosity reducing agent.
One exemplary therapeutic antibody which is approved for subcutaneous application and formulated as a liquid formulation comprising a sugar and L-arginine as an additional viscosity reducing agent is the anti-IL-4Ra antibody dupilumab (CAS Registry Number 1190264-60-8).
The recombinant, human, monoclonal IgG4 antibody dupilumab is specifically directed against the interleukin (IL)-4Ra subunit present at type I and II receptors on immune cells such as B or T cells and thus inhibits the reaction cascades triggered via interleukin-4 and interleukin- 13 (IL-4 and IL- 13). Dupilumab thus has an antiphlogistic and immunosuppressive effect in diseases whose pathophysiology is based on overactive IL- 4 and IL- 13 signaling.
A liquid pharmaceutical formulation comprising dupilumab is described in EP 2 624 865 BL An especially preferred liquid pharmaceutical formulation comprises
(i) dupilumab at a concentration up to 200 mg/mL, (ii) acetate at a concentration of 12.5 mM ± 2 mM; (iii) histidine at a concentration of 20 mM ± 3 mM; (iv) sucrose at a concentration of 5% w/v ± 0.75% w/v; (v) polysorbate 20 or polysorbate 80 at a concentration of 0.2% w/v ± 0.03% w/v; and (vi) arginine at a concentration of 25 mM ± 3.75 mM or at a concentration of 50 mM ± 7.5 mM; wherein the formulation has a pH of 5.6 to 6.2.
According to EP 2 624 865 Bl, it is critical to formulate the anti-hIL-4Ra antibody (i.e. dupilumab) at a concentration of from about 100 mg/mL up to 200 mg/mL with sucrose as a thermal stabilizer and arginine as a viscosity enhancer to obtain stable and low to moderate viscosity liquid formulations.
However, as particularly arginine concentrations of 5% triggered undesirable high levels of degradation products, i.e. aggregates, cleavage products, and charge variants (see Table 3 of EP 2 624 865 Bl), formulations containing low amounts of arginine (25 mM or 50 mM) have been claimed. As a further relevant factor EP 2 624 865 Bl suggests the use of sucrose as an essential stabilizing ingredient in amounts of about 5%.
Similarly, US 2023/348532 Al, which generally relates to methods for manufacturing high titer antibody products, e.g. dupilumab, discloses in a side aspect excipient buffer solutions to achieve a certain target concentration of the antibody such as 150 mg/mL or 175 mg/mL. However, no final drug formulations are described.
A liquid pharmaceutical formulation comprising dupilumab in accordance with EP 2 624 865 Bl is currently marketed under the tradename Dupixent® as a solution for injection containing dupilumab at a concentration of 150 mg/mL for dosages of 100 mg and 300 mg or 175 mg/mL for dosages of 200 mg, and further comprising arginine hydrochloride at a concentration of 25 mM and 50 mM, respectively, histidine, histidine hydrochloride monohydrate, polysorbate 80 (E 433), sodium acetate trihydrate, acetic acid 99% (E 260), sucrose, and water for injection, wherein the formulation has a pH of about 5.9 (Strickley R.G. and Lambert W.J. A review of Formulations of Commercially Available Antibodies. J Pharm Sci. 2021 Jul;110(7):2590-2608.).
While Dupixent® as an approved medication satisfies the requirements of sufficiently low viscosity, enhanced storage stability of a liquid formulation comprising dupilumab is desirable. In that respect, it is particularly desirable that a high proportion of dupilumab
remains in its native state upon storage. Further, it is desirable that a single formulation fits all dosages and devices to be marketed.
It is therefore an object of the present invention to provide a stable liquid formulation of a therapeutic antibody, preferably dupilumab.
In particular, it is an object of the present invention to provide a stable liquid formulation of a therapeutic antibody containing from 150 mg/mL to 200 mg/mL of a therapeutic antibody, preferably dupilumab, by addressing the above issues including acceptable solution viscosity, chemical stabilization, physical stabilization by limiting aggregation and fragmentation, and osmolality adjustment.
It is a further object of the present invention to provide a stable liquid formulation of a therapeutic antibody containing from 150 mg/mL to 200 mg/mL of a therapeutic antibody, preferably dupilumab, wherein a high proportion of the therapeutic antibody, preferably dupilumab remains in its native state upon storage and/or under stress conditions such as shaking, temperature variations or freezing.
A further object of the present invention is the provision of a stable liquid antibody formulation, preferably a stable liquid dupilumab formulation, having advantageous, e.g. enhanced physical stability, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting sufficiently low aggregation propensity, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, having a sufficiently low proportion of high molecular weight species (HMWS), e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, wherein the hydrodynamic radius of the antibody is maintained, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, wherein the inter-
molecular attractive forces are advantageously low, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting sufficiently low fragmentation propensity, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, having a sufficiently low proportion of fragments/low molecular weight species (LMWS), e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, having improved, e.g. enhanced chemical stability, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting an acceptable, i.e. stabilized, main peak of dupilumab, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting an acceptable, i.e. reduced, formation of acidic variants, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a high concentrated therapeutic antibody, preferably dupilumab, exhibiting an acceptable, i.e. reduced, formation of basic species, e.g. upon storage and/or under stress conditions.
A further object of the present invention is the provision of a stable liquid formulation of a therapeutic antibody, preferably dupilumab, having acceptable solution viscosity and exhibiting an osmolality close to an osmolality of body fluid, i.e. near 300 mOsmol/kg.
A further object of the present invention is the provision of a stable liquid formulation of a therapeutic antibody, preferably dupilumab, which is particularly suited for drug approval.
Another object of the present invention is the provision of a liquid formulation of a therapeutic antibody, preferably dupilumab, meeting the requirements for marketing authorization as a biosimilar.
The above objects should be achieved by a formulation containing as little excipients as possible. In particular, the use of further excipients like stabilizers, e.g. sugars, sugar alcohols, other amino acids such as methionine or proline should be avoided. Especially, the use of sucrose should be avoided.
Accordingly, it is an object of the present invention to provide a liquid formulation of dupilumab containing a reduced number of excipients while maintaining the properties required for obtaining marketing authorization as a biosimilar.
A further object of the present invention is the provision of a single stable liquid formulation of a therapeutic antibody, preferably dupilumab, which is suited for using the same concentration (e.g. 150 mg/mL or 175 mg/mL) in different antibody dosages (e.g. 100 mg, 200 mg or 300 mg) and/or in different application forms (e.g. a pre-filled syringe, autoinjector, pre-filled pen, pre-filled cartridge for on-body injector, vial and microinfuser).
According to the present invention, it was contrary to the teaching of the prior art surprisingly found that the inclusion of a high concentration of L-arginine of from 150 to 300 mM in a liquid antibody formulation comprising an antibody, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0, not only allows to lower the viscosity of the formulation, but also minimizes the formation of aggregates, while, surprisingly, at the same time less degradation products (i.e. fragments) are formed and less undesired chemical modifications, such as charge variants, are observed, thus resulting in a liquid antibody formulation exhibiting acceptable viscosity and improved storage stability and, thus, extended shelf-life.
This was particularly surprising as EP 2 624 865 Bl discloses that high amounts of arginine may cause stability issues, and particularly suggests including arginine only at a concentration of 25 mM or at a concentration of 50 mM. This recommendation was later put into practice in the authorized product Dupixent®. Thus, based on the prior art, there was no reasonable expectation that a liquid antibody formulation comprising high concentrated dupilumab and further comprising arginine at a concentration beyond 100 mM might solve the above-mentioned problems, e.g. exhibit minimized formation of aggregates, and particularly reduced formation of degradation products (i.e. fragments) and charge variants, while having acceptable viscosity and enhanced stability upon long-
term storage and use as well as further advantages set forth below.
Based on the prior art, there was further no reasonable expectation that reducing the number of excipients might provide a liquid antibody formulation, particularly a liquid dupilumab formulation, having enhanced physico-chemical stability without any negative influence on parameters such as osmolality, viscosity, and turbidity.
Accordingly, the present invention relates to a liquid antibody formulation comprising (a) an antibody, preferably dupilumab or an antibody having at least 90% sequence identity with dupilumab, (b) L-arginine at a concentration of from 150 to 300 mM, (c) a surfactant, and (d) optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0.
It was surprisingly found that in the liquid antibody formulation of the present invention the hydrodynamic radius of the antibody molecules is maintained, intermolecular attractive forces of the antibody molecules are reduced and colloidal stability of the antibody molecules is improved.
The term “hydrodynamic radius”, as used herein, refers to the Stokes radius or effective hydrated radius in solution. Degradation products of samples can be either fragments with smaller radii than the radius of the monomer (e.g of a therapeutic protein such as dupilumab), or aggregates with higher radii. Maintenance of the hydrodynamic radius is a parameter demonstrating colloidal stability, a periodical determination by e.g. dynamic light scattering (DLS) of a sample might demonstrate particles aggregating over time by increasing the hydrodynamic radius of the particles.
According to the present invention, it was surprisingly found that the hydrodynamic radius of the antibody molecules, preferably dupilumab, is maintained in the liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM. Dupilumab is thus prevented from aggregation during storage and/or under stress conditions, leading to improved colloidal stability.
The term “colloidal stability”, as used herein, generally relates to particle size change (e.g. aggregation or agglomeration) of particles in a dispersion, e.g. of antibody molecules in a liquid antibody formulation. If particles are not subject to size variation, the dispersion is considered as having colloidal stability. Colloidal stability can depend on several types of interactions such as van der Waals and electrostatic interactions, steric
interactions (e.g. polymer adsorption), and hydrophobic effect, which renders it difficult to theoretically predict the colloidal stability of a dispersion. The ko value (i.e. diffusion interaction parameter) as measured by dynamic light scattering (DLS) is indicative of colloidal stability. Higher negative values for ko usually indicate an increase of proteinprotein attractive forces, thus affecting the generation of high molecular weight species, i.e. aggregates, during storage and/or under stress conditions, which leads to reduced colloidal stability. In contrast, less negative (i.e. more positive) ko values suggest repulsive intermolecular forces, thus preventing dupilumab from aggregation during storage and/or under stress conditions and leading to improved colloidal stability.
According to the present invention, it was surprisingly found that protein-protein attractive forces are reduced in the liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM, thus preventing dupilumab from aggregation during storage and/or under stress conditions and leading to improved colloidal stability. In other words, in the liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM, intermolecular forces are sufficiently repulsive for preventing dupilumab from aggregation during storage and/or under stress conditions and leading to improved colloidal stability.
Further surprisingly, the liquid antibody formulation of the present invention exhibits enhanced physical stability, including stability against aggregation (i.e. reduced HMWS formation) and against degradation/fragmentation (i.e. reduced LMWS formation), and at the same time enhanced chemical stability including stabilization of the main peak of the antibody, lower increase of acidic and/or basic species.
The term “physical stability”, as used herein, can refer to stability against aggregation (i.e. reduced HMWS formation) of a therapeutic antibody and against degradation/fragmentation (i.e. reduced LMWS formation) of a therapeutic antibody.
The term “high molecular weight species” (HMWS), as used herein, can refer to aggregated, soluble antibody, e.g. dupilumab, starting from dimeric antibody to higher aggregation levels. HMWS are measured by size-exclusion chromatography (SE-HPLC) and include the sum of all peaks of HMWS in SE-HPLC, not differing between the size, respectively the aggregation level, nor differing between covalent or non-covalent aggregation as both are unwanted species with immunogenic potential.
