WO2015057910A1 - Buffer formulations for enhanced antibody stability - Google Patents

Buffer formulations for enhanced antibody stability Download PDF

Info

Publication number
WO2015057910A1
WO2015057910A1 PCT/US2014/060810 US2014060810W WO2015057910A1 WO 2015057910 A1 WO2015057910 A1 WO 2015057910A1 US 2014060810 W US2014060810 W US 2014060810W WO 2015057910 A1 WO2015057910 A1 WO 2015057910A1
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
buffer
antibody
buffered
antibody formulation
Prior art date
Application number
PCT/US2014/060810
Other languages
French (fr)
Inventor
John Cini
Athena NAGI
Maria TADDEI
Original Assignee
Oncobiologics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oncobiologics, Inc. filed Critical Oncobiologics, Inc.
Priority to JP2016549199A priority Critical patent/JP6526025B2/en
Priority to EP14854535.3A priority patent/EP3057616B1/en
Priority to AU2014337263A priority patent/AU2014337263B2/en
Priority to CA2926588A priority patent/CA2926588C/en
Priority to MX2016004926A priority patent/MX2016004926A/en
Priority to CN201480068235.2A priority patent/CN106170298B/en
Priority to US15/025,088 priority patent/US10376582B2/en
Publication of WO2015057910A1 publication Critical patent/WO2015057910A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype

Definitions

  • the invention relates generally to the field of antibody formulation chemistry. More particularly, the invention relates to buffered formulations for antibody storage, which enhance the thermal stability, conformational and colloidal sta bility of the antibody, thereby enhancing long term storage of the antibody.
  • a biological drug product (produced in or derived from living organisms) may be demonstrated to be "biosimilar” if data show that, among other things, the product is "highly similar” to an already-approved biological product.
  • the biosimilar product should retain at least the biologic function a nd treatment efficacy of the U.S. Food and Drug Agency-approved biological product.
  • the biosimilar product may be formulated differently, however, from the a pproved biological product. The formulation may improve stability a nd shelf storage of the biologic drug product, and may also improve the efficacy in treating a particular disease or condition.
  • the formulation may also improve other aspects of administration, including a reduction in patient discomfort or other untoward effects that a patient may experience upon administration of the approved biological product.
  • Antibody molecules may be used as biological drugs, and many such antibodies are approved for use in human beings. Antibody molecules may be produced as a biosimilar, and reformulated accordingly. There remains a need in the art for high-quality antibody biosimilars.
  • the invention features buffered antibody formulations, comprising (a) an antibody.
  • the antibody may specifically bind to tumor necrosis factor alpha.
  • the antibody may comprise a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2.
  • the formulation in addition to the antibody, comprises (b) an aqueous buffer comprising from about 0.7 mM to about 1.3 mM of an acetate salt, preferably sodium acetate trihydrate, from about 200 mM to about 206 mM of mannitol, from about 16 mM to about 22 mM of glacial acetic acid, and from about 24 mM to about 28 mM of sodium chloride, and (c) about 0.07% (v/v) to about 0.15% (v/v) of a non-ionic surfactant such as polysorbate 80.
  • the buffered antibody formulation has a pH of from about 5.1 to about 5.3, preferably about 5.2.
  • the formulation comprises from about 30 mg to about 50 mg of the antibody. In some preferred aspects, the formulation comprises from about 35 mg to about 45 mg of the antibody. In some preferred aspects, the formulation comprises from about 37 mg to about 43 mg of the antibody. In some preferred aspects, the formulation comprises about 40 mg of the antibody.
  • the buffer may comprise from about 0.8 mM to about 1.2 mM of sodium acetate trihydrate, or from about 0.9 mM to about 1.1 mM of sodium acetate trihydrate, or about 1 mM of sodium acetate trihydrate.
  • the buffer may comprise from about 201 mM to about 205 mM of mannitol, or from about 202 mM to about 204 mM of mannitol, or about 203 mM of mannitol.
  • the buffer may comprise from about 17 mM to about 21 mM of glacial acetic acid, or from about 18 mM to about 20 mM of glacial acetic acid, or about 19 mM of glacial acetic acid.
  • the buffer may comprise from about 25 mM to about 27 mM of sodium chloride, or about 26 mM of sodium chloride, or about 27 mM of sodium chloride, or about 26.35 mM of sodium chloride.
  • the buffered antibody formulation includes a non-ionic surfactant, which preferably is polysorbate 80.
  • the formulation comprises from about 0.08% (v/v) to about 0.12% (v/v) of polysorbate 80. In some aspects, the formulation comprises from about 0.09% (v/v) to about 0.11% (v/v) of polysorbate 80. In some aspects, the formulation comprises about 0.1% (v/v) of polysorbate 80.
  • a buffered antibody formulation comprises (a) about 30 mg to about 50 mg of an antibody comprising a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ. ID NO: 2, (b) a buffer comprising about 1 mM of an acetate salt, preferably sodium acetate trihydrate, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and (c) about 0.1% (by volume) of polysorbate 80.
  • the buffered antibody formulation has a pH of from about 5.1 to about 5.3, preferably about 5.2.
  • the formulation comprises from about 35 mg to about 45 mg of the antibody.
  • the formulation comprises from about 37 mg to about 43 mg of the antibody.
  • the formulation comprises about 40 mg of the antibody.
  • the buffered antibody formulations may be used as a medicament, and may be used in methods of treatment.
  • the buffered antibody formulations may be for use in the treatment of arthritis.
  • the buffered antibody formulations may be for use in the treatment of Rheumatoid Arthritis, or Juvenile Idiopathic Arthritis, or Psoriatic Arthritis.
  • the buffered antibody formulations may be for use in the treatment of Ankylosing Spondylitis.
  • the buffered antibody formulations may be for use in the treatment of Crohn's Disease.
  • the buffered antibody formulations may be for use in the treatment of Ulcerative Colitis.
  • the buffered antibody formulations may be for use in the treatment of Plaque Psoriasis.
  • the methods of treatment include methods for treating arthritis, including
  • the methods of treatment also include methods for treating Ankylosing Spondylitis, methods for treating Crohn's Disease, methods for treating Plaque Psoriasis, and methods for treating Ulcerative Colitis.
  • methods of treatment comprise administering to an arthritis patient, including a Rheumatoid Arthritis, Juvenile Idiopathic Arthritis, or Psoriatic Arthritis, an amount of the buffered antibody formulations described or exemplified herein effective to treat the arthritis in the patient.
  • methods of treatment comprise administering to an Ankylosing Spondylitis patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Ankylosing Spondylitis in the patient.
  • methods of treatment comprise administering to a Crohn's Disease patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Crohn's Disease in the patient.
  • methods of treatment comprise administering to an Ulcerative Colitis patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Ulcerative Colitis in the patient.
  • methods of treatment comprise administering to a Plaque Psoriasis patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Plaque Psoriasis in the patient.
  • the buffered antibody formulations are preferably administered subcutaneously to the patient, for example, by subcutaneous injection.
  • the patient preferably is a human being.
  • kits which may be used, for example, in accordance with the methods of treatment.
  • the kits generally comprise any of the buffered antibody formulations described or exemplified herein and instructions for using the formulation in a method of treatment.
  • the method of treatment may be a method for treating arthritis.
  • the method of treatment may be a method for treating Rheumatoid Arthritis.
  • the method of treatment may be a method for treating Juvenile Idiopathic Arthritis.
  • the method of treatment may be a method for treating Psoriatic Arthritis.
  • the method of treatment may be a method for treating Ankylosing Spondylitis.
  • the method of treatment may be a method for treating Crohn's Disease.
  • the method of treatment may be a method for treating Ulcerative Colitis.
  • the method of treatment may be a method for treating Plaque Psoriasis.
  • the kits may include a device for administering the antibody formulation to a patient.
  • the device may comprise a syringe a nd a needle.
  • the device may comprise a catheter.
  • Fig. 1 shows an overlay of SE-UPLC chromatograms from representative
  • Fig. 2 shows trends in SE-UPLC % high molecular weight species (HMWS) as a function of solution pH.
  • Fig. 3 shows a DSC thermograms for representative experimental series 1 formulation conditions.
  • Fig. 4 shows a DLS and pH ranges for formulation solutions at 50 mg/ml protein concentration, coded by buffer composition.
  • Fig. 5 shows an overlay of SE-UPLC chromatograms for the adalimumab reference formulation under duration of stressed stability experiment (55° C up to 14 days).
  • Fig. 6 shows an overlay of representative SE-UPLC chromatograms from
  • Fig. 7 shows trends in SE-UPLC aggregation over time at stressed conditions.
  • Fig. 8 shows trends in SE-UPLC fragmentation over time at stressed conditions.
  • Fig. 9 shows ONS-3010 experimental series 3 DLS results.
  • Fig. 10 shows an overlay of SE-UPLC chromatograms for the adalimumab reference formulation under duration of stressed stability experiment (55° C up to 7 days).
  • Fig. 11 shows an overlay of representative SE-UPLC chromatograms from
  • Fig. 12 shows trends in experimental series 3 SE-UPLC aggregation over time at stressed conditions.
  • Fig. 13 shows an overlay of CEX-HPLC chromatograms for 55° C-incubated samples in the adalimumab reference formulation, days 0, 1, and 2.
  • Fig. 14 shows CEX-HPLC Main Peak Percentages for 55° C-incubated samples up to 2 days.
  • Fig. 15 shows CEX-HPLC Acidic Peak Percentages for 55° C-incubated samples up to 2 days.
  • Fig. 16 shows CEX-HPLC Basic Peak Percentages for 55° C-incubated samples up to 2 days.
  • Fig. 17 shows the total percentage of isomerized species in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition number).
  • Fig. 18 shows the percentage of cyclized N-terminal peptides in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition number).
  • Fig. 19 shows the total percentage of oxidized methionine peptides in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition number).
  • Fig. 20 shows the total percentage of deamidated peptides in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition num ber).
  • Fig. 21 shows an overlay of SE-UPLC chromatograms for adalimumab reference formulation samples incubated at 37° C, time zero and day 28.
  • Fig. 22 shows an overlay of SE-UPLC chromatograms for 37° C-incubated day 28 samples.
  • Fig. 25 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated day 28 samples.
  • Fig. 26 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated 5 month samples.
  • Fig. 27 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated 12 month samples, conditions 1 and 3.
  • Fig. 28 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated 18 month samples, conditions 1 and 3.
  • a nd include any a nimal.
  • Subjects include mamma ls, including companion and farm mammals, as well as rodents, including mice, rabbits, and rats, and other rodents.
  • rodents including mice, rabbits, and rats, and other rodents.
  • Non-human primates preferred subjects. Human beings are highly preferred subjects.
  • formulations of ONS- 3010 which specifically binds to tumor necrosis factor alpha, can be buffered with mannitol and acetate, while minimizing sodium chloride, with the buffers enhancing the thermal and colloidal stability of the antibody, even more so than formulations of adalimumab currently approved for patient use. It was observed that there is a fine balance in establishing and maintaining an acidic pH of about 5.2 with the appropriate salts and buffer components. It was observed, for example, that high levels of salt may induce aggregation and degradation, which could be improved by lowering the salt level. Accordingly, the disclosure features buffered formulations for antibodies, which formulations include an aqueous carrier comprising buffer comprising acetate and mannitol, as well as a non-ionic surfactant, but with minimal sodium chloride.
  • the antibody specifically binds to an epitope on tumor necrosis factor alpha, and the epitope may be linear or conformational.
  • the antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1.
  • the antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 2.
  • the antibody comprises a heavy chain constant domain and/or a light chain constant domain.
  • the antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, an example of which is ONS-3010.
  • the heavy and light chain amino acid sequences of the antibody may comprise those of U.S. Pat. No. 6,090,382.
  • the antibody is a full length antibody, comprising both variable and constant regions, although in some aspects, the antibody may comprise a derivative or fragment or portion of a full-length antibody that retains the antigen-binding specificity, and also preferably retains most or all of the affinity, of the full length antibody molecule.
  • the antibody may comprise post-translational modifications (PTMs) or moieties, which may impact antibody activity or stability.
  • PTMs post-translational modifications
  • the antibody may be methylated, acetylated, glycosylated, sulfated, phosphorylated, carboxylated, and/or amidated, and may comprise other moieties that are well known in the art.
  • Common PTMs for ONS-3010 include N- glycosylation, C-terminal variants (e.g., cleavage of lysine, proline amidation), N-terminal pyro-E formation, oxidation, isomerization, deamidation, succinimide formation,
  • Moieties include any chemical group or combinations of groups commonly found on immunoglobulin molecules in nature, or otherwise added to antibodies by recombinant expression systems, including prokaryotic and eukaryotic expression systems.
  • the formulation preferably comprises a therapeutically effective amount of an antibody.
  • the antibody may be any antibody compatible with the aqueous buffer formulation.
  • a preferred a ntibody comprises a heavy chain having the amino acid sequence of SEQ. I D NO: 1 and a light chain having the amino acid sequence of SEQ. I D NO: 2.
  • a therapeutically effective amount may vary, depending on the disease or condition being treated upon administration of the antibody, and/or depending on the characteristics of the subject to which the antibody is administered, such as age, gender, height, weight, state of advancement or stage of the disease or condition, the number and efficacy of previous administrations, other therapeutic agents administered to the subject, and other characteristics that are known to the practitioner or that would otherwise be taken into account in determining appropriate dosing.
  • a therapeutically effective amount is an amount that is effective to treat Rheumatoid Arthritis.
  • a therapeutically effective amount is an amount that is effective to treat Juvenile Idiopathic Arthritis, Psoriatic Arthritis, Ankylosing Spondylitis, Crohn's Disease, Plaque Psoriasis, Ulcerative Colitis, I nflammatory Bowel Disease, Hidradenitis Suppurativa, or Refractory Asthma.
  • the formulation may comprise from about 10 mg to about 70 mg of the antibody.
  • the formulation comprises from about 20 mg to about 60 mg of the antibody.
  • the formulation comprises from about 30 mg to about 50 mg of the antibody.
  • the formulation comprises from about 35 mg to about 45 mg of the antibody.
  • the formulation comprises from about 37 mg to a bout 43 mg of the antibody.
  • the formulation comprises from about 38 mg to about 42 mg of the antibody.
  • the formulation comprises from about 39 mg to about 41 mg of the antibody.
  • the formulation comprises from about 30 mg to about 60 mg of the antibody.
  • the formulation comprises from about 35 mg to about 55 mg of the antibody.
  • the formulation comprises from about 40 mg to about 60 mg of the antibody. These ranges include the lower and upper amounts that define the range.
  • the formulation comprises about 40 mg of the antibody.
  • the antibody is preferably formulated with a buffered aqueous carrier, and the carrier preferably comprises water.
  • the buffered antibody formulation is preferably in liquid form, and more preferably in liquid form suitable for subcutaneous administration. Thus, the amount of water in the buffered formulation may vary in accordance with the desired volume of the injectable bolus.
  • the buffer comprises sodium acetate trihydrate, mannitol, sodium chloride, glacial acetic acid, and a non-ionic surfactant, and maintains the antibody formulation at an acidic pH. When stored in the buffered formulation, the antibody is shelf-stable under normal storage conditions.
  • the buffer may comprise from about 0.1 mM to about 5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.3 mM to about 3 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.5 mM to about 2 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.5 mM to about 1.5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.6 mM to about 1.4 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.7 mM to about 1.5 mM of an acetate salt.
  • the buffer may comprise from about 0.7 mM to about 1.3 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.8 mM to about 1.2 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.8 mM to about 1.5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.8 mM to about 1.1 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.9 mM to about 1.2 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.9 mM to about 1.4 mM of an acetate salt.
  • the buffer may comprise from about 0.9 mM to about 1.1 mM of an acetate salt. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 1 mM of an acetate salt.
  • the acetate salt may comprise any suitable acetate salt. Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
  • the buffer may comprise from about 100 mM to about 300 mM of mannitol. In some aspects, the buffer may comprise from about 110 mM to about 290 mM of mannitol. In some aspects, the buffer may comprise from about 120 mM to about 280 mM of mannitol. In some aspects, the buffer may comprise from about 150 mM to about 250 mM of mannitol. In some aspects, the buffer may comprise from about 175 mM to about 225 mM of mannitol. In some aspects, the buffer may comprise from about 180 mM to about 220 mM of mannitol. In some aspects, the buffer may comprise from about 185 mM to about 215 mM of mannitol.
  • the buffer may comprise from about 190 mM to about 215 mM of mannitol. In some aspects, the buffer may comprise from about 195 mM to about 210 mM of mannitol. In some aspects, the buffer may comprise from about 197 mM to about 209 mM of mannitol. In some aspects, the buffer may comprise from about 198 mM to about 208 mM of mannitol. In some aspects, the buffer may comprise from about 198 mM to about 205 mM of mannitol. In some aspects, the buffer may comprise from about 199 mM to about 207 mM of mannitol.
  • the buffer may comprise from about 200 mM to about 210 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 207 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 206 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 205 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 203 mM of mannitol. In some aspects, the buffer may comprise from about 201 mM to about 205 mM of mannitol.