The term “low molecular weight species” (LMWS), as used herein, can refer to fragmented/degraded, soluble antibody, e.g. dupilumab. LMWS are measured by sizeexclusion chromatography (SE-HPLC) and include the sum of all peaks of LMWS in SE- HPLC, not differing between the size of the fragments/degradation products.
Both, HMWS and LMWS, can be indicative of the content of impurities of the liquid antibody formulation.
According to the present invention, it was surprisingly found that a liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM exhibits advantageous low formation of HMWS and LMWS. Therefore, the liquid antibody formulation has advantageous high stability against aggregation and fragmentation/degradation and, thus desirable, e.g. enhanced physical stability, e.g. upon storage and/or under stress conditions.
The term “chemical stability”, as used herein, can refer to stability against undesired chemical modifications such as the formation of acidic and/or basic species of a therapeutic antibody. Acidic or basic species, which are also referred to as “charge variants”, are defined as the sum of the antibody (e.g. dupilumab) peaks that elute from a cation exchange (CEX-HPLC) column with earlier or later retention times than the main peak, respectively. Accordingly, “chemical stability” also refers to the stabilization of the main peak of the antibody, which is a measure of the native antibody.
Both, acidic and basic species, are indicative of the content of impurities of the liquid antibody formulation, whereas the main peak is an indicator of the purity of the liquid antibody formulation.
Hence, as a measure of chemical stability, the change (i.e. the “DELTA” or “A”) between the main peak, the peak for the acidic species and the peak for the basic species, respectively, is determined in the formulation as prepared, i.e. at to, and at a later point in time.
In general, the three absolute DELTAs between (1) the main peak at to and the main peak at tiater point in time, and (2) the peak for the acid species at to and the peak for the acidic species at hater point in time, and (3) the peak for the basic species at to and the peak for the acid species at tiater point in time, are calculated.
Thus, if, for example, the peak for the acid species at to is 15% relative area and the peak for the acid species at hater point in time is 25% relative area, then the absolute DELTA relative area is 10%. In the opposite case, if, for example, the peak for the acid species at to is 15% relative area and the peak for the acid species at tiater point in time is 10% relative area, then the absolute DELTA relative area is 5%.
Those three absolute DELTAs (between the main/acid/basic peaks at to and at tiater point in time) can be considered as an indicator for the stability of the antibody in a specific formulation. The smaller the DELTA, the more stable the antibody in the specific formulation or, vice versa, the greater the DELTA, the more instable the antibody in the specific formulation.
Consequently, the comparison of the main peak at to with the main peak at tiater point in time, and the peak for the acid species at to with the peak for the acid species at tiater point in time, and the peak for the basic species at to with the peak for the acid species at tiater point in time, leads tO absolute DELTA values (Amain peak, Abasic species peak, Aacid species peak) which are indicative for the chemical stability of the antibody in a specific liquid formulation.
According to the present invention, it was surprisingly found that a liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM exhibits a reduced formation of acidic and basic species and, concomitantly has a stabilized main peak of the antibody, e.g. dupilumab, e.g. upon storage and/or under stress conditions. Therefore, the liquid antibody formulation has desirable, e.g. enhanced chemical stability.
Thus, a liquid antibody formulation containing L-arginine at a concentration of from 150 to 300 mM surprisingly exhibits improved physical stability and/or improved chemical stability. Importantly, these desirable properties can be achieved while maintaining the remaining desirable properties of the formulation, e.g. maintaining the desirable level of osmolality and viscosity.
This was especially surprising in view of the prior art as EP 2 624 865 Bl provides data in Table 3 suggesting that inclusion of 5% arginine in a liquid antibody formulation comprising dupilumab might reduce formation of acidic species, but at the same time induces elevated levels of aggregation and enhanced formulation of basic species. Thus, based on EP 2624865 B 1 it could not have been expected that improved physical stability in combination with improved chemical stability might be achieved by including L-
arginine in a liquid antibody formulation comprising dupilumab.
Accordingly, the present invention provides for liquid formulations of high concentrated antibodies, particularly high concentrated dupilumab, exhibiting optimized stability and, hence, enhanced stability upon long-term storage and/or under stress conditions.
The liquid antibody formulation of the present invention can exhibit high levels of stability.
A liquid antibody formulation of the present invention is considered stable, if one of the following stability criteria (1) to (6) is fulfilled.
1) High proportion of native antibody
Stability can be measured, for example, by determining the percentage of native antibody that remains in the formulation after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions.
The percentage of native antibody can be determined, for example, by size exclusion chromatography, e.g., size exclusion high performance liquid chromatography (SE- HPLC). Alternatively, the percentage of native antibody can be determined, for example, by non-reducing capillary electrophoresis (nRCE-SDS or non-reducing cGE).
The term “stable”, as used herein in reference to the liquid antibody formulation, means that the antibody within the liquid antibody formulation, preferably dupilumab, remains in its native form or retains an acceptable degree of chemical structure or biological function upon storage and/or under stress conditions. A formulation may be stable even though the antibody contained therein does not remain 100% native or maintain 100% of its chemical structure or biological function upon storage and/or under stress conditions. Under certain circumstances, a liquid antibody formulation may be regarded as “stable”, if about 90%, about 95%, about 96%, about 97%, about 98% or about 99% of the antibody’s structure or function are maintained upon storage and/or under stress conditions may. Analogously, a liquid antibody formulation may be regarded as “stable”, if about 90%, about 95%, about 96%, about 97%, about 98% or about 99% of the antibody are maintained in its native form upon storage and/or under stress conditions.
In one embodiment, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of the native form of the antibody, preferably dupilumab, can be detected
in the formulation upon storage and/or under stress conditions.
The term “storage conditions”, as used herein, may refer to at least 1 week, at least 2 weeks (i.e. at least 0.5 month), at least 3 weeks, at least 4 weeks, at least 1 month, at least
2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, at least 18 months, at least 24 months, at least 36 months, or more at either 2 to 8°C such as 5 °C, room temperature such as 25°C, or 40°C.
According to the present invention, the term “long-term storage” may refer to a storage period of at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, at least 18 months, at least 24 months, at least 36 months, or more.
The term “stress conditions”, as used herein, may refer to storage of at least 1 week, at least 2 weeks (i.e. at least 0.5 month), at least 3 weeks, at least 4 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or at least 6 months at room temperature such as 25 °C ± 2°C / 60% relative humidity (RH) ± 5% RH or at 40°C ± 2°C/75% RH ± 5% RH. The term “stress conditions”, as used herein, may also refer to storage with mechanical stressing (e.g. orbital shaking or overhead rotation (OH)) overhead rotation for 24 h at 25°C/300 rpm. The term “stress conditions”, as used herein, may also refer to application of at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 freeze/thaw cycles of -80°C/room temperature such as +25 °C.
In one embodiment, the liquid antibody formulation of the invention is deemed stable, if at least 97% or at least 98% or at least 99% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 40°C, as determined by SE-HPLC. In other words, the delta of the percentage of native antibody between to and ti month at 4o°c is less than 3% or less than 2% or less than 1%, as determined by SE-HPLC.
In one embodiment, the liquid antibody formulation of the invention is deemed stable, if at least 98% or at least 99% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 25°C, or after 3 months of storage at 5°C, or after
3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by SE-HPLC. In other
words, the delta of the percentage of native antibody between to and t3 months at 5°c or between to and t3 months at 25°c or between to and ts months at 5°c or between to and ts months at 25 °C or between to and to months at 5°c is less than 2% or less than 1%, as determined by SE-HPLC.
In one embodiment, the liquid antibody formulation of the invention is deemed stable, if at least 95% or at least 96% or at least 97% or at least 98% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 40°C, as determined by nRCE-SDS. In other words, the delta of the percentage of native antibody between to and ti month at 4o°c is less than 5% or less than 4% or less than 3% or less than 2%, as determined by nRCE-SDS.
In one embodiment, the liquid antibody formulation of the invention is deemed stable, if at least 97% or at least 98% or at least 99% of the native form of the antibody, preferably dupilumab, are recovered after one month of storage at 25 °C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by nRCE- SDS. In other words, the delta of the percentage of native antibody between to and t3 months at 5°c or between to and t3 months at 25°c or between to and ts months at 5°c or between to and ts months at 25°c or between to and to months at 5°c is less than 3% or less than 2% or less than 1%, as determined by nRCE-SDS.
In a preferred embodiment, at least 90% or at least 95% or at least 96% or at least 97% or at least 98% of the native form of the antibody, preferably dupilumab, is recovered from the liquid antibody formulation after one month of storage at 40°C or after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by size exclusion chromatography.
2) Low proportion of aggregated antibody (HMWS)
Stability can be measured by determining the percentage of antibody that forms aggregates (i.e. HMWS) within the liquid antibody formulation after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the proportion of aggregates that is formed. The percentage of aggregated antibody can be determined by, for example, size exclusion chromatography, e.g., size exclusion high performance liquid chromatography
(SE-HPLC).
The term “stable”, as used herein in reference to the liquid antibody formulation, usually means that at most 5% of the antibody is in an aggregated form detected in the formulation upon storage and/or under stress conditions. Under certain circumstances, a liquid antibody formulation may be regarded as “stable”, if at most about 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of the antibody can be detected in an aggregated form in the formulation upon storage and/or under stress conditions.
In a preferred embodiment, less than 3% or less than 2% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C or after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, is aggregated, as determined by SE-HPLC.
3) Low proportion of fragmented antibody (LMWS)
Stability can be measured by determining the percentage of antibody that degrades to form fragments (i.e. LMWS) within the liquid antibody formulation after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the proportion of fragments that is formed. The percentage of fragmented antibody can be determined by, for example, by non-reducing capillary electrophoresis (nRCE-SDS or non-reducing cGE).
The term “stable”, as used herein in reference to the liquid antibody formulation, usually means that at most 5% of the antibody is in a fragmented form detected in the formulation upon storage and/or under stress conditions. Under certain circumstances, a liquid antibody formulation may be regarded as “stable”, if at most about 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of the antibody can be detected in a fragmented form in the formulation upon storage and/or under stress conditions.
In a preferred embodiment, less than 5% or less than 4% or less than 3% or less than 2% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C or after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, is fragmented, as determined by nRCE-SDS.
4) High proportion of antibody in the main peak/main fraction
Stability can be measured by determining the percentage of antibody remaining in the main fraction/main peak of antibody during ion exchange after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is proportional to the fraction of antibody in the main fraction/main peak. The percentage of antibody in the main fraction/main peak can be determined, for example, by ion exchange chromatography, e.g., cation exchange high performance liquid chromatography (CEX-HPLC) or CZE (capillary zone electrophoresis).
The term “stable”, as used herein in reference to the liquid antibody formulation, means that the antibody, preferably dupilumab, when measured by an appropriate analytical method such as by capillary zone electrophoresis, demonstrates a loss of main peak area relative to the main peak area at to (i.e. Amain peak) of less than 20% or less than 15% or less than 14% or less than 13% or less than 12% or less than 11% or less than 10% or less than 9% or less than 8% or less than 7% or less than 6% or less than 5% after the liquid antibody formulation has undergone a stress condition and/or storage conditions.
In a preferred embodiment, the liquid antibody formulation demonstrates a loss of main peak area relative to the main peak area at to (i.e. Amain peak) of less than 15% or less than 13% or less than 11% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C, as determined by CZE.
In a preferred embodiment, the liquid antibody formulation demonstrates a loss of main peak area relative to the main peak area at to (i.e. Amain peak) of less than 5% or less than 4% or less than 3% or less than 2% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by CZE.