  • the buffer may comprise from about 201 mM to about 204 mM of mannitol. In some aspects, the buffer may comprise from about 201 mM to about 203 mM of mannitol. In some aspects, the buffer may comprise from about 202 mM to about 204 mM of mannitol. In some aspects, the buffer may comprise from about 202 mM to about 203 mM of mannitol. In some aspects, the buffer may comprise from about 202 mM to about 206 mM of mannitol. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 203 mM of mannitol.
  • the buffer may comprise from about 9 mM to about 30 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 10 mM to about 30 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 9 mM to about 29 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 10 mM to about 28 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 11 mM to about 27 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 12 mM to about 26 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 13 mM to about 25 mM of glacial acetic acid.
  • the buffer may comprise from about 14 mM to about 24 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 15 mM to about 23 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 15 mM to about 21 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 15 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 16 mM to about 22 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 16 mM to about 20 mM of glacial acetic acid.
  • the buffer may comprise from about 17 mM to about 21 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 17 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 19 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 23 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 19 mM to about 20 mM of glacial acetic acid. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 19 mM of glacial acetic acid.
  • the buffer preferably includes minimal amounts of sodium chloride, and in some aspects, includes no sodium chloride.
  • the buffer may comprise from about 15 mM to about 36 mM of sodium chloride.
  • the buffer may comprise from about 16 mM to about 36 mM of sodium chloride.
  • the buffer may comprise from about 18 mM to about 34 mM of sodium chloride.
  • the buffer may comprise from about 20 mM to about 32 mM of sodium chloride.
  • the buffer may comprise from about 22 mM to about 30 mM of sodium chloride.
  • the buffer may comprise from about 23 mM to about 29 mM of sodium chloride.
  • the buffer may comprise from about 23 mM to about 27 mM of sodium chloride. In some aspects, the buffer may comprise from about 24 mM to about 28 mM of sodium chloride. In some aspects, the buffer may comprise from about 24 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 25 mM to about 27 mM of sodium chloride. In some aspects, the buffer may comprise from about 25 mM to about 28 mM of sodium chloride. In some aspects, the buffer may comprise from about 25 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.5 mM to about 27.5 mM of sodium chloride.
  • the buffer may comprise from about 25.3 mM to about 27.3 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.4 mM to about 27.4 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.35 mM to about 27.35 mM of sodium chloride. In some aspects, the buffer may comprise from about 26 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 26 mM to about 28 mM of sodium chloride. In some aspects, the buffer may comprise from about 26 mM to about 27 mM of sodium chloride. In some aspects, the buffer may comprise from about 26.3 mM to about 27.3 mM of sodium chloride.
  • the buffer may comprise from about 26.4 mM to about 27.4 mM of sodium chloride. In some aspects, the buffer may comprise from about 26.3 mM to about 26.4 mM of sodium chloride. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 26 mM of sodium chloride. In some aspects, the buffer comprises about 27 mM of sodium chloride. In some aspects, the buffer comprises about 26.3 mM of sodium chloride. In some aspects, the buffer comprises about 26.4 mM of sodium chloride. In some aspects, the buffer comprises about 26.35 mM of sodium chloride.
  • the antibody formulation preferably comprises a non-ionic surfactant. More preferably, the non-ionic surfactant comprises polysorbate 80.
  • the antibody formulation, including the antibody and the aqueous buffer preferably comprises from about 0.01% to about 1% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.03% to about 0.7% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.05% to about 0.4% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.075% to about 0.3% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.25% (by volume) of polysorbate 80.
  • the antibody formulation comprises from about 0.07% to about 0.2% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.15% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.14% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.3% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.2% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.15% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.12% (by volume) of polysorbate 80.
  • the antibody formulation comprises from about 0.08% to about 0.1% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.15% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.2% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.18% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.11% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.1% (by volume) of polysorbate 80. These ranges include the lower and upper amounts that define the range. In some aspects, the antibody formulation comprises about 0.1% (by volume) of polysorbate 80.
  • the antibody formulation preferably is buffered to an acidic pH.
  • the formulation preferably has a pH of about 4.8 to about 5.6. In some aspects, the formulation has a pH of about 4.9 to about 5.5. In some aspects, the formulation has a pH of about 5.0 to about 5.4. In some preferred aspects, the formulation has a pH of about 5.0 to about 5.3. In some preferred aspects, the formulation has a pH of about 5.0 to about 5.2. In some aspects, the formulation has a pH of about 5.1 to about 5.3. In some aspects, the formulation has a pH of about 5.1 to about 5.5. In some preferred aspects, the formulation has a pH of about 5.1 to about 5.2.
  • the formulation has a pH of about 5.1 to about 5.4 In some aspects, the formulation has a pH of about 5.2 to about 5.4. In some aspects, the formulation has a pH of about 5.2 to about 5.5. In some preferred aspects, the formulation has a pH of about 5.2 to about 5.3. These ranges include the lower and upper amounts that define the range. In some aspects, the formulation has a pH of about 5.2.
  • the antibody formulation comprises about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.7 mM to about 1.3 mM of sodium acetate trihydrate, about 200 mM to about 206 mM of mannitol, about 16 mM to about 22 mM of glacial acetic acid, and about 24 mM to about 28 mM of sodium chloride, and about 0.07% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody formulation consists essentially of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting essentially of about 0.7 mM to about 1.3 mM of sodium acetate trihydrate, about 200 mM to about 206 mM of mannitol, about 16 mM to about 22 mM of glacial acetic acid, and about 24 mM to about 28 mM of sodium chloride, and about 0.07% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody formulation consists of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 0.7 mM to about 1.3 mM of sodium acetate trihydrate, about 200 mM to about 206 mM of mannitol, about 16 mM to about 22 mM of glacial acetic acid, and about 24 mM to about 28 mM of sodium chloride, and about 0.07% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody may be present in the formulation at about 37 mg to about 43 mg, or about 38 mg to about 42 mg, or about 39 mg to about 41 mg, or about 40 mg.
  • the antibody formulation comprises about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody formulation consists essentially of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting essentially of about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody formulation consists of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody may be present in the formulation at about 37 mg to about 43 mg, or about 38 mg to about 42 mg, or about 39 mg to about 41 mg, or about 40 mg.
  • the acetate salt may comprise any suitable acetate salt.
  • Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
  • the antibody formulation comprises about 39 mg to about 41 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about
  • the antibody formulation consists essentially of about 39 mg to about 41 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ.
  • a buffer consisting essentially of about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about 18 mM to about 20 mM of glacial acetic acid, and about 25.35 mM to about 26.35 mM of sodium chloride, and about 0.09% to about 0.11% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody formulation consists of about 39 mg to about 41 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about 18 mM to about 20 mM of glacial acetic acid, and about 25.35 mM to about 26.35 mM of sodium chloride, and about 0.09% to about 0.11% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3.
  • the antibody may be present in the formulation at about 37 mg to about 43 mg, or about 38 mg to about 42 mg, or about 39 mg to about 41 mg, or about 40 mg.
  • the acetate salt may comprise any suitable acetate salt.
  • Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
  • the antibody formulation comprises about 40 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 1 mM of an acetate salt, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, and has a pH of about 5.2.
  • the antibody formulation consists essentially of about 40 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting essentially of about 1 mM of an acetate salt, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, and has a pH of about 5.2.
  • the antibody formulation consists of about 40 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 1 mM of an acetate salt, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, and has a pH of about 5.2.
  • the acetate salt may comprise any suitable acetate salt.
  • Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
  • the formulation stabilizes the antibody for improved shelf storage, particularly over a period of months to years.
  • the antibody maintains thermal and colloidal stability during the period of storage.
  • the antibody when stored in the formulation, the antibody is stable and exhibits minimal aggregation, flocculation, fragmentation, and denaturation, and the antibody retains it tumor necrosis factor alpha binding activity.
  • the antibody formulation be stored under refrigerated conditions, and preferably at a temperature of from about 2° C to about 8° C, including from about 2° C to about 6° C, and including about 2° C, about 3° C, about 4° C, about 5° C, about 6° C, about 7° C, or about 8° C.
  • the antibody formulation may be stored at such temperatures for at least about 3 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 6 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 9 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 12 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 15 months.
  • the antibody formulation may be stored at such temperatures for at least about 18 months. During the storage period the antibody is stable and exhibits minimal aggregation, flocculation, fragmentation, and denaturation, and the antibody retains it tumor necrosis factor alpha binding activity such that the antibody formulation may be removed from storage, administered to a patient, and still exhibit therapeutic efficacy against the condition for which the formulation is administered.
  • the formulation comprises about 10 mg to about 70 mg of antibody.