5) Low proportion of acidic species
Stability can be measured by determining the percentage of antibody that migrates in a more acidic fraction during capillary zone electrophoresis (“acidic form”, “acidic species”) than in the main fraction/main peak of antibody (“neutral conformation”) after storage for a defined period of time at a defined temperature, i.e. upon storage and/or
under stress conditions. In this case, stability is inversely proportional to the fraction of antibody in the acidic form. The percentage of “acidified” or “deamidated” antibody, i.e. acidic species, can be determined, for example, by capillary zone electrophoresis (CZE) or ion exchange chromatography, e.g., cation exchange high performance liquid chromatography (CEX-HPLC).
The term “stable”, as used herein in reference to the liquid antibody formulation, means that the antibody, preferably dupilumab, when measured by an appropriate analytical method such as by capillary zone electrophoresis, demonstrates an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. Aacidic species peak) of less than 20% or less than 15% or less than 14% or less than 13% or less than 12% or less than 11% or less than 10% or less than 9% or less than 8% or less than 7% or less than 6% or less than 5% after the liquid antibody formulation has undergone a stress condition and/or storage conditions.
In a preferred embodiment, the liquid antibody formulation demonstrates an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. Aacidic species peak) of less than 15% or less than 13% or less than 11% or less than 10% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C, as determined by CZE.
In a preferred embodiment, the liquid antibody formulation demonstrates an increase of peak area of acidic species relative to the peak area of acidic species at to (i.e. Aacidic species peak) of less than 5% or less than 4% or less than 3% or less than 2% or less than 1% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5 °C, as determined by CZE.
6) Low proportion of basic species
Stability can be measured by determining the percentage of antibody that migrates in a more basic fraction during ion exchange (“basic form”, “basic species”) than in the main fraction/main peak of antibody (“neutral conformation”) after storage for a defined period of time at a defined temperature, i.e. upon storage and/or under stress conditions. In this case, stability is inversely proportional to the fraction of antibody in the basic form. The
percentage of “basified” antibody, i.e. basic species, can be determined, for example, by ion exchange chromatography, e.g., cation exchange high performance liquid chromatography (CEX-HPLC) or capillary zone electrophoresis (CZE).
The term “stable”, as used herein in reference to the liquid antibody formulation, means that the antibody, preferably dupilumab, when measured by an appropriate analytical method such as by capillary zone electrophoresis, demonstrates an increase of peak area of basic species relative to the peak area of basic species at to (i.e. Abasic species peak) of less than 5% or less than 4% or less than 3% or less than 2% or less than 1% or less than 0.9% or less than 0.8% or less than 0.7% or less than 0.6% or less than 0.5% after the liquid antibody formulation has undergone a stress condition and/or storage conditions.
In a preferred embodiment, the liquid antibody formulation demonstrates an increase of peak area of basic species relative to the peak area of basic species at to (i.e. Abasic species peak) of less than 3%, less than 2% or less than 1% or less than 0.8% or less than 0.5% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 40°C, as determined by CZE.
In a preferred embodiment, the liquid antibody formulation demonstrates an increase of peak area of basic species relative to the peak area of basic species at to (i.e. Abasic species peak) of less than 2% or less than 1% or less than 0.8% or less than 0.5% of the antibody, which is recovered from the liquid antibody formulation after one month of storage at 25°C, or after 3 months of storage at 5°C, or after 3 months of storage at 25°C, or after 6 months of storage at 5°C, or after 6 months of storage at 25°C, or after 9 months of storage at 5°C, as determined by CZE.
Other methods may be used to assess the stability of the formulations of the present invention such as, e.g., differential scanning calorimetry (DSC) to determine thermal stability, controlled agitation to determine mechanical stability, and absorbance at about 350 nm or about 405 nm to determine solution turbidities. For example, a liquid antibody formulation may be considered stable if, after 6 or more months of storage at about 5°C to about 25°C, the change in OD405 of the formulation is less than about 0.05 (e.g., 0.04, 0.03, 0.02, 0.01, or less) from the OD405 of the formulation at time zero.
Stability may also be assessed by measuring the biological activity or binding affinity of the antibody to its target. For example, a formulation may be regarded as stable if, after
storage at 5°C etc. for a defined period of time of at least six months (e.g., 6 to 12 months), the antibody contained within the formulation, preferably dupilumab, binds to IL-4Ra with an affinity that is at least 90%, 95%, or more of the binding affinity of the antibody prior to said storage. Binding affinity may be determined by e.g., ELISA or plasmon resonance. Biological activity may be determined by an IL-4Ra activity assay, such as e.g., contacting a cell that expresses IL-4Ra with the formulation comprising the anti IL- 4Ra antibody. The binding of the antibody to such a cell may be measured directly, such as e.g., via FACS analysis. Alternatively, the downstream activity of the IL-4Ra system may be measured in the presence of the antibody, preferably dupilumab, and an IL-4Ra agonist, and compared to the activity of the IL-4Ra system in the absence of antibody. In some embodiments, the IL-4Ra may be endogenous to the cell. In other embodiments, the IL-4Ra may be ectopically expressed in the cell.
The liquid antibody formulation of the present invention further generally exhibits desirable low solution viscosity and, thus, can be suitable to be administered by subcutaneous injection e.g. using thin needles for injection which is associated with reduced pain during injection.
The term “viscosity”, as used herein, may be “kinematic viscosity” or “absolute viscosity”. “Absolute viscosity” which is also referred to as dynamic or simple viscosity, is the product of kinematic viscosity and fluid density (absolute viscosity = kinematic viscosity x density). According to the invention, a liquid antibody formulation having a “sufficiently low viscosity” or a “suitable viscosity” or an “acceptable viscosity” will exhibit an absolute viscosity of less than 15 mPa-s, or less than 13 mPa-s, or from 10 to 13 mPa-s or from 10 to 12.5 mPa-s, wherein the shear viscosity is measured by rotational rheometry at 20°C.
According to the present invention, it was surprisingly found that a liquid antibody formulation containing an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, optionally a buffer, and a surfactant, and having a pH of from 5.0 to 7.0, exhibits sufficiently low viscosity and sufficiently high stability without requiring a further viscosity reducing agent or a further stabilizing agent. In particular it was found that the use of common stabilizers like methionine, proline, sucrose could be omitted.
Accordingly, in a preferred embodiment, the liquid antibody formulation of the present invention comprises an antibody, preferably dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant and optionally a buffer, and has a pH of from 5.0 to 7.0, wherein the formulation does not comprise sucrose and/or treahlose and/or methionine and/or proline, especially the formulation does not comprise sucrose.
Furthermore, the liquid antibody formulation of the present invention generally has an osmolality that is physiologically compatible.
A “physiologically compatible” or “physiologically isotonic” osmolality, as used herein, refers to an osmolality of body fluid of about 300 mOsmol/kg, which allows to avoid pain and tissue damage during application.
In one embodiment, the liquid antibody formulation has an osmolality of 200 to 400 mOsm/kg, preferably 250 to 350 mOsmol/kg, preferably 270 to 330 mOsm/kg, more preferably 290 ± 20 mOsm/kg.
Further, also the visual appearance of the liquid antibody formulation of the present invention, met the acceptance criteria, being slightly yellowish, opalescent and more viscous than water. The visual appearance was assessed using an inspection light box equipped with non-flickering fluorescent lamps and a black and a white background plate (Portable Inspection Hood MIH-PORT, Bosch). The samples were evaluated in their storage container without magnification to assess sample appearance in respect to visible particles, color and clarity.
All of the methods described herein were performed as described in the Examples.
Additionally, the liquid antibody formulation of the present invention requires less excipients for obtaining a stable formulation. In particular, the liquid antibody formulation of the present invention does not require a sugar. Further, the liquid antibody formulation of the present invention does not require a sugar alcohol. Further, the liquid antibody formulation of the present invention does not require amino acids other than L- arginine, i.e. the liquid antibody formulation of the present invention does not require amino acids such as methionine or proline. Thus, this leads to a simpler formulation, which is easier and safer as compared to prior art formulations.
Accordingly, in one embodiment, the present invention relates to a liquid antibody
formulation consisting of (a) dupilumab; (b) L-arginine; (c) a surfactant, selected from the group consisting of polyoxyethylene(20)sorbitan monolaurate, polyoxy- ethylene(20)sorbitan monooleate, and a polyoxyethylene-polyoxypropylene-block polymer of the following formula (I)
wherein x and z = 75, and y=30, preferably polyoxy ethylene(20)sorbitan monooleate; (d) a buffer, selected from the group consisting of L-histidine/L-histidine HC1 and succinic acid/succinate, preferably succinic acid/succinate; and optionally HC1 for adjusting the pH; wherein the pH of the liquid antibody formulation is from 5.5 to 6.2.
In a further preferred embodiment, the liquid antibody formulation consists of (a) dupilumab at a concentration of 150 mg/mL or 175 mg/mL; (b) L-arginine at a concentration of from 150 to 250 mM; (c) a surfactant, selected from the group consisting of polyoxyethylene(20)sorbitan monolaurate, polyoxyethylene(20)sorbitan monooleate, and a polyoxy ethylene-polyoxypropylene-block polymer of the following formula (I)
wherein x and z = 75, and y=30, preferably polyoxyethylene(20)sorbitan monooleate, at a concentration of from 0.2 to 2.0 mg/mL, preferably 2.0 mg/mL; (d) a buffer, selected from the group consisting of L-histidine/L-histidine HC1 and succinic acid/succinate, preferably succinic acid/succinate, at a concentration of 10 mM; and optionally HC1 or NaOH for adjusting the pH; wherein the pH of the liquid antibody formulation is from 5.5 to 6.2.
According to the present invention, a particularly stable liquid antibody formulation, preferably a particularly stable liquid dupilumab formulation is obtained. Said stable liquid antibody formulation, preferably dupilumab formulation, is particularly suitable for drug approval. Concerning suitability for drug approval, it is particularly advantageous that the liquid antibody formulation, preferably the liquid dupilumab
formulation, is particularly stable and requires only a minimum number of excipients.
According to the present invention, the liquid antibody formulation comprises (a) an antibody.
As used herein, the term “antibody” refers to monoclonal or polyclonal antibodies. The term “antibody” includes but is not limited to recombinant antibodies that are generated by recombinant technologies as known in the art. The term “antibody” includes antibodies of any species, in particular of mammalian species; such as human antibodies of any isotype, including IgAl, lgA2, IgD, IgGl, lgG2a, lgG2b, lgG3, lgG4, IgE and IgM, as well as modified variants thereof; non-human primate antibodies, e.g. from chimpanzee, baboon, rhesus or cynomolgus monkey; rodent antibodies, e.g. from mouse, rat or rabbit; goat or horse antibodies; and camelid antibodies (e.g. from camels or lamas such as nanobodies) and derivatives thereof; or of bird species such as chicken antibodies; or of fish species such as shark antibodies. The term “antibody” also refers to “chimeric” antibodies in which a first portion of at least one heavy and/or light chain antibody sequence is from a first species and a second portion of the heavy and/or light chain antibody sequence is from a second species.
The term “antibody”, as used herein, further includes antigen-binding portions or antigenbinding fragments of an antibody. The term “antigen-binding fragment” also refers to an antibody that comprises at least one heavy or light chain immunoglobulin domain as known in the art and binds to one or more antigen(s). Examples of antibody fragments that can be used as biologies include Fab, Fab', F(ab')2, and Fv and scFv fragments; as well as diabodies, triabodies, tetrabodies, minibodies, domain antibodies, single-chain antibodies, bispecific, trispecific, tetraspecific or multispecific antibodies formed from antibody fragments or antibodies, including but not limited to Fab-Fv constructs. In some aspects, the antibody is a vNAR, a camelid antibody, a VHH antibody, or an antigenbinding portion thereof. Antibody fragments as defined above are known in the art.