  • this amount of antibody protein is a percentage of antibody monomers in active, native form, as well as a percentage of antibody fragments, antibody aggregates, and denatured or partially denatured antibodies that have reduced or no tumor necrosis binding activity. It is highly preferred that the formulation include a maximal amount of functional antibody monomers and a minimal amount of antibody fragments, aggregates, and structurally altered forms of the antibody with reduced binding activity and/or therapeutic efficacy (relative to the unaltered monomer).
  • the antibody formulation preferably contains at least about 85% by weight of antibody monomers, and less than about 15% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months.
  • the antibody formulation contains at least about 90% by weight of antibody monomers, and less than about 10% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 93% by weight of antibody monomers, and less than about 7% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months.
  • the antibody formulation contains at least about 95% by weight of antibody monomers, and less than about 5% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 96% by weight of antibody monomers, and less than about 4% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months.
  • the antibody formulation contains at least about 97% by weight of antibody monomers, and less than about 3% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 98% by weight of antibody monomers, and less than about 2% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months.
  • the antibody formulation contains at least about 99% by weight of antibody monomers, and less than about 1% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months.
  • the amount of antibody monomers and antibody fragments, aggregates, and structurally altered forms may be determined according to any technique suitable in the art, including those described or exemplified herein, including any one or combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), size exclusion chromatography (SE-UPLC), non-reducing and reducing capillary electrophoresis SDS (NR CE- SDS and R CE-SDS), peptide mapping and particle counting (PC).
  • DLS dynamic light scattering
  • DSC differential scanning calorimetry
  • SE-UPLC size exclusion chromatography
  • NR CE- SDS and R CE-SDS non-reducing and reducing capillary electrophoresis SDS
  • PC peptide mapping and particle counting
  • the antibody formulation contains at least about 90% by weight of antibody monomers, and less than about 10% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 93% by weight of antibody monomers, and less than about 7% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months.
  • the antibody formulation contains at least about 95% by weight of antibody monomers, and less than about 5% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 96% by weight of antibody monomers, and less than about 4% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months.
  • the antibody formulation contains at least about 97% by weight of antibody monomers, and less than about 3% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 98% by weight of antibody monomers, and less than about 2% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months.
  • the antibody formulation contains at least about 99% by weight of antibody monomers, and less than about 1% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months.
  • the amount of antibody monomers and antibody fragments, aggregates, and structurally altered forms may be determined according to any technique suitable in the art, including those described or exemplified herein, including any one or combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), size exclusion chromatography (SE-UPLC), non-reducing and reducing capillary electrophoresis SDS (NR CE-SDS and R CE-SDS), peptide mapping and particle counting (PC).
  • DLS dynamic light scattering
  • DSC differential scanning calorimetry
  • SE-UPLC size exclusion chromatography
  • NR CE-SDS and R CE-SDS non-reducing and reducing capillary electrophoresis SDS
  • PC peptide mapping and particle counting
  • the antibody formulation contains at least about 90% by weight of antibody monomers, and less than about 10% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 93% by weight of antibody monomers, and less than about 7% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months.
  • the antibody formulation contains at least about 95% by weight of antibody monomers, and less than about 5% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 96% by weight of antibody monomers, and less than about 4% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months.
  • the antibody formulation contains at least about 97% by weight of antibody monomers, and less than about 3% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 98% by weight of antibody monomers, and less than about 2% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months.
  • the antibody formulation contains at least about 99% by weight of antibody monomers, and less than about 1% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months.
  • the amount of antibody monomers and antibody fragments, aggregates, and structurally altered forms may be determined according to any technique suitable in the art, including those described or exemplified herein, including any one or combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), size exclusion chromatography (SE-UPLC), non-reducing and reducing capillary electrophoresis SDS (NR CE-SDS and R CE-SDS), peptide mapping and particle counting (PC).
  • DLS dynamic light scattering
  • DSC differential scanning calorimetry
  • SE-UPLC size exclusion chromatography
  • NR CE-SDS and R CE-SDS non-reducing and reducing capillary electrophoresis SDS
  • PC peptide mapping and particle counting
  • the invention also features methods for treating Rheumatoid Arthritis in a subject in need thereof by administering a therapeutically effective amount of any of the antibody formulations described or exemplified herein.
  • the invention also features methods for treating Juvenile Idiopathic Arthritis, Psoriatic Arthritis, Ankylosing Spondylitis, Crohn's Disease, Plaque Psoriasis, Ulcerative Colitis, Inflammatory Bowel Disease, Hidradenitis Suppurativa, or Refractory Asthma by administering a therapeutically effective amount of any of the antibody formulations described or exemplified herein.
  • Therapeutic efficacy is attained, for example, by the ONS-3010 antibody present in the administered formulation.
  • Administration of the antibody formulation may be according to any suitable route, preferably by injection, and more preferably by subcutaneous injection. Administration may be carried out under the direction or supervision of a medical practitioner.
  • the antibody formulations described and exemplified herein may be for use as a medicament.
  • the antibody formulations described and exemplified herein may be for use in the manufacture of a medicament.
  • the formulations may be for use in the treatment of Rheumatoid Arthritis.
  • the formulations may be for use in the treatment of Juvenile
  • the formulations may be for use in the treatment of Psoriatic Arthritis.
  • the formulations may be for use in the treatment of Ankylosing Spondylitis.
  • formulations may be for use in the treatment of Crohn's Disease.
  • the formulations may be for use in the treatment of Plaque Psoriasis.
  • the formulations may be for use in the treatment of Ulcerative Colitis.
  • the formulations may be for use in the treatment of Inflammatory Bowel Disease.
  • the formulations may be for use in the treatment of
  • Hidradenitis Suppurativa The formulations may be for use in the treatment of Refractory Asthma.
  • kits may be used, for example, to practice any of the methods described or exemplified herein.
  • a kit comprises any antibody formulation described or exemplified herein, and instructions for using the antibody formulation in any of the methods or uses described or exemplified herein.
  • the kit may comprise a device for injecting the antibody formulation into a subject, including but not limited to a syringe and needle, or catheter.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Rheumatoid Arthritis, including instructions for injecting the antibody formulation into a Rheumatoid Arthritis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Juvenile Idiopathic Arthritis, including instructions for injecting the antibody formulation into a Juvenile Idiopathic Arthritis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Psoriatic Arthritis, including instructions for injecting the antibody formulation into a Psoriatic Arthritis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Ankylosing Spondylitis, including instructions for injecting the antibody formulation into a Ankylosing Spondylitis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Crohn's Disease, including instructions for injecting the antibody formulation into a Crohn's Disease patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Ankylosing Spondylitis, including instructions for injecting the antibody formulation into a Ankylosing Spondylitis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Crohn's Disease, including instructions for injecting the antibody formulation into a Crohn's Disease patient in need thereof.
  • instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Plaque Psoriasis, including instructions for injecting the antibody formulation into a Plaque Psoriasis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Ulcerative Colitis, including instructions for injecting the antibody formulation into a Ulcerative Colitis patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Inflammatory Bowel Disease, including instructions for injecting the antibody formulation into an Inflammatory Bowel Disease patient in need thereof.
  • the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Hidradenitis Suppurativa, including instructions for injecting the antibody formulation into a Hidradenitis Suppurativa patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Refractory Asthma, including instructions for injecting the antibody formulation into a Refractory Asthma patient in need thereof.
  • Antibody ONS-3010 represents a biosimilar of adalimumab, and has been reformulated for enhanced storage stability. It is believed that the modifications to the buffer of the formulation composition may reduce the incidence of injection-site reaction, including injection pain and a burning sensation observed from subcutaneous administration of adalimumab (Kaiser C et al. (2012) Rheumatol. Int. 32:295-9, and
  • adalimumab formulations include (in addition to the antibody), sodium chloride, monobasic sodium phosphate dihydrate, dibasic sodium phosphate dihydrate, sodium citrate, citric acid monohydrate, mannitol, polysorbate 80, and sterile water for injection.
  • the first experimental series of studies focused on buffer composition, strength, and ability to achieve the desired pH of about 5.2.
  • the second experimental series of experiments utilized stressed stability studies with refined set of formulation conditions based on results of experimental series 1. Sodium chloride concentration was probed in experimental series 2.
  • the third experimental series of formulation development studies compared three conditions, including a control of the adalimumab reference product buffer (per 0.8 ml: 40 mg adalimumab, 4.93 mg sodium chloride, 0.69 mg monobasic sodium phosphate dihydrate, 1.22 mg dibasic sodium phosphate dihydrate, 0.24 mg sodium citrate, 1.04 mg citric acid monohydrate, 9.6 mg mannitol, 0.8 mg polysorbate 80, and Q..S.