In some embodiments of the present invention, the antibody is an IgGl, IgG2, IgG3 or IgG4 antibody. In some embodiments, the antibody is an IgG4 antibody. In some embodiments the antibody is a human antibody, a humanized antibody, or a chimeric antibody.
In some embodiments, the antibody is an anti-interleukin-4 receptor alpha (anti-IL-4Ra)
antibody. In the preferred embodiments, the antibody is dupilumab, i.e. dupilumab having CAS Registry Number 1190264-60-8. Alternatively, the antibody can be an antibody having at least 90% sequence identity with dupilumab having CAS Registry Number 1190264-60-8.
Preferably, the antibody included in the liquid antibody formulation of the present invention is dupilumab.
In a preferred embodiment, the liquid antibody formulation comprises the antibody, preferably dupilumab, at a concentration of from 150 to 200 mg/mL, preferably at a concentration of 150 mg/mL or 175 mg/mL.
The liquid antibody formulations of the present invention are particularly suitable for parenteral application including intradermal, subcutaneous, intramuscular, intraosseous, intraperitoneal, and intravenous administration. Therefore, the liquid antibody formulations of the present invention can be formulated as dosage forms, particularly as dosage forms for parenteral application, e.g. dosage forms for intradermal, subcutaneous, intramuscular, intraosseous, intraperitoneal, and intravenous administration.
Hence, another subject of the present invention is a dosage form containing the liquid antibody formulation of the present invention. Usually, the dosage form comprises an application device (e.g. as further described below) said device containing the antibody formulation of the present invention.
In a preferred embodiment, the dosage form contains the antibody formulation, wherein the antibody, preferably dupilumab, is present in the dosage range from 50 mg to 500 mg, in particular at dosages of 100 mg or 200 mg or 300 mg.
In a further preferred embodiment, the dosage form contains the antibody formulation, wherein the antibody, preferably dupilumab, is present at a concentration of 150 mg/mL and at dosages of 100 mg or 200 mg or 300 mg.
In an alternative further preferred embodiment, the dosage form contains the antibody formulation, wherein the antibody, preferably dupilumab, is present at a concentration of 175 mg/mL and at dosages of 100 mg or 200 mg or 300 mg.
According to the invention, the liquid antibody formulation comprises (b) the amino acid L-arginine (also referred to as “arginine” herein) at a concentration of from 150 mM to
300 mM.
“Amino acid”, as used herein, refers to one of the 20 known amino acids including alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, L-histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine. All amino acids except for glycine are stereoisomers (mirror images of their structure). These are labelled L-amino acids and D-amino acids to distinguish. Whenever any of the amino acids are mentioned herein, it is meant as L-amino acid. Therefore, if the term “arginine” is used, L-arginine is meant and so on. When an individual amino acid is mentioned, it encompasses its derivatives. For example, if L-arginine is used, L-arginine HC1 is encompassed.
In some embodiments, the liquid antibody formulation comprises L-arginine at a concentration of from 150 mM, 155 mM, 160 mM, 165 mM, 170 mM up to 300 mM, 295 mM, 290 mM, 285 mM, 280 mM, 275 mM, 270 mM, 265 mM, 260 mM, 255 mM, 250 mM, 245 mM, 240 mM, 235 mM, 230 mM.
In some embodiments, the liquid antibody formulation comprises L-arginine at a concentration from 150 mM to 290 mM, from 150 mM to 280 mM, from 150 mM to 270 mM, from 150 mM to 260 mM, from 150 mM to 250 mM, from 150 mM to 240 mM, from 150 mM to 230 mM, from 150 mM to 220 mM, from 150 mM to 210 mM, from 150 mM to 200 mM.
In some embodiments, the liquid antibody formulation comprises L-arginine at a concentration from 160 mM to 300 mM, from 170 mM to 300 mM, from 180 mM to 300 mM, from 190 mM to 300 mM, from 200 mM to 300 mM, from 210 mM to 300 mM, from 220 mM to 300 mM, from 230 mM to 300 mM, from 240 mM to 300 mM, from 250 mM to 300 mM, from 260 mM to 300 mM, from 270 mM to 300 mM, from 280 mM to 300 mM, from 290 mM to 300 mM.
Preferably, the liquid antibody formulation of the present invention comprises L-arginine at a concentration of from 150 to 250 mM, preferably at a concentration of from 150 to 230 mM, preferably at a concentration of from 170 to 250 mM, preferably at a concentration of from 170 to 230 mM, preferably at a concentration of 170 mM.
In a preferred embodiment, the liquid antibody formulation of the present invention comprises L-arginine at a concentration of 170 mM.
In a preferred embodiment, the liquid antibody formulation of the present invention comprises L-arginine at a concentration of 230 mM
The liquid antibody formulation of the present invention further comprises (c) a surfactant.
According to the invention, the surfactant comprises preferably a non-ionic surfactant.
The term “non-ionic surfactant” means a surfactant that contains neither positively nor negatively charged functional groups. In contrast to anionic and cationic surfactants, non- ionic surfactants do not ionize in solution.
The non-ionic surfactant can be a poloxamer or a polysorbate.
Poloxamers are non-ionic triblock copolymers composed of a central hydrophobic chain of poly(propyleneoxide) flanked by two hydrophilic chains of polyethylene oxide). The length of the polymer blocks can be customized, leading to different poloxamers with slightly different properties. Accordingly, the non-ionic surfactant can be Pluronic F127 (poloxamer 407), Pluronic F123 (poloxamer 403), Pluronic F-68 (poloxamer 188), Pluronic P123, Pluronic P85, or other polyethylene oxide-polypropylene oxide (EO-PO) block copolymers of greater than 3,000-4,000 MW or combinations thereof.
In some embodiments, the surfactant is a poloxamer, i.e. a poly oxy ethylene-poly oxy - propylene-block polymer, such as a polyoxyethylene-polyoxypropylene-block polymer of the following formula (I)
wherein x is an integer from 5 to 146, y is an integer from 18 to 60, and z is an integer from 5 to 146, preferably wherein x = 75-85, y = 25-30, and z = 79-84 (i.e. poloxamer 188 as defined according to Pschyrembel Online, 2023, ISSN 2510-1668), e.g. wherein x and z = 75, and y = 30 (i.e. poloxamer 188 as defined according to Fiedler Lexikon der Hilfsstoffe fur Pharmazie, Kosmetik und angrenzende Gebiete, 5th edition, volume 2, 2002), i.e. a polyoxyethylene-polyoxypropylene-block polymer which is also referred to as poloxamer 188.
Polysorbates are composed of ethylene oxide and sorbitan fatty acid esters. These emulsifiers are much more widely known as polysorbates, e.g., polysorbate 20, 60, and 80. Polysorbate 20, 60, and 80 utilize laurate, stearate, and oleate, respectively, for the fatty acid portion of the molecule.
In some embodiments, the liquid antibody formulation comprises a polysorbate. In some embodiments, the liquid antibody formulation comprises polyoxyethylene(20)sorbitan monolaurate (i.e. polysorbate 20). In some embodiments, the liquid antibody formulation comprises polyoxyethylene(20)sorbitan monostearate (i.e. polysorbate 60). In some embodiments, the liquid antibody formulation comprises polyoxyethylene(20)sorbitan monooleate (i.e. polysorbate 80).
According to the invention, it has further surprisingly been found that in the liquid antibody formulation of the invention, the concentration of surfactant can be reduced, while maintaining the properties required for obtaining marketing authorization as a biosimilar.
Thus, in some embodiments, the liquid antibody formulation comprises a polysorbate at a concentration of from 0.1 to 4.0 mg/mL, from 0.2 to 3.5 mg/mL, from 0.5 to 3.0 mg/mL, from 1.0 to 2.5 mg/mL, from 1.5 to 2.0 mg/mL, such as 0.2 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises a polysorbate at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises a polysorbate at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises a polysorbate at a concentration of 0.2 mg/mL or 2.0 mg/mL.
In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of from 0.1 to 4.0 mg/mL, from 0.2 to 3.5 mg/mL, from 0.5 to 3.0 mg/mL, from 1.0 to 2.5 mg/mL, from 1.5 to 2.0 mg/mL, such as 0.2 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 20 at a concentration of 0.2 mg/mL or 2.0 mg/mL.
In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a
concentration of from 0.1 to 4.0 mg/mL, from 0.2 to 3.5 mg/mL, from 0.5 to 3.0 mg/mL, from 1.0 to 2.5 mg/mL, from 1.5 to 2.0 mg/mL, such as 0.2 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises polysorbate 80 at a concentration of 0.2 mg/mL or 2.0 mg/mL.
In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of from 0.1 to 4.0 mg/mL, from 0.5 to 3.5 mg/mL, from 1.0 to 3.0 mg/mL, from 1.5 to 2.5 mg/mL, such as 0.5 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises a poloxamer at a concentration of 0.5 mg/mL or 2.0 mg/mL.
In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of 0.1 to 4.0 mg/mL, from 0.5 to 3.5 mg/mL, from 1.0 to 3.0 mg/mL, from 1.5 to 2.5 mg/mL, such as 0.5 mg/mL or 2.0 mg/mL. In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of from 1.0 to 3.0 mg/mL. In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of from 1.5 to 2.5 mg/mL. In some embodiments, the liquid antibody formulation comprises poloxamer 188 at a concentration of 0.5 mg/mL or 2.0 mg/mL.
In a preferred embodiment, the surfactant included in the liquid antibody formulation of the present invention is polyoxyethylene(20)sorbitan monolaurate, also referred to as polysorbate 20 or polyoxyethylene(20)sorbitan monooleate, also referred to as polysorbate 80, or a polyoxyethylene-polyoxypropylene-block polymer, preferably a polyoxy ethylene-polyoxypropylene-block polymer of the following formula (I)
wherein x is an integer from 5 to 146, y is an integer from 18 to 60, and z is an integer from 5 to 146.
In a preferred embodiment, the surfactant is a polyoxyethylene-polyoxypropylene-block polymer of formula (I), wherein x = 75-85, y = 25-30, and z = 79-84, preferably wherein x and z = 75, and y = 30, also referred to as poloxamer 188.
In another preferred embodiment, the surfactant is polyoxyethylene(20)sorbitan monooleate, also referred to as polysorbate 80.
In a preferred embodiment, the liquid antibody formulation comprises the surfactant at a concentration of from 0.1 to 4.0 mg/mL, preferably at a concentration of 0.2 mg/mL or 0.5 mg/mL or 2.0 mg/mL.
In a preferred embodiment, the liquid antibody formulation comprises a polyoxy ethylene- polyoxypropylene-block polymer of formula (I), wherein x = 75-85, y = 25-30, and z = 79-84, preferably wherein x and z = 75, and y = 30 (i.e. poloxamer 188), as a surfactant at a concentration of 0.5 mg/mL or 2.0 mg/mL.
In another preferred embodiment, the surfactant is polyoxyethylene(20)sorbitan monooleate (i.e. polysorbate 80) at a concentration of 0.2 mg/mL or 2.0 mg/mL.
In a particularly preferred embodiment, the surfactant is polyoxyethylene(20)sorbitan monooleate (i.e. polysorbate 80) at a concentration of 2.0 mg/mL.
The liquid antibody formulation of the present invention optionally comprises (d) a buffer.
The term “buffer”, as used herein, refers to a substance that allows a solution to maintain its pH or only marginally change its pH after addition of acidic or basic substances.
In some embodiments, the buffer is a single buffer. In some embodiments, the buffer may be a salt buffer or an amino acid buffer.
In some embodiments the amino acid buffer is alanine, aspartic acid, glutamic acid, L- histidine, lysine, cysteine, tyrosine, or serine.
According to a preferred embodiment, component (b) L-arginine is not considered a buffer.
In some embodiments, a L-histidine buffer is used, i.e. L-histidine/L-histidine HC1. Said buffer is also referred to as “histidine” or “L-histidine” herein.
The concentration of the amino acid buffer in the liquid antibody formulation may range from 5 mM to 20 mM. In some embodiments, the concentration of the amino acid buffer in the liquid antibody formulation is from 5 mM to 15 mM, such as 10 mM.
Thus, when L-histidine is used as the buffer, the concentration of L-histidine in the liquid antibody formulation may range from 5 to 20 mM, or 5 mM to 15 mM, such as 10 mM.
In some embodiments, the concentration of L-histidine in the liquid antibody formulation is 10 mM.
In some embodiments, a salt buffer is used. In some embodiments, the salt buffer system is citrate; adipate, bicarbonate; phosphate, succinate, or tartrate.
In some embodiments, a succinate buffer is used i.e. succinic acid/succinate. Said buffer is also referred to as “succinate” herein.
In some embodiments, the liquid antibody formulation does not comprise an acetate buffer.
The concentration of the salt buffer in the liquid antibody formulation may range from 5 mM to 20 mM. In some embodiments, the concentration of the salt buffer in the liquid antibody formulation is from 5 mM to 15 mM, such as 10 mM.
Thus, when succinate is used as the buffer, the concentration of succinate in the liquid antibody formulation may range from 2 mM to 25 mM, 5 mM to 20 mM or 5 mM to 10 mM. In some embodiments, the concentration of succinate in the liquid antibody formulation is 10 mM.
In some embodiments, a single buffer is used. In some embodiments a single buffer is used, which is an amino acid buffer. In some embodiments a single buffer is used, which is a salt buffer. In some embodiments the liquid antibody formulation comprises L-histidine as the buffer and comprises no other buffer. In some embodiments the liquid antibody formulation comprises succinate as the buffer and comprises no other buffer.
In a preferred embodiment, the liquid antibody formulation comprises a buffer. Preferably, the buffer is L-histidine/L-histidine HC1 or succinic acid/succinate.
In a preferred embodiment, the liquid antibody formulation comprises the buffer, preferably the L-histidine/L-histidine HC1 or succinic acid/succinate buffer at a concentration of from 5 to 20 mM, preferably at a concentration of 10 mM.
In a preferred embodiment, the liquid antibody formulation comprises as the buffer succinic acid/succinate buffer at a concentration of from 5 to 20 mM, preferably at a concentration of 10 mM.
In a preferred embodiment, the liquid antibody formulation of the present invention does not comprise an acetate buffer.
The liquid antibody formulation of the present invention may further comprise (e) L-glycine.
In some embodiments, the liquid antibody formulation comprises L-glycine at a concentration of from 20 to 150 mM. In some embodiments, the concentration of L-glycine in the liquid antibody formulation is from 20 to 150 mM, from 25 mM to 150 mM, from 30 mM to 150 mM, from 35 mM to 150 mM, from 40 mM to 150 mM, from 45 mM to 150 mM, from 50 mM to 150 mM, from 55 mM to 150 mM, from 60 mM to 150 mM, from 70 mM to 150 mM, from 75 mM to 150 mM, from 80 mM to 150 mM, from 90 mM to 150 mM, from 95 mM to 150 mM, from 100 mM to 150 mM.
The liquid antibody formulation of the present invention may further comprise (f) a sugar alcohol.
Sugars and sugar alcohols are used as common stabilizers in liquid antibody formulations.
“Sugar”, as used herein, refers to monosaccharides, disaccharide sugars or polysaccharide sugars. Disaccharides form when two monosaccharides undergo a dehydration reaction (or a condensation reaction or dehydration synthesis). During this process, one monosaccharide's hydroxyl group combines with another monosaccharide's hydrogen, releasing a water molecule and forming a covalent bond. This is called a glycosidic bond. Glycosidic bonds (or glycosidic linkages) can be alpha or beta type. An alpha bond is formed when the OH group on the carbon- 1 of the first glucose is below the ring plane,
and a beta bond is formed when the OH group on the carbon- 1 is above the ring plane. The term “sugar”, as used herein, does not include “sugar alcohol”.
“Sugar alcohol”, as used herein, refers to organic compounds, typically derived from sugars, containing one hydroxyl group (-OH) attached to each carbon atom. Sugar alcohols have the general formula HOCH2(CHOH)nCH2OH. In contrast, sugars have two fewer hydrogen atoms, for example H0CH2(CH0H)nCH0 or H0CH2(CH0H)n IC(0)CH20H. The sugar alcohols differ in chain length. Most have five- or six-carbon chains, because they are derived from pentoses (five-carbon sugars) and hexoses (six-carbon sugars), respectively. They have one -OH group attached to each carbon. They are further differentiated by the relative orientation (stereochemistry) of these -OH groups. Unlike sugars, which tend to exist as rings, sugar alcohols do not - although they can be dehydrated to give cyclic ethers (e.g., sorbitol can be dehydrated to isosorbide).
According to the present invention it has surprisingly been found that a sugar is not necessary for a liquid antibody formulation. The inventors found that a sugar such as trehalose that comprises two monomer units is not necessary for a liquid antibody formulation. Since trehalose and sucrose are both non-reducing sugars commonly used as a stabilizer in polypeptide formulations, and since it is known that trehalose may even be better as compared to sucrose for polypeptide formulations, it was surprisingly found that sugars, in general are not required for a liquid antibody formulation.
This was particularly surprising, as prior art formulations such as the currently marketed Dupixent® formulations usually comprise sucrose as an essential stabilizing ingredient, as also suggested by EP 2 624 865 Bl and US 2023/348532 Al.
In some embodiments, the liquid antibody formulation does not comprise a sugar comprising two monomer units. In some embodiments, the liquid antibody formulation does not comprise a sugar. In some embodiments, the liquid antibody formulation does not comprise a sugar and does not comprise a sugar alcohol.
In some embodiments, the liquid antibody formulation does not comprise a sugar comprising two monomer units but comprises a sugar alcohol. In some embodiments, the liquid antibody formulation does not comprise a sugar but comprises a sugar alcohol.
In some embodiments, the liquid antibody formulation does not comprise a sugar
comprising two monomer units and does not comprise a sugar alcohol. In some embodiments, the liquid antibody formulation does not comprise a sugar and does not comprise a sugar alcohol.
In some embodiments, the liquid antibody formulation does not comprise a sugar but comprises a sugar alcohol. In some embodiments, the sugar alcohol is present at a concentration of from 50 mM to 250 mM. In some embodiments, the sugar alcohol is present at a concentration of from 50 mM to 250 mM, from 50 mM to 200 mM, from 50 mM to 150 mM, from 50 mM to 130 mM. In some embodiments, the sugar alcohol is present at a concentration of from 50 mM to 150 mM. In some embodiments, the sugar alcohol is present at a concentration of 60 mM or 120 mM.
In some embodiments, the sugar alcohol is mannitol. In some embodiments, mannitol is present at a concentration of from 50 mM to 250 mM. In some embodiments, mannitol is present at a concentration of from 50 mM to 250 mM, from 50 mM to 200 mM, from 50 mM to 150 Mm, from 50 mM to 130 mM. In some embodiments, mannitol is present at a concentration of from 50 mM to 150 mM. In some embodiments, mannitol is present at a concentration of 60 mM or 120 mM.
In some embodiments, the sugar alcohol is sorbitol. In some embodiments, sorbitol is present at a concentration of from 50 mM to 250 mM. In some embodiments, sorbitol is present at a concentration of from 50 mM to 250 mM, from 50 mM to 200 mM, from 50 mM to 150 MM, from 50 mM to 130 mM. In some embodiments, sorbitol is present at a concentration of from 50 mM to 150 mM. In some embodiments, sorbitol is present at a concentration of 60 mM or 120 mM.
In a preferred embodiment, the liquid antibody formulation does not comprise a sugar. Preferably, the liquid antibody formulation does not comprise a sugar comprising more than one monomer unit.
In a preferred embodiment, the liquid antibody formulation does not comprise a sugar alcohol.
In a preferred embodiment of the liquid antibody formulation of the invention, the peak area of HMWS in %, as measured by size exclusion chromatography, is lower as compared to a liquid antibody formulation of the same pH or the same pH range comprising, (a) the same antibody, preferably dupilumab, at the same concentration or at
the same concentration range, (b) L-arginine, (c) the same surfactant and (d) the same buffer at the same concentration range, but additionally comprising a sugar, e.g. a sugar comprising more than one monomer unit, and/or a sugar alcohol.
In another preferred embodiment, the liquid antibody formulation comprises (f) a sugar alcohol. Preferably, the sugar alcohol is mannitol or sorbitol. In a preferred embodiment, the liquid antibody formulation comprises mannitol as a sugar alcohol at a concentration from 50 to 250 mM such as 60 or 120 mM. In another preferred embodiment, the liquid antibody formulation comprises sorbitol as a sugar alcohol at a concentration from 50 to 250 mM such as 60 or 120 mM.
The liquid antibody formulation of the present invention may further comprise a salt. In some embodiments, the liquid antibody formulation comprises a salt at a concentration of from 10 mM to 100 mM, such as 20 mM to 80 mM, or 30 mM to 50 mM. In some embodiments, the salt can be selected from a group consisting of sodium chloride (NaCl), potassium chloride (KCL), monosodium dihydrogen phosphate (NaH^PC ), disodium hydrogen phosphate (ISfeHPCU), and potassium dihydrogen phosphate (KH2PO4). In some embodiments, the liquid antibody formulation comprises sodium chloride. In some embodiments, the liquid antibody formulation comprises sodium chloride at a concentration range from 10 mM to 100 mM such as 20 mM to 80 mM, or 30 mM to 50 mM.
According to the present invention, the pH of the liquid antibody formulation is from 5.0 to 7.0, preferably from 5.5 to 6.2 such as 5.9. In a preferred embodiment, the pH of the liquid antibody formulation is about 5.9.
For pH adjustment, hydrochloric acid (HC1) or sodium hydroxide (NaOH) can be added. Accordingly, the liquid antibody formulation of the present invention may further comprise HC1 or NaOH.
Preferably, the liquid antibody formulation of the present invention comprises 0.05 to 0.2 N HC1, more preferably 0.1 N HC1.
The liquid antibody formulation of the present invention may comprise one or more further excipient(s) and/or additive(s). The terms “excipient” and “additive”, are used interchangeably herein and refer to any non-therapeutic agent added to the formulation to provide a desired consistency, viscosity or stabilizing effect.
“Stabilizer”, as used herein, refers to an excipient and/or an additive that protects and stabilizes a protein in the composition in dry form or in the absence of water. Further, a stabilizer also protects the protein from degradation under storage conditions. For example, a stabilizer prevents denaturation or aggregation by preserving the tertiary or quaternary structure of said protein. The stabilizer can be a thermal stabilizer. The stabilizer, e.g. thermal stabilizer can be included at a concentration of from about 0.9% ± 0.135% w/v to about 10% ± 1.5% w/v. In one embodiment, the thermal stabilizer is a sugar. In one embodiment, the sugar is selected from the group consisting of sucrose, mannitol and trehalose. In a specific embodiment, the thermal stabilizer is sucrose at a concentration of about 5% 6 0.75% w/v.
In a preferred embodiment, the liquid antibody formulation of the present invention does not comprise a sugar, preferably the formulation does not comprise sucrose and/or trehalose.