  • adalimumab reference formulation level of NaCI and mannitol there was one condition for the adalimumab reference formulation level of NaCI and mannitol, and a condition where those levels were modified relative to the adalimumab reference formulation (lower NaCI, higher mannitol (LS/HM)). These modifications resulted in formulations of comparable osmolality to the adalimumab reference formulation, while maintaining isotonicity.
  • DLS testing method used a Wyatt DynaProTM Plate Reader to provide information on protein size distribution and overall colloidal stability in solution. Hydrodynamic radius provided information on the presence of aggregation and confirmation of the molecule's structure in solution. DLS testing provided an orthogonal measure of size distribution in solution under non-denaturing conditions.
  • Differential Scanning Calorimetry measured the melting transitions for the protein and, thus, provided information on protein thermal stability in solution. Calorimetry was performed using a GE VP Capillary DSC system. The protein was heated from 25° C to 95° C at an optimized scan rate allowing the melting transitions (Tm) to occur while the protein is unfolding. A buffer control was heated alongside the sample and used to calculate melting temperatures and transitions. The DSC profile was typical of antibodies and demonstrated that the protein folded into distinct domains.
  • SE-UPLC Size Exclusion Chromatography
  • NonReducing and Reducing Capillary Electrophoresis SDS (NR CE-SDS and R CE-SDS).
  • CE-SDS analysis was used to compare ONS-3010 size variants under denaturing conditions, with both non-reducing and reducing conditions, using a Beckman PA800 plus instrument.
  • Capillary gel electrophoresis provides automated analysis of reduced and non-reduced proteins by size to determine protein purity and/or heterogeneity. Samples were treated with either an alkylation or reducing agent and SDS was bound to all proteins via a sample buffer. A polymer matrix was filled into the capillary prior to sample analysis. Samples were electrokinetically introduced to the capillary by an applied voltage, then electrophoresis was performed by applying a constant voltage to the capillary.
  • the SDS treated proteins have mass to charge properties that are proportional to the protein weights, which allows for the separation of the SDS-bound proteins by the differences in molecular weight. Test article proteins were quantified by UV detection at 220 nm.
  • L929 Cell-Based Bioassay The primary mechanism of action of adalimumab is the neutralization of circulating TNF-alpha.
  • L929 cell-based bioassay measures cell death/viability. TNF-alpha induces cytotoxicity in L929 cells; relative potency of adalimumab was measured by monitoring live cells through a luminescent tag.
  • N-terminal sequence variants N-terminal sequence variants, C-terminal sequence variants, oxidation, deamidation, succinimide formation, isomerization are measured using peptide mapping LC-MS methodologies.
  • Experimental series 1 The first experimental series of studies focused on buffer composition, strength, and ability to achieve the desired pH of 5.2. Buffers tested included citrate and phosphate (which are used in the reference product formulation) and acetate (Table 1). Sodium chloride and mannitol concentrations (equivalent to those in adalimumab reference formulation) were added to conditions throughout experimental series 1 experiments. From this experimental series of experiments, it was observed that some buffers were better than others at achieving and maintaining the desired pH in the range of 4.9 - 5.5 (0.3 pH units outside of the adalimumab reference formulation). SE-UPLC purity, in particular, was highly correlated with pH, and the use of acetate buffer resulted in preferable profiles ( Figures 1 and 2).
  • Y NaCI and mannitol included at Adalimumab reference buffer concentrations of 4.93 mg/0.08 mL and 9.6 mg/0.8 mL, respectively
  • Dynamic light scattering was used to monitor the hydrodynamic radius Rh (size) of protein molecules in solution. Hydrodynamic radius size in the 5 - 6 nanometer range under lower ( ⁇ Img/mL) protein concentration are typical for monomeric monoclonal antibodies (about 140 kDa in size); this size increases with protein concentration, possibly due to crowding, self-association, or aggregation. Such higher sizes should typically be avoided under formulation conditions since they are indicative of an inherently unstable condition. Hydrodynamic radii of ONS-3010 in experimental series 1 formulation conditions were monitored at two protein concentrations for more complete picture of colloidal stability. Rh was not dominated by pH ( Figure 4): there was considerable variation in Rh even within a relatively narrow pH range, underscoring the impact of buffer composition on colloidal stability. The conditions that had Rh ⁇ adalimumab reference formulation of 8.0 nm were selected for further evaluation in experimental series 2.
  • Experimental series 2 utilized stressed stability studies with a refined set of formulation conditions based on results of experimental series 1.
  • Acetate buffer is of particular interest at the 20 mM level.
  • Y NaCI and mannitol included at Adalimumab reference (AR) buffer concentrations (4.93 mg / 0.8 mL and 9.6 mg / 0.8 mL, respectively).
  • N no NaCI added
  • Table 3 summarizes the experimental series 2 conditions and their analytical results, highlighting reasons for their inclusion or exclusion from experimental series 3 investigations.
  • conditions selected for experimental series 3 showed comparable or improved stability toward thermal and chemical denaturation as monitored by a variety of orthogonal techniques (SE-UPLC, CEX-HPLC, CE-SDS).
  • Relative potency was also assessed using the L929 cell-based potency assay, and colloidal stability was monitored with DLS. Finally, all samples were visually monitored throughout the study (and haziness upon dilution for testing became an exclusion criterion).
  • Experimental series 3 The final experimental series of formulation development studies compares three conditions including the adalimumab reference formulation as a control (Table 4). The other two reformulation conditions use acetate buffer. There is one acetate buffer matching the adalimumab reference level of NaCI and mannitol, and an acetate buffer where those levels are modified relative to the adalimumab reference (AR) formulation (lower NaCI, higher mannitol(LS/HM)). These modifications result in
  • Y NaCI and mannitol included at Adalimumab reference (AR) buffer concentrations (4.93 mg / 0.8 mL and 9.6 mg / 0.8 mL, respectively).
  • LS/HM lower NaCI and higher mannitol levels.
  • CEX-HPLC peptide comparable to Adalimumab mapping
  • L929 reference for CEX-HPLC SE- bioassay
  • UPLC UPLC
  • CE-SDS peptide map appearance and bioassay.
  • Particle count T 0 comparable or better than Adalimumab reference.
  • Dynamic Light Scattering was used to measure the hydrodynamic radius (Rh) of ONS-3010 at four different protein concentrations (see Figure 9 for graphical representation of the results).
  • the adalimumab reference formulation at 50 mg/ml, has an average hydrodynamic radius of 8.0 nm.
  • Condition 2 acetate shows similar values at the full 50 mg/ml concentration
  • condition 3 acetate LS/HM with lower salt shows a smaller Rh value, correlated with better control of self-association at higher protein concentrations.
  • Lower amounts of sodium chloride as used in condition 3 are enough to disrupt the crowding/association taking place at 50 mg/mL used under typical formulation conditions.
  • Rh values converge on values in the 5 - 6 nm range, typical for monomeric monoclonal antibodies.
  • Condition 3 (Acetate LS/H M) results in the lowest Rh values over the entire range of protein concentrations measured.
  • Osmolarity Osmolality values for the three conditions were measured using the Nova Flex instrument. All conditions were similar to one another and to the adalimumab reference formulation, and were in the isotonic range of 290 - 340 mOsm/kg (Table 7).
  • LS/HM Lower NaCI and higher mannitol levels
  • Particle Count Particle analysis was conducted using a HIAC Model 9703+ system following a modified USP method (allowing for detection of particles as small as 2 ⁇ ). Cumulative results for all size ranges are shown in Table 8, with counts per container calculated based on the 0.8 ml pre-filled syringe presentation. The 10 and 25 ⁇ size bins are specifically tracked per the USP method ⁇ 788>. Values for all conditions are well below the limits of ⁇ 600 cumulative counts per container for > 25 micron particles, and ⁇ 6000 cumulative counts per container for > 10 micron particles. Lower salt (formulation condition 3) appears to further reduce particles at 2-10 micron relative to the adalimumab reference formulation and the higher salt formulation (Table 8).
  • LS/HM Lower NaCI and higher mannitol levels
  • SE-UPLC Exposure of ONS-3010 to 55° C generated both higher and lower molecular weight species (HMWS and LMWS) (Table 9), both of which can be monitored by SE-UPLC. From time zero to day 7, there were marked increases in dimer (with a retention time of approximately 3.1 minutes) and species larger-than-dimer with earlier retention times, both counted as HMWS. For fragmentation, a distinct peak was formed off the backside of the monomer peak at ⁇ 3.9 minutes, plus an additional peak at 4.5 minutes ( Figure 10). After 7 days of incubation, there were clear differences between the SE-UPLC chromatograms of the formulation conditions ( Figure 11).
  • LS/HM Lower NaCI and higher mannitol levels
  • CEX-HPLC Cation exchange chromatography of samples treated at 55° C provides a broad view of many physicochemical changes that can manifest themselves as cha nges in molecular charge. This includes specific charge based modifications such as deamidation, isomerization, and pyroglutamine formation, but can also reveal more subtle
  • CEX-HPLC profiles were monitored for time zero through day 2 sam ples (Table 10).
  • CE-SDS R/NR To provide an orthogonal view of size variants (denatured vs. the non- denatured SE-UPLC methodology), CE-SDS was used under both non-reducing and reducing conditions. The strong differentiation between formulation conditions observed with SE- UPLC is not seen under analysis by denaturing techniques, indicating that the size variants formed are largely noncovalent in nature.
  • Bioassay In general, all samples tested showed comparable relative potency in the bioassay within method variability. There was no measurable change in potency after the 7 day treatment at 55 degrees C, demonstrating stability toward thermal denaturation.
  • Peptide Map allowed a more specific view of modifications in ONS-3010.
  • the 55° C incubation temperature fostered certain chemical modifications that are not commonly observed at lower temperatures, such as pyroglutamic acid formation of the N-terminal heavy chain and specific isomerization events.
  • Condition 3 (Acetate LS/HM) appeared to have the most protection against this assortment of chemical modifications ( Figure 17, Figure 18, Figure 19, Figure 20, and Table 11).
  • C-terminal variants and glycosylation levels remained constant throughout the treatment. Table 11.
  • CEX-HPLC CEX-HPLC analysis of accelerated stability samples at 14 day, 21 day and 28 days of treatment at 37° C (Table 13) showed acetate conditions comparable to the adalimumab reference formulation.
  • LS/HM Lower NaCI and higher mannitol levels CE-SDS (R/NR). For the 37° C incubated samples, CE-SDS testing reveals similar trends in NR denatured size variants between the three conditions after 28 days. For 37° C R CE-SDS, all conditions relative to the adalimumab reference formulation are comparable after 28 days.
  • IP particle visible
  • 5P many particles visible
  • LS/HM Lower NaCI and higher mannitol levels
  • a forced oxidation study utilized 1% t-butyl hydrogen peroxide treatment to induce oxidation in ONS-3010 formulation candidates.
  • SE-UPLC, CEX-HPLC, and tryptic peptide map methodologies were used to monitor changes in product quality attributes and specific amino acid residues that are susceptible to oxidation. Oxidative modification is one of the major chemical degradation pathways. Sites of oxidation damage on backbone or side-chain can change hydrophobicity of protein surfaces. The fingerprint of oxidation by using LC-MS peptide mapping enables a fast and reliable approach for formulation selection.
  • Formulation condition #3 provided the most protection overall to oxidation. Oxidized species of residues M4 and M83 were below the method LOO even in stressed conditions. Upon stress, oxidized M34 was at the LOO, with the three conditions comparable within method variability.
  • Adalimumab Reference T 0 2-8° C 15.3 65.2 18.6 0.8
  • LS/HM Lower NaCI and higher mannitol levels
  • Freeze-Thaw Cycling Freeze-thaw cycling was conducted for sam ples in the candidate formulations at two temperatures: -20° C and -80° C. Samples were placed in freezers set to the appropriate temperature and allowed to freeze thoroughly (for at least one hour). Samples were then removed from the freezer and allowed to thaw at 25° C (approximately 1 hour). This freezing step plus the thawing step constituted a single cycle. Samples were subjected to up to 5 freeze-thaw cycles, and then a nalyzed together by SE- UPLC, with a subset of samples also tested by NR CE-SDS.
  • LS/HM Lower NaCI and higher mannitol levels
  • CEX-HPLC results showed no significant difference in the acetate formulations compared to the adalimumab reference formulation (Tables 25-27).
  • Day 28 samples displayed full potency in the L929 bioassay, and CE-SDS (R/NR) values are similar across conditions. At day 28, all conditions showed some visible particulate formation: condition 3 showed more protection than the other conditions (Tables 28 and 29).
  • IP particle visible
  • 5P many particles visible
  • IP particle visible
  • 5P many particles visible
  • LS/HM Lower NaCI and higher mannitol levels
  • IP particle visi ble
  • 5P mar
  • LS/HM Lower NaCI and higher mannitol levels
  • LS/HM Lower NaCI and higher mannitol levels
  • LS/HM Lower NaCI and higher mannitol levels
  • LS/HM Lower NaCI and higher mannitol levels
  • LS/HM Lower NaCI and higher mannitol levels
  • SE-UPLC monitors ONS-3010 size homogeneity under non- denaturing conditions.
  • the SE-UPLC testing method separates proteins based on size. The method is isocratic with a sodium phosphate running buffer, using a Waters Acquity UPLC BEH200 SEC column (1.7 ⁇ , 4.6x150 mm). Peaks are monitored using absorbance at 280 nm. Species eluting before the monomer peak are aggregates (HMWS) and peaks eluting after the monomer peak are degradants (LMWS).
  • HMWS aggregates
  • LMWS degradants
  • CE-SDS analysis is used to monitor ONS-3010 size homogeneity under denaturing conditions, with non-reducing conditions, using a Beckman PA800 plus instrument.
  • Samples are treated with an alkylation agent and SDS is bound to all proteins via a sample buffer.
  • a polymer matrix is filled into the capillary prior to sample analysis.
  • Samples are electrokinetically introduced to the capillary by an applied voltage, then electrophoresis is performed by applying a constant voltage to the capillary.
  • the SDS treated proteins have a mass to charge properties that are proportional to the protein weights which allows for the separation of the SDS-bound proteins by the differences in molecular weight.
  • Test article proteins are quantified by UV detection at 220 nm.
  • Peptide Map UPLC Peptide mapping is used to characterize a protein's primary structure. ONS-3010 is digested with trypsin and the resulting peptides are separated by RP-UPLC. The characteristic peptide map fingerprint is analyzed for peak relative retention time and overall peak pattern compared with the reference.
  • Adalimumab reference formulation displaying enhanced stability at 55°C for aggregation and % intact protein on non-reduced CE as well as peptide map. There was also a visual difference due to the antibody in the Adalimumab reference buffer becoming opalescent by day 10 of treatment compared to the antibody in the acetate LS/HM buffer formulation, which remained clear throughout 10 days of treatment.
  • Tables 48-50 show the longest timepoints with significant differences. There consistent trend throughout the treatment, however.
  • adalimumab reference citrate-phosphate buffer
  • acetate LS/HM buffer offers greater stability against aggregation at pH 3.0.
  • the acetate LS/HM buffer formulation shows greater protection at low pH compared to the adalimumab reference formulation.
  • Formulation condition #3 (acetate LS/HM) used in experimental series 3, provides a protective effect to ONS-3010 relative to the adalimumab reference buffer. This observation is consistent in forced oxidation and shaking studies, as well as at elevated temperatures.
  • the acetate LS/HM buffer formulation provides improved thermal, conformational and colloidal stability to ONS-3010, which indeed translates into comparable shelf life and/or improved product quality.
  • Condition 1 (adalimumab reference) and Condition 3 (acetate LS/HM buffer formulation) were stable and showed no significant change.

Abstract

The invention provides buffered formulations of adalimumab. The formulations comprise a buffer comprising an acetate salt, mannitol, glacial acetic acid, sodium chloride, and polysorbate 80. The formulations have an acidic pH, and enhance the thermal, conformational and colloidal stability of antibodies, including the adalimumab antibody.

Description

BUFFER FORMULATIONS FOR ENHANCED ANTIBODY STABILITY
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application No. 61/891,485 filed on October 16, 2013, the contents of which are incorporated by reference herein, in their entirety and for all purposes.
REFERENCE TO A SEQUENCE LISTING
This application includes a Sequence Listing submitted electronically as a text file named Buffered Adalimumab ST25.txt, created on October 10, 2014, with a size of 16,000 bytes. The Sequence Listing is incorporated by reference herein.
FIELD OF THE INVENTION
The invention relates generally to the field of antibody formulation chemistry. More particularly, the invention relates to buffered formulations for antibody storage, which enhance the thermal stability, conformational and colloidal sta bility of the antibody, thereby enhancing long term storage of the antibody.
BACKGROUND OF THE INVENTION
Various publications, including patents, published applications, accession numbers, technical articles and scholarly articles are cited throughout the specification. Each of these cited publications is incorporated by reference herein, in its entirety and for all purposes.