In a preferred embodiment, the liquid antibody formulation of the present invention does not comprise a sugar and/or a sugar alcohol and/or methionine and/or proline and/or an acetate buffer, preferably the formulation does not comprise sucrose and/or trehalose, more preferably the formulation does not comprise sucrose.
Exemplary preferred liquid antibody formulations according to the invention comprise 150 mg/mL or 175 mg/mL of dupilumab, L-arginine at a concentration of from 150 to 300 mM, a surfactant, and optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM histidine, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM histidine, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM histidine, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.2 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 20.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.5 mg/mL pol oxamer 188.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL pol oxamer 188.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM histidine, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM histidine, 200 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM histidine, 230 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.2 mg/mL polysorbate 80.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 20.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 0.5 mg/mL pol oxamer 188.
In a preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL pol oxamer 188.
In a particularly preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a particularly preferred embodiment, the liquid antibody formulation according to the invention comprises 150 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80 and does not comprise sucrose.
In a particularly preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80.
In a particularly preferred embodiment, the liquid antibody formulation according to the invention comprises 175 mg/mL dupilumab, 10 mM succinate, 170 mM L-arginine, and 2.0 mg/mL polysorbate 80 and does not comprise sucrose.
All of the above formulations preferably have a pH of 5.9.
As mentioned above, the liquid antibody formulation of the invention can be provided as a dosage form, comprising an application device, said device containing the liquid formulation. Usually, the application device comprises a container. Preferred embodiments for respective devices or containers are a pre-filled syringe, an autoinjector, a pre-filled pen, a pre-filled cartridge for on -body injector, a glass vial, and a microinfuser, wherein a pre-filled syringe or an autoinjector is preferred. A preferred container size of the pre-filled syringe or the autoinjector is 2.25 ml. An alternatively preferred container size of the pre-filled syringe or the autoinjector is 1 ml.
In a preferred embodiment, the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 100 mg antibody, preferably dupilumab, preferably at a concentration of 150 mg/mL.
In a preferred embodiment, the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 100 mg antibody, preferably dupilumab, preferably at a concentration of 175 mg/mL.
In a preferred embodiment, the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 200 mg antibody, preferably dupilumab, preferably at a concentration of 150 mg/mL.
In a preferred embodiment, the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 200 mg antibody, preferably dupilumab, preferably at a concentration of 175 mg/mL.
In a preferred embodiment, the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation
contains 300 mg antibody, preferably dupilumab, preferably at a concentration of 150 mg/mL.
In a preferred embodiment, the dosage form comprises a pre-filled syringe containing the liquid antibody formulation according to the present invention, wherein the formulation contains 300 mg antibody, preferably dupilumab, preferably at a concentration of 175 mg/mL.
In a preferred embodiment, each of the devices can be filled with the same liquid antibody formulation according to the invention. In other words, it was surprisingly found that a single formulation can be used in a set of dosage forms. This was particularly surprising as the currently marketed Dupixent® formulations comprise different amounts of the excipients for different dupilumab concentration of 150 mg/mL and 175 mg/mL. Similarly, also US 2023/348532 Al suggests different excipient buffer solutions for a target concentration of 150 mg/mL and for a target FDS concentration of 175 mg/mL.
Hence, another subject of the present invention is a set of dosage forms. The set comprise a plurality of dosage forms according to the present invention. Preferably, the set comprises at least three dosage forms containing the same antibody formulation in different volumes. By using the same formulation with different volumes different dosages can be achieved, e.g. (e.g. 100 mg, 200 mg and 300 mg of antibody, preferably dupilumab).
Further, the set preferably comprises dosage forms containing at least two different devices selected from pre-filled syringe, autoinjector, pre-filled pen, pre-filled cartridge for on-body injector, glass vial and microinfuser.
A preferred set comprises the following plurality of dosage forms: a pre-filled syringe containing a dosage of 100 mg antibody; a pre-filled syringe containing a dosage of 200 mg antibody; a pre-filled syringe containing a dosage of 300 mg antibody; an autoinjector containing a dosage of 100 mg antibody; an autoinjector containing a dosage of 200 mg antibody; an autoinjector containing a dosage of 300 mg antibody.
The set of dosage forms according to the present invention preferably contains the same formulation. For the set of dosage forms different dosages are preferably achieved by using different amounts of formulation, but not different concentrations.
The present invention further relates to a method of preparing the liquid antibody formulation disclosed herein comprising mixing (a) an antibody, preferably dupilumab, (b) L-arginine at a concentration of from 150 to 300 mM, and (c) a surfactant, and optionally (d) a buffer and/or optionally one or more further excipient(s).
The present invention further relates to the use of arginine at a concentration of from 110 to 320 mM for stabilization of an antibody, preferably dupilumab, in a liquid antibody formulation.
In a preferred embodiment, the present invention further relates to the use of arginine at a concentration of from 150 to 250 mM, preferably at a concentration of from 150 to 230 mM, preferably at a concentration of from 170 to 250 mM, preferably at a concentration of from 170 to 230 mM, preferably at a concentration of 170 mM for stabilization of dupilumab in a liquid antibody formulation.
The present invention further relates to the liquid antibody formulation disclosed herein for use in the treatment of allergic bronchopulmonary aspergillosis, allergic fungal rhinosinusitis, allergic rhinitis, allergic rhinoconjunctivitis (grass pollen allergy), allergies food milk, alopecia areata, aspirin-exacerbated respiratory disease, asthma, allergic asthma, atopic dermatitis (AD; moderate to severe atopic dermatitis/dermatitis/eczema/skin diseases/genetic skin diseases/inbom genetic diseases/eczematous skin diseases/hypersensitivity/immediate hypersensitivity/immune system diseases), atopic keratoconjunctivitis, bullous pemphigoid, cholinergic urticaria, chronic obstructive pulmonary disease (COPD), chronic rhinosinusitis with nasal polyps (CRSwNP), chronic rhinosinusitis without nasal polyps (CRsNP; sinusitis/chronic sinusitis/sinus disorder/respiratory disorder), chronic spontaneous urticaria (CSU; recurrent angioedema), cold urticaria, ulcerative colitis, eosinophilic esophagitis, eosinophilic gastritis/eosinophilic duodenitis/eosinophilic gastrointestinal disease/eosinophilic gastritis/eosinophilic gastroenteritis, hand eczema, keloid, moderate to severe atopic hand and foot dermatitis, nasal polyps, neurodermatitis, peanut allergy, prostate cancer, and pruritus.
In particular, the present invention further relates to the liquid antibody formulation disclosed herein for use in the treatment of atopic dermatitis (AD), e.g. moderate to severe atopic dermatitis, atopic eczema, asthma, e.g. moderate to severe eosinophilic or oral steroid dependent asthma, chronic rhinosinusitis with nasal polyposis (CRSwNP), chronic rhinosinusitis without nasal polyps (CRsNP), eosinophilic esophagitis (EoE), prurigo nodularis (PN), skin infections, chronic obstructive pulmonary disease (COPD), chronic spontaneous urticaria (CSU), chronic inducible cold urticaria (CINDU), allergic fungal rhinosinusitis (AFRS).
It is to be understood that the embodiments disclosed herein can be combined with each other for an individualized embodiment that is also disclosed as part of the invention.
The invention is further illustrated by the following examples.
Examples
Formulation development of high concentrated dupilumab formulations
Example 1: Method panel
The following method panel for analytical characterization of high concentrated dupilumab formulations was used throughout the examples. All of the following methods were performed at 25 °C unless otherwise indicated.
Analysis of protein content by UV-VIS
The concentration of dupilumab was determined by absorption spectroscopy at 280 nm with correction for any aggregated particles at 320 nm using a Nanophotometer N120 from Implen. Formulated drug substance (DS) was diluted with the respective placebo solution and measured against the respective placebo solution. Protein concentrations were calculated using the absorption coefficient of 1.412 L*g'1*cm'1. Concentrations were reported based on the mean of three replicate measurements.
Analysis of acidic and basic species of dupilumab by capillary zone electrophoresis (CZE)
For the CZE method, separation was performed by forward injection in a neutral, bare fused silica capillary (20 cm effective length and 50 pm diameter) with a PA800 plus instrument from ABSciex and using the standard kit from Sciex for CZE analysis. Protein
separation was performed by applying a voltage of 30 kV for 10 min. UV absorption was measured at 214 nm using the DAD (diode array detector) with 4 Hz data collection rate. The capillary temperature was kept constant at 25°C for all steps. Data were evaluated in terms of peak integration using the 32Karat software (Beckman Coulter). Peak areas were determined as velocity-corrected relative peak areas, reporting acidic species and basic species of dupilumab.
Analysis of HMWS by size exclusion chromatography SE-HPLC
5 pg of formulated dupilumab (diluted to 1 mg/mL with mobile phase 50 mmol/L sodium phosphate, 300 mmol/L sodium chloride, pH 6.8) were separated on a TSKgel® UP- SW3000 SEC column (4.6 x 150 mm, 2 pm, from Tosoh Bioscience) using a ThermoFisher Ultimate 3000 HPLC system with an isocratic flow of 0.35 mL/min (mobile phase: 50 mmol/L sodium phosphate, 300 mmol/L sodium chloride, pH 6.8) to analyze presence of high molecular weight species of dupilumab (HMWS) during a run time of 8 min. Data were recorded at 280 nm by a UV detector. Chromatogram profiles were compared and evaluated for HMWS and main peak relative area. All measurements were performed as single measurement.
Analysis of LMWS by non-reducing CE-SDS (LabChip)
Non-reducing capillary electrophoresis (nRCE-SDS or non-reducing cGE) was performed to quantify LMWS in dupilumab formulations, using a Perkin Elmer Labchip GXII Touch Protein Characterization System, a HT Protein Express Chip (760499, Perkin Elmer) and Protein Express assay reagent kit (CLS960008, Perkin Elmer). The sample preparation was performed according to the manufacturer general instruction as described in the Kit handbook (PN CLS140159, Rev. F) and following:
Formulated dupilumab samples were diluted with water to 1 mg/mL and 10 pl thereof were further diluted with 35 pl of non-reducing denaturation solution. Non-reducing conditions were established with stocks of alkylating reagent, namely 200 n- Ethylmaleimide (NEM, cat#E1271, from Sigma Aldrich) added at 1 :20 (v/v) ratio to the sample buffer provided with the kit. Denaturation solution was supplemented with lithium dodecyl sulfate (LDS, #L4632, from Sigma Aldrich) to a final concentration of 1%. Sample denaturation was performed at 75 °C for 10 min in a Bio-Rad real-time PCR cycler without shaking. Analysis of samples was performed in triplicate wells at the assay
concentration of 0.091 pg/pl. Electrophoretic separation was performed using the LabChip GXII Touch HT software, running the preset P200 Antibody Analysis method (Perkin Elmer). Raw data were exported and analyzed using Chromeleon software suite, version 7.2.10 from Thermo Fisher. The peak results were evaluated as normalized relative peak area (RFU). Normalization was achieved by division of the peak integral (RFU*min) by the peak migration time. Electropherogram profiles as well as purity (main peak) and impurity (LMWS, HMWS) content was monitored.
Visual appearance
Visual appearance of samples was assessed using an inspection light box equipped with non-flickering fluorescent lamps and a black and a white background plate (Portable Inspection Hood MIH-PORT, Bosch). The samples were evaluated by in their storage container without magnification to assess sample appearance with respect to visible particles, color and clarity. pH measurement
Determination of pH was performed with 150 pl aliquots of the samples and either calibrated Ultra Micro ISM pH-electrode or Micro Pro-ISM pH-electrode (both Mettler Toledo) connected to a SevenExcellence pH-Meter (Mettler Toledo) at room temperature. Samples were equilibrated at room temperature for at least 30 min before measurement.
Determination of dynamic viscosity
Measurements were performed at 20°C on a Kinexus ultra plus rheometer and using a cone-plated geometry (diameter: 40 mm; cone angle: 0.5°; sample volume 160 pl). The samples were equilibrated for 2 minutes at 20°C prior to each measurement. After ramping up from 0.1 to 1000 s’1, the dynamic viscosity was measured for one minute at constant shear rate of 1000 s’1. All samples were analyzed as single measurements.
Determination of Osmolality
Osmolality was determined by freezing point depression on an osmometer (Osmomat 030 D-RS from Gonotec, Germany. Calibration was performed using sodium chloride standards (500 and 850 mOmol/kg) and purified water. Samples were measured diluted at 50 mg/mL dupilumab concentration (50 pl sample volume) and recalculated. Dilution was performed by using purified water.
Determination of hydrodynamic radius by DLS
The hydrodynamic radius of samples containing dupilumab was determined by dynamic light scattering (DLS) at 25°C, using a DynaPro Plate Reader from Wyatt Technology (Santa Barbara, California, USA). Degradation products of samples can be either fragments with smaller radii than the radius of the monomer of dupilumab, or aggregates with higher radii. Samples were diluted to 2 mg/mL (diluted with 0.02 pm filtered water). Analysis was performed using the software Dynamics from Wyatt (version 8.1.2.144). Two replicates of each sample were transferred to a 384 well plate and measured twice. 35 acquisitions of 1 second each were performed. The radii for each population (x-axis) and the D50 value of all particles measured (y-axis) were plotted in a log-log scatter plot.
Diffusion Interaction Parameter ko (using DLS)
Protein-protein interactions were quantified by ko by the use of a DLS (dynamic light scattering) system from Wyatt Technology (Santa Barbara, California, USA), ko value is a measure of the balance between repulsive and attractive intermolecular forces. A positive ko value reveals repulsive forces between the proteins in solutions and hence a reduced tendency for aggregation as the surface net charge of the proteins keeps them separated, negative values for ko are related to attractive net forces between the molecules.
Before analysis all samples were 0.02 pm filtered and diluted with 0.02 pm filtered placebo solution to seven concentrations ranging from 1 mg/mL to 10 mg/mL. For analysis (using Dynamics software, version 8.1.2.144 from Wyatt) the default settings for refractive index and viscosity of water were used. Measurements were performed as duplicate measurement, each with 35 acquisitions of 1 second each as measurement parameter. KD value for each formulation was derived by linear regression using the equation D = Do (1+ko x c), whereas D = diffusion coefficient (cm2/s), Do = diffusion coefficient (cm2/s) at c = 0, ko = diffusion interaction parameter (mL/mg), c = concentration of dupilumab.
Determination of Turbidity (according to Ph.Eur. Method 8.0/2.02.01.00)
For experiments using the NEPHELOstar Plus, 200 pl of turbidity standards or samples were pipetted into the 96 well transparent microplate. The measurement was performed in duplicates, resulting in 2 technical replicates. Turbidity was measured with
Nepheolostar by Omega Software and selected Method: “LC-Standard_Methode_MT”. Analysis was performed by using MARS data analysis software.
Melting point by Nano-Differential Scanning Flourometry (nano-DSF)
Nano-DSF measurements were performed on a NanoTemper Prometheus NT.plex in 24- channel glass capillaries with the PR.ThermControl software (v2.3.1). Conformational stability of dupilumab was determined by intrinsic fluorescence measurement at 330 nm and 350 nm during thermal unfolding with a l°C/min temperature ramp from maximum 25°C to 95°C. Samples were analyzed with PR.Stability Analysis software (vl.l) and compared regarding the onset and midpoint of protein unfolding, TON and Tm, respectively. TON and inflection points were determined manually by identifying the transition and inflection points respectively on the curves with the PR.ThermControl software. Samples were measured diluted to 10 mg/mL in duplicates.
Subvisible Particles (SvP, According to USP Method <1787>)
Micron-sized protein aggregates and particles (subvisible particles, SvP) are important quality attributes of therapeutic protein formulations due to their risk of enhancing an immunogenic response. Hence, pharmacopoeias require quantification of SvP larger than 10 pm and 25 pm. Currently, the pharmacopeial acceptance criteria for parenteral administration in small volume measured by light obscuration are: NMT 6000 particles > 10 pm per container and NMT 600 particles > 25 pm per container. Moreover, SvP < 10 pm must be monitored, however, specific acceptance criteria are not defined.
Here, formulations were analyzed by flow imaging microscopy using a FlowCam® 8100 system (ANASYSTA). This method captures SvP more efficiently compared to light obscuration since it is more sensitive at detecting particles with refractive indices similar to the solvent. To achieve an optimal flow imaging efficiency in the range of 60-70 %, the following analysis setting was used: 0.070 ml/min flow rate with imaging rate of 13 frames per second.
For the analysis of formulations 120 pl of sample was manually injected, of which -100 pl was imaged by the system. The SvP data was size grouped according to EP/USP utilizing the following filters: Total particle count, above 1 pm, above 2 pm, above 5 pm, above 10 pm, above 25 pm, above 50 pm, above 100 pm, above 250 pm. The SvP of all formulations at all storage time points was determined. For each formulation two
technical replicates were measured non-diluted.
Quantification of oxidized species of dupilumab by reversed phase high performance liquid chromatography (RP-HPLC)
Reversed phase high performance liquid chromatography (RP-HPLC) was performed to quantify oxidized species in dupilumab formulations.
For this formulated dupilumab samples were diluted to a final concentration of approx. 0.4 g/L during the IdeS digestion and reduction. IdeS digestion and reduction was proceeded as follows:
12 pl of dupilumab samples were mixed with 3.75 pl of the IdeS solution (66 U/pl of FabRICATOR reagent (Genovis, # GKPC-503), solved in water) as well as 259 pl of SVP 50 solution (50 mmol/L ammonium hydrogen carbonate in water). Samples were 30 minutes incubated for digestion at 37°C and 500 rpm. After digestion the solution was mixed with 6 pl IM DDT (in water) and 275 pl TFE solution (Sigma-Aldrich, #91683). After this, samples were incubated for further 15 minutes at a temperature of 60°C and 500 rpm. The reduction was stopped by adding 62 pl of 10% formic acid (solved in water).
The measurement was performed using a Waters Aquity UPLC H-class system and Empower 3 software, also from Waters.
For this 2.4 pg of digested samples were separated on a BioResolve RP mAb Polyphenyl column 450A (2.7 pm; 1.1 x 150 mm) from Waters, using a gradient elution of two mobile phases:
Mobile Phase A: 0.1 % TFA in water
Mobile phase B: 0.1 % TFA in acetonitrile
Separation was performed at a flow rate of 0.6 mL/min at a column temperature of 80°C using following gradient:
Data were recorded at 214 nm using a UV detector from Waters. After reduction three fragment peaks were detected. The peaks directly before the main fragments were integrated as oxidations of the fragments and reported as relative areas [%]. All measurements were performed as duplicate measurements.
Example 2: Liquid formulations of dupilumab selected for stability study
Liquid formulations of dupilumab adjusted to 150 mg/mL as well as 175 mg/mL dupilumab were prepared as set forth in Table 1. In addition, marketed reference formulations of Dupixent® will be tested. Table 1 : Detailed information of formulations prepared within this study
Example 3: Stability study of liquid formulations of dupilumab
The formulations of Table 1 are tested according to Table 2 in a 12-month long-term stability at target storage condition 2°C to 8°C (i.e. 5°C ± 3°C) as well as accelerated conditions at 25°C/60% RH for up to 6 months and at 40°C/75% RH for up to 1 month. In addition, samples will undergo mechanical stressing as well as five freeze/thaw cycles.
X Only 175 mg/mL formulations analyzed
X* 175 mg/mL and 150 mg/mL formulations analyzed
Protein stability was determined by size exclusion chromatography (SE-HPLC) for the presence of high molecular weight species (HMWS) and by non-reducing SDS-cGE (nRCE-SDS) for the presence of low molecular weight species (LMWS). CZE was used to detect modifications leading to charge heterogeneities, i.e. to detect acidic and basic species. Oxidation was monitored by RP-HPLC chromatography after IdeS digestion. DLS was analyzed, also to calculate ko values for characterizing protein-protein interactions. Melting temperature (Tm) and aggregation temperature (Tagg) were
analyzed by nano-differential scanning fluorometry (nano-DSF). In addition, samples were analyzed for visual appearance, dynamic viscosity, turbidity, sub-visible particle content and particle size. Osmolality of the samples was determined. The protein concentration of the samples was determined by UV-VIS spectroscopy. Also, pH was monitored during the stability study.
Results of the stability study of liquid formulations of dupilumab are as follows.
Protein concentration
Target concentration (150 mg/mL and 175 mg/mL ± 10%) post upconcentration was achieved for all formulations. Osmolality
Osmolality measurements were performed for all formulations of Table 1. Osmolality for all formulations is within the range of 260 to 390 mOsm/kg.
1111
All formulations maintained stable pH over storage time for up to 9 months. Viscosity
All formulations tend to exhibit reduced viscosity compared to the reference formulations and are within a suitable range for subcutaneous injections.
Visual appearance
All formulations were inconspicuous over storage time for up to 9 months. Visual appearance of all samples was slightly viscous, slightly opalescent, slightly yellowish, and clear for all conditions. Turbidity
All formulations show low turbidity after preparation and after storage for 1 month, thus complying with visual appearance. After storage for up to 9 months most formulations showed an acceptable and expected slight increase in turbidity.
Colloidal stability The ko value as measured by dynamic light scattering (DLS) is indicative of colloidal stability. Measurements of the ko value was performed for all formulations of Table 1, the results are provided in Table 4.
Table 4: ko value of formulations prepared within this study along with adjusted correlation coefficient R2
The ko values are observed with high confidence explaining >85% of the data.
As shown in Table 4 most formulations of the invention exhibit less negative (i.e. more positive) ko values as compared to the reference formulations.
Physical stability - HMWS/Stabilization of main peak
SE-HPLC was performed to determine the main peak and HMWS of dupilumab in the formulations of Table 1, the results are provided in
Table 5.
Table 5: HMWS and main peak of dupilumab as Arei.area [%] in the formulations prepared within this study
5 As shown in Table 5 formulations of the invention exhibit enhanced physical stability as compared to the reference formulations.
Physical stability - LMWS/Stabilization of main peak nRCE-SDS was performed to determine the main peak and LMWS of dupilumab in the formulations of Table 1, the results are provided in Table 6.
Table 6: LMWS and main peak of dupilumab as Arei.area [%] in the formulations prepared within this study
5 As shown in Table 6 formulations of the invention exhibit enhanced physical stability as compared to the reference formulations.
Chemical stability - Acidic and basic variants/Stabilization of main peak
CZE was performed to determine the main peak, acidic species/acidic variants (AV) and basic species/basic variants (BV) of dupilumab in the formulations of Table 1, the results are provided in Table 7.
Table 7: Acidic variants, main peak and basic variants of dupilumab as Arei.area [%] in the formulations prepared within this study
5 As shown in Table 7 formulations of the invention exhibit enhanced chemical stability as compared to the reference formulation.
Example 4: Characterization of liquid dupilumab formulations with or without sugar
Liquid formulations of dupilumab adjusted to 175 mg/mL dupilumab were prepared as set forth in Table 8. Formulation WP6 01 corresponds to the marketed formulation of Dupixent® comprising 175 mg/mL dupilumab.