As part of the Biologies Price Competition and I nnovation Act (BPCIA), a biological drug product (produced in or derived from living organisms) may be demonstrated to be "biosimilar" if data show that, among other things, the product is "highly similar" to an already-approved biological product. The biosimilar product should retain at least the biologic function a nd treatment efficacy of the U.S. Food and Drug Agency-approved biological product. The biosimilar product may be formulated differently, however, from the a pproved biological product. The formulation may improve stability a nd shelf storage of the biologic drug product, and may also improve the efficacy in treating a particular disease or condition. The formulation may also improve other aspects of administration, including a reduction in patient discomfort or other untoward effects that a patient may experience upon administration of the approved biological product. Antibody molecules may be used as biological drugs, and many such antibodies are approved for use in human beings. Antibody molecules may be produced as a biosimilar, and reformulated accordingly. There remains a need in the art for high-quality antibody biosimilars.
SUMMARY OF THE INVENTION
The invention features buffered antibody formulations, comprising (a) an antibody. The antibody may specifically bind to tumor necrosis factor alpha. The antibody may comprise a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2. The formulation, in addition to the antibody, comprises (b) an aqueous buffer comprising from about 0.7 mM to about 1.3 mM of an acetate salt, preferably sodium acetate trihydrate, from about 200 mM to about 206 mM of mannitol, from about 16 mM to about 22 mM of glacial acetic acid, and from about 24 mM to about 28 mM of sodium chloride, and (c) about 0.07% (v/v) to about 0.15% (v/v) of a non-ionic surfactant such as polysorbate 80. The buffered antibody formulation has a pH of from about 5.1 to about 5.3, preferably about 5.2.
In some aspects, the formulation comprises from about 30 mg to about 50 mg of the antibody. In some preferred aspects, the formulation comprises from about 35 mg to about 45 mg of the antibody. In some preferred aspects, the formulation comprises from about 37 mg to about 43 mg of the antibody. In some preferred aspects, the formulation comprises about 40 mg of the antibody.
The buffer may comprise from about 0.8 mM to about 1.2 mM of sodium acetate trihydrate, or from about 0.9 mM to about 1.1 mM of sodium acetate trihydrate, or about 1 mM of sodium acetate trihydrate. The buffer may comprise from about 201 mM to about 205 mM of mannitol, or from about 202 mM to about 204 mM of mannitol, or about 203 mM of mannitol. The buffer may comprise from about 17 mM to about 21 mM of glacial acetic acid, or from about 18 mM to about 20 mM of glacial acetic acid, or about 19 mM of glacial acetic acid. The buffer may comprise from about 25 mM to about 27 mM of sodium chloride, or about 26 mM of sodium chloride, or about 27 mM of sodium chloride, or about 26.35 mM of sodium chloride.
The buffered antibody formulation includes a non-ionic surfactant, which preferably is polysorbate 80. In some aspects, the formulation comprises from about 0.08% (v/v) to about 0.12% (v/v) of polysorbate 80. In some aspects, the formulation comprises from about 0.09% (v/v) to about 0.11% (v/v) of polysorbate 80. In some aspects, the formulation comprises about 0.1% (v/v) of polysorbate 80.
In a detailed aspect, a buffered antibody formulation comprises (a) about 30 mg to about 50 mg of an antibody comprising a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ. ID NO: 2, (b) a buffer comprising about 1 mM of an acetate salt, preferably sodium acetate trihydrate, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and (c) about 0.1% (by volume) of polysorbate 80. The buffered antibody formulation has a pH of from about 5.1 to about 5.3, preferably about 5.2. In some preferred aspects, the formulation comprises from about 35 mg to about 45 mg of the antibody. In some preferred aspects, the formulation comprises from about 37 mg to about 43 mg of the antibody. In some preferred aspects, the formulation comprises about 40 mg of the antibody.
The buffered antibody formulations may be used as a medicament, and may be used in methods of treatment. For example, the buffered antibody formulations may be for use in the treatment of arthritis. In some aspects, the buffered antibody formulations may be for use in the treatment of Rheumatoid Arthritis, or Juvenile Idiopathic Arthritis, or Psoriatic Arthritis. In some aspects, the buffered antibody formulations may be for use in the treatment of Ankylosing Spondylitis. In some aspects, the buffered antibody formulations may be for use in the treatment of Crohn's Disease. In some aspects, the buffered antibody formulations may be for use in the treatment of Ulcerative Colitis. In some aspects, the buffered antibody formulations may be for use in the treatment of Plaque Psoriasis.
The methods of treatment include methods for treating arthritis, including
Rheumatoid Arthritis, Juvenile Idiopathic Arthritis, and Psoriatic Arthritis. The methods of treatment also include methods for treating Ankylosing Spondylitis, methods for treating Crohn's Disease, methods for treating Plaque Psoriasis, and methods for treating Ulcerative Colitis.
In some aspects, methods of treatment comprise administering to an arthritis patient, including a Rheumatoid Arthritis, Juvenile Idiopathic Arthritis, or Psoriatic Arthritis, an amount of the buffered antibody formulations described or exemplified herein effective to treat the arthritis in the patient. I n some aspects, methods of treatment comprise administering to an Ankylosing Spondylitis patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Ankylosing Spondylitis in the patient. I n some aspects, methods of treatment comprise administering to a Crohn's Disease patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Crohn's Disease in the patient. I n some aspects, methods of treatment comprise administering to an Ulcerative Colitis patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Ulcerative Colitis in the patient. I n some aspects, methods of treatment comprise administering to a Plaque Psoriasis patient an amount of the buffered antibody formulations described or exemplified herein effective to treat the Plaque Psoriasis in the patient. The buffered antibody formulations are preferably administered subcutaneously to the patient, for example, by subcutaneous injection. The patient preferably is a human being.
The invention also provides kits, which may be used, for example, in accordance with the methods of treatment. Thus, for example, the kits generally comprise any of the buffered antibody formulations described or exemplified herein and instructions for using the formulation in a method of treatment. The method of treatment may be a method for treating arthritis. The method of treatment may be a method for treating Rheumatoid Arthritis. The method of treatment may be a method for treating Juvenile Idiopathic Arthritis. The method of treatment may be a method for treating Psoriatic Arthritis. The method of treatment may be a method for treating Ankylosing Spondylitis. The method of treatment may be a method for treating Crohn's Disease. The method of treatment may be a method for treating Ulcerative Colitis. The method of treatment may be a method for treating Plaque Psoriasis. The kits may include a device for administering the antibody formulation to a patient. The device may comprise a syringe a nd a needle. The device may comprise a catheter.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 shows an overlay of SE-UPLC chromatograms from representative
experimental series 1 formulation conditions.
Fig. 2 shows trends in SE-UPLC % high molecular weight species (HMWS) as a function of solution pH. Fig. 3 shows a DSC thermograms for representative experimental series 1 formulation conditions.
Fig. 4 shows a DLS and pH ranges for formulation solutions at 50 mg/ml protein concentration, coded by buffer composition.
Fig. 5 shows an overlay of SE-UPLC chromatograms for the adalimumab reference formulation under duration of stressed stability experiment (55° C up to 14 days).
Fig. 6 shows an overlay of representative SE-UPLC chromatograms from
experimental series 2 formulation conditions of ONS-3010.
Fig. 7 shows trends in SE-UPLC aggregation over time at stressed conditions.
Fig. 8 shows trends in SE-UPLC fragmentation over time at stressed conditions.
Fig. 9 shows ONS-3010 experimental series 3 DLS results.
Fig. 10 shows an overlay of SE-UPLC chromatograms for the adalimumab reference formulation under duration of stressed stability experiment (55° C up to 7 days).
Fig. 11 shows an overlay of representative SE-UPLC chromatograms from
experimental series 3 ONS-3010 formulation conditions.
Fig. 12 shows trends in experimental series 3 SE-UPLC aggregation over time at stressed conditions.
Fig. 13 shows an overlay of CEX-HPLC chromatograms for 55° C-incubated samples in the adalimumab reference formulation, days 0, 1, and 2.
Fig. 14 shows CEX-HPLC Main Peak Percentages for 55° C-incubated samples up to 2 days.
Fig. 15 shows CEX-HPLC Acidic Peak Percentages for 55° C-incubated samples up to 2 days.
Fig. 16 shows CEX-HPLC Basic Peak Percentages for 55° C-incubated samples up to 2 days.
Fig. 17 shows the total percentage of isomerized species in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition number).
Fig. 18 shows the percentage of cyclized N-terminal peptides in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition number).
Fig. 19 shows the total percentage of oxidized methionine peptides in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition number). Fig. 20 shows the total percentage of deamidated peptides in peptide maps of 55° C stressed ONS-3010 samples (x axis refers to formulation condition num ber).