The formulations of Table 8 are characterized as follows.
Protein concentration
Target concentration (175 mg/mL ± 10%) post upconcentration was achieved for all formulations.
Osmolality
Osmolality measurements were performed for all formulations of Table 8. Osmolality for all formulations was within the range of 290 to 360 mOsm/kg.
Viscosity Table 9: Viscosity measured at constant shear rate.
Formulation no. Shear.viscosity.mPa.s
WP6 01 17.2±0.1
WP6_02 12.0±0.2
WP6_03 20.0±0.1
WP6_04 15.2±0.0
Formulations WP6 01 and WP6 03 containing sucrose tend to exhibit increased viscosity. The syringeability of formulations WP6 01 and WP6 03 might not be optimal.
Colloidal stability
Measurements of the ko value were performed for all formulations of Table 8, the results are provided in Table 10.
Table 10: ko value of formulations prepared within this study along with adjusted correl ati on coeffi ci ent R2
Formulation no. kD*1000 Adj.R.squared
WP6 01 -10.89 0.96
WP6_02 -5.29 0.94
WP6 03 -11.56 0.95
WP6_04 -5.22 0.89
The ko values are observed with high confidence explaining > 89% of the data.
As shown in Table 11, lowest intermolecular interaction is observed in formulations WP6 02 and WP6 04; containing high concentration of arginine.
Claims
1. A liquid antibody formulation comprising:
(a) dupilumab,
(b) L-arginine at a concentration of from 150 to 300 mM,
(c) a surfactant, and
(d) optionally a buffer, wherein the pH of the liquid antibody formulation is from 5.0 to 7.0.
2. The liquid antibody formulation of claim 1, comprising the antibody at a concentration of from 150 to 200 mg/mL, preferably at a concentration of 150 mg/mL or 175 mg/mL.
3. The liquid antibody formulation of claim 1 or claim 2, comprising L-arginine at a concentration of from 150 to 250 mM, preferably at a concentration of from 150 to 230 mM, preferably at a concentration of from 170 mM.
4. The liquid antibody formulation of any one of the preceding claims, wherein the buffer is L-histidine/L-histidine HC1 or succinic acid/ succinate, preferably wherein the buffer is succinic acid/succinate.
5. The liquid antibody formulation of any one of the preceding claims, comprising the buffer at a concentration of from 5 to 20 mM, preferably at a concentration of 10 mM and/or comprising the surfactant at a concentration of from 0.1 to 4.0 mg/mL, preferably at a concentration of 0.2 mg/mL or 0.5 mg/mL or 2.0 mg/mL, preferably at a concentration of 2.0 mg/mL.
6. The liquid antibody formulation of any one of the preceding claims, wherein the surfactant is polyoxyethylene(20)sorbitan monolaurate, polyoxy- ethylene(20)sorbitan monooleate or a polyoxyethylene-polyoxypropylene-block polymer, preferably a polyoxyethylene-polyoxypropylene-block polymer of the following formula (I)
wherein x is an integer from 5 to 146, y is an integer from 18 to 60, and z is an integer from 5 to 146, preferably wherein x and z = 75, and y=30, more preferably wherein the surfactant is polyoxy ethylene(20)sorbitan monooleate.
7. The liquid antibody formulation of any one of the preceding claims, wherein the formulation does not comprise sucrose.
8. The liquid antibody formulation of any one of the preceding claims consisting of
(a) dupilumab, preferably at a concentration of 150 mg/mL or 175 mg/mL,
(b) L-arginine at a concentration of from 150 to 250 mM, preferably 170 mM,
(c) a buffer, selected from the group consisting of L-histidine/L-histidine HC1 and succinic acid/succinate, preferably succinic acid/succinate, preferably at a concentration of 10 mM.
(d) a surfactant, selected from the group consisting of polyoxy- ethylene(20)sorbitan monolaurate, polyoxy ethylene(20)sorbitan monooleate, and a polyoxyethylene-polyoxypropylene-block polymer of the following formula (I)
wherein x and z = 75, and y=30, preferably polyoxyethylene(20)sorbitan monooleate, preferably at a concentration of 2.0 mg/mL, and optionally HC1 for adjusting the pH, wherein the pH of the liquid antibody formulation is from 5.5 to 6.2, preferably about 5.9.
9. The liquid antibody formulation of any one of the preceding claims, consisting of
(a) dupilumab at a concentration of 150 mg/mL or 175 mg/mL,
(b) L-arginine at a concentration of from 150 to 250 mM, preferably 170 mM,
(c) succinic acid/succinate at a concentration of 10 mM,
(d) polyoxyethylene(20)sorbitan monooleate at a concentration of 2.0 mg/mL, and optionally HC1 for adjusting the pH, wherein the pH of the liquid antibody formulation is from 5.5 to 6.2 preferably about 5.9.
10. The liquid antibody formulation of any one of the preceding claims, which is a stable liquid antibody formulation.
11. The liquid antibody formulation of any one of the preceding claims for use in the treatment of moderate to severe atopic dermatitis (AD), atopic eczema, moderate to severe eosinophilic or oral steroid dependent asthma, chronic rhinosinusitis with nasal polyposis (CRSwNP), eosinophilic esophagitis (EoE), prurigo nodularis (PN), skin infections, and chronic obstructive pulmonary disease.
12. A method of preparing the liquid antibody formulation of any one of the preceding claims comprising mixing (a) an antibody, preferably dupilumab, (b) L-arginine, and (c) a surfactant, and optionally (d) a buffer.
13. A dosage form comprising a device preferably selected from a pre-filled syringe, an autoinjector, a pre-filled pen, a pre-filled cartridge for on-body injector, a glass vial, and a microinfuser, said device containing a liquid antibody formulation according to any of claims 1 to 11, wherein the amount of the formulation is adapted to the antibody dosage to be provided.
14. A set of dosage forms comprising a plurality of dosage forms according to claim 13, wherein the set comprises at least three dosage forms containing the same antibody formulation in different volumes.
15. Use of arginine at a concentration of from 110 to 320 mM for stabilization of dupilumab in a liquid antibody formulation.
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23185958.8 | 2023-07-17 | ||
| EP23185959.6 | 2023-07-17 | ||
| EP23185958 | 2023-07-17 | ||
| EP23185959 | 2023-07-17 | ||
| EP23211549.3 | 2023-11-22 | ||
| EP23211549 | 2023-11-22 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025017034A1 true WO2025017034A1 (en) | 2025-01-23 |
Family
ID=91898549
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/070169 Pending WO2025017034A1 (en) | 2023-07-17 | 2024-07-16 | Stable liquid antibody formulation for dupilumab |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025017034A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP2624865A1 (en) | 2010-10-06 | 2013-08-14 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies |
| WO2023011502A1 (en) * | 2021-08-02 | 2023-02-09 | 甘李药业股份有限公司 | Stable formulation comprising anti-il-4r antibody |
| US20230348532A1 (en) | 2022-03-02 | 2023-11-02 | Regeneron Pharmaceuticals, Inc. | Manufacturing process for high titer antibody |
| WO2023215267A1 (en) * | 2022-05-02 | 2023-11-09 | Regeneron Pharmaceuticals, Inc. | Anti-interleukin-4 receptor (il-4r) antibody formulations |
-
2024
- 2024-07-16 WO PCT/EP2024/070169 patent/WO2025017034A1/en active Pending
Patent Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP2624865A1 (en) | 2010-10-06 | 2013-08-14 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies |
| EP2624865B1 (en) | 2010-10-06 | 2018-08-01 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies |
| WO2023011502A1 (en) * | 2021-08-02 | 2023-02-09 | 甘李药业股份有限公司 | Stable formulation comprising anti-il-4r antibody |
| US20230348532A1 (en) | 2022-03-02 | 2023-11-02 | Regeneron Pharmaceuticals, Inc. | Manufacturing process for high titer antibody |
| WO2023215267A1 (en) * | 2022-05-02 | 2023-11-09 | Regeneron Pharmaceuticals, Inc. | Anti-interleukin-4 receptor (il-4r) antibody formulations |
Non-Patent Citations (4)
| Title |
|---|
| EMA: "CHMP assessment report on extension of marketing authorisation and an extension of indication variation Dupixent", 28 February 2019 (2019-02-28), pages 1 - 161, XP093211800, Retrieved from the Internet <URL:https://www.ema.europa.eu/en/documents/variation-report/dupixent-h-c-4390-ii-0012-epar-assessment-report-variation_en.pdf> * |
| FIEDLER: "Lexikon der Hilfsstoffe für Pharmazie, Kosmetik und angrenzende Gebiete", vol. 2, 2002 |
| REN STEVEN: "Effects of arginine in therapeutic protein formulations: a decade review and perspectives", vol. 6, no. 4, 12 October 2023 (2023-10-12), GB, pages 265 - 276, XP093133197, ISSN: 2516-4236, Retrieved from the Internet <URL:https://academic.oup.com/abt/article-pdf/6/4/265/54152757/tbad022.pdf> DOI: 10.1093/abt/tbad022 * |
| STRICKLEY R.G.LAMBERT W.J.: "A review of Formulations of Commercially Available Antibodies", J PHARM SCI, vol. 110, no. 7, July 2021 (2021-07-01), pages 2590 - 2608, XP055799313, DOI: 10.1016/j.xphs.2021.03.017 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP5631591B2 (en) | Stable antibody formulation | |
| AU2007240732B2 (en) | Buffering agents for biopharmaceutical formulations | |
| JP5577098B2 (en) | Stable buffered formulations containing polypeptides | |
| KR20170005864A (en) | Liquid pharmaceutical composition | |
| TWI787161B (en) | Pharmaceutical composition comprising anti-human tslp receptor antibody | |
| Garidel et al. | Stability of buffer-free freeze-dried formulations: A feasibility study of a monoclonal antibody at high protein concentrations | |
| JP7407723B2 (en) | Pharmaceutical composition containing human anti-IL-33 monoclonal antibody | |
| WO2020173431A2 (en) | Preparations containing anti-cd47 antibody, and preparation method and use therefor | |
| JP2023512961A (en) | Stable anti-PD-1 antibody pharmaceutical formulation | |
| EP4070817A1 (en) | Liquid preparation containing anti-il-17 antibody | |
| TWI857996B (en) | Protein solution formulation containing high concentration of an anti-vegf antibody | |
| AU2019274826A1 (en) | Stable fusion protein formulation | |
| CN114146174B (en) | Anti-PD-L1/OX40 bispecific antibody preparation and preparation method and use thereof | |
| AU2019274827A1 (en) | CTLA4-Ig fusion protein formulation | |
| CN117545501A (en) | Methods to improve the stability of antibody preparations | |
| JP7755766B2 (en) | Anti-IL-6 antibody preparation | |
| KR20240053633A (en) | Preparations for VEGF receptor fusion proteins | |
| CN111683681B (en) | Formulations comprising anti-OX 40 antibodies, methods of making, and uses thereof | |
| WO2025017034A1 (en) | Stable liquid antibody formulation for dupilumab | |
| EP3434283A1 (en) | Medicinal composition comprising peg anti-human ngf antibody fab' fragment | |
| EA045592B1 (en) | LIQUID PREPARATION CONTAINING ANTIBODY TO IL-17 | |
| TW202128220A (en) | Stable liquid composition, preparation method therefor and formulation containing the same | |
| HK1259609A1 (en) | Pharmaceutical composition comprising peglated fab' fragment of anti-human ngf antibody |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24740483 Country of ref document: EP Kind code of ref document: A1 |