Fig. 21 shows an overlay of SE-UPLC chromatograms for adalimumab reference formulation samples incubated at 37° C, time zero and day 28.
Fig. 22 shows an overlay of SE-UPLC chromatograms for 37° C-incubated day 28 samples.
Fig. 23 shows the percentage of oxidized methionine-256 peptides in ONS-3010 forced oxidation study peptide maps (x axis refers to formulation condition number). Top = full view, bottom = zoomed.
Fig. 24 shows the percentage of oxidized methionine-432 peptides in ONS-3010 forced oxidation study peptide maps (x axis refers to formulation condition number). Top = full view, bottom = zoomed.
Fig. 25 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated day 28 samples.
Fig. 26 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated 5 month samples.
Fig. 27 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated 12 month samples, conditions 1 and 3.
Fig. 28 shows an overlay of SE-UPLC chromatograms for 2 - 8° C-incubated 18 month samples, conditions 1 and 3.
DETAILED DESCRIPTION OF THE INVENTION
Various terms relating to aspects of the present invention are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art, unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definition provided herein.
As used herein, the singular forms "a," "a n," and "the" include plural referents unless expressly stated otherwise.
As used herein, the terms "comprising," "having," and "including" encompass the more restrictive terms "consisting essentially of" and "consisting of."
The terms subject and patient are used interchangeably, a nd include any a nimal. Subjects include mamma ls, including companion and farm mammals, as well as rodents, including mice, rabbits, and rats, and other rodents. Non-human primates preferred subjects. Human beings are highly preferred subjects.
It has been observed in accordance with the invention that formulations of ONS- 3010, which specifically binds to tumor necrosis factor alpha, can be buffered with mannitol and acetate, while minimizing sodium chloride, with the buffers enhancing the thermal and colloidal stability of the antibody, even more so than formulations of adalimumab currently approved for patient use. It was observed that there is a fine balance in establishing and maintaining an acidic pH of about 5.2 with the appropriate salts and buffer components. It was observed, for example, that high levels of salt may induce aggregation and degradation, which could be improved by lowering the salt level. Accordingly, the disclosure features buffered formulations for antibodies, which formulations include an aqueous carrier comprising buffer comprising acetate and mannitol, as well as a non-ionic surfactant, but with minimal sodium chloride.
In some preferred aspects, the antibody specifically binds to an epitope on tumor necrosis factor alpha, and the epitope may be linear or conformational. In some preferred aspects, the antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1. In some preferred aspects, the antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 2. Preferably, the antibody comprises a heavy chain constant domain and/or a light chain constant domain. In highly preferred aspects, the antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, an example of which is ONS-3010. The heavy and light chain amino acid sequences of the antibody may comprise those of U.S. Pat. No. 6,090,382.
Preferably, the antibody is a full length antibody, comprising both variable and constant regions, although in some aspects, the antibody may comprise a derivative or fragment or portion of a full-length antibody that retains the antigen-binding specificity, and also preferably retains most or all of the affinity, of the full length antibody molecule. The antibody may comprise post-translational modifications (PTMs) or moieties, which may impact antibody activity or stability. The antibody may be methylated, acetylated, glycosylated, sulfated, phosphorylated, carboxylated, and/or amidated, and may comprise other moieties that are well known in the art. Common PTMs for ONS-3010 include N- glycosylation, C-terminal variants (e.g., cleavage of lysine, proline amidation), N-terminal pyro-E formation, oxidation, isomerization, deamidation, succinimide formation,
mannosylation, K98 glycation, and fragmentation. Moieties include any chemical group or combinations of groups commonly found on immunoglobulin molecules in nature, or otherwise added to antibodies by recombinant expression systems, including prokaryotic and eukaryotic expression systems.
The formulation preferably comprises a therapeutically effective amount of an antibody. The antibody may be any antibody compatible with the aqueous buffer formulation. A preferred a ntibody comprises a heavy chain having the amino acid sequence of SEQ. I D NO: 1 and a light chain having the amino acid sequence of SEQ. I D NO: 2. A therapeutically effective amount may vary, depending on the disease or condition being treated upon administration of the antibody, and/or depending on the characteristics of the subject to which the antibody is administered, such as age, gender, height, weight, state of advancement or stage of the disease or condition, the number and efficacy of previous administrations, other therapeutic agents administered to the subject, and other characteristics that are known to the practitioner or that would otherwise be taken into account in determining appropriate dosing. Preferably, a therapeutically effective amount is an amount that is effective to treat Rheumatoid Arthritis. I n some preferred aspects, a therapeutically effective amount is an amount that is effective to treat Juvenile Idiopathic Arthritis, Psoriatic Arthritis, Ankylosing Spondylitis, Crohn's Disease, Plaque Psoriasis, Ulcerative Colitis, I nflammatory Bowel Disease, Hidradenitis Suppurativa, or Refractory Asthma.
The formulation may comprise from about 10 mg to about 70 mg of the antibody. I n some aspects, the formulation comprises from about 20 mg to about 60 mg of the antibody. I n some aspects, the formulation comprises from about 30 mg to about 50 mg of the antibody. I n some aspects, the formulation comprises from about 35 mg to about 45 mg of the antibody. I n some aspects, the formulation comprises from about 37 mg to a bout 43 mg of the antibody. I n some aspects, the formulation comprises from about 38 mg to about 42 mg of the antibody. In some aspects, the formulation comprises from about 39 mg to about 41 mg of the antibody. I n some aspects, the formulation comprises from about 30 mg to about 60 mg of the antibody. I n some aspects, the formulation comprises from about 35 mg to about 55 mg of the antibody. In some aspects, the formulation comprises from about 40 mg to about 60 mg of the antibody. These ranges include the lower and upper amounts that define the range. In some aspects, the formulation comprises about 40 mg of the antibody.
The antibody is preferably formulated with a buffered aqueous carrier, and the carrier preferably comprises water. The buffered antibody formulation is preferably in liquid form, and more preferably in liquid form suitable for subcutaneous administration. Thus, the amount of water in the buffered formulation may vary in accordance with the desired volume of the injectable bolus. The buffer comprises sodium acetate trihydrate, mannitol, sodium chloride, glacial acetic acid, and a non-ionic surfactant, and maintains the antibody formulation at an acidic pH. When stored in the buffered formulation, the antibody is shelf-stable under normal storage conditions.
The buffer may comprise from about 0.1 mM to about 5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.3 mM to about 3 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.5 mM to about 2 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.5 mM to about 1.5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.6 mM to about 1.4 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.7 mM to about 1.5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.7 mM to about 1.3 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.8 mM to about 1.2 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.8 mM to about 1.5 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.8 mM to about 1.1 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.9 mM to about 1.2 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.9 mM to about 1.4 mM of an acetate salt. In some aspects, the buffer may comprise from about 0.9 mM to about 1.1 mM of an acetate salt. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 1 mM of an acetate salt. The acetate salt may comprise any suitable acetate salt. Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
The buffer may comprise from about 100 mM to about 300 mM of mannitol. In some aspects, the buffer may comprise from about 110 mM to about 290 mM of mannitol. In some aspects, the buffer may comprise from about 120 mM to about 280 mM of mannitol. In some aspects, the buffer may comprise from about 150 mM to about 250 mM of mannitol. In some aspects, the buffer may comprise from about 175 mM to about 225 mM of mannitol. In some aspects, the buffer may comprise from about 180 mM to about 220 mM of mannitol. In some aspects, the buffer may comprise from about 185 mM to about 215 mM of mannitol. In some aspects, the buffer may comprise from about 190 mM to about 215 mM of mannitol. In some aspects, the buffer may comprise from about 195 mM to about 210 mM of mannitol. In some aspects, the buffer may comprise from about 197 mM to about 209 mM of mannitol. In some aspects, the buffer may comprise from about 198 mM to about 208 mM of mannitol. In some aspects, the buffer may comprise from about 198 mM to about 205 mM of mannitol. In some aspects, the buffer may comprise from about 199 mM to about 207 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 210 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 207 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 206 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 205 mM of mannitol. In some aspects, the buffer may comprise from about 200 mM to about 203 mM of mannitol. In some aspects, the buffer may comprise from about 201 mM to about 205 mM of mannitol. In some aspects, the buffer may comprise from about 201 mM to about 204 mM of mannitol. In some aspects, the buffer may comprise from about 201 mM to about 203 mM of mannitol. In some aspects, the buffer may comprise from about 202 mM to about 204 mM of mannitol. In some aspects, the buffer may comprise from about 202 mM to about 203 mM of mannitol. In some aspects, the buffer may comprise from about 202 mM to about 206 mM of mannitol. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 203 mM of mannitol.
The buffer may comprise from about 9 mM to about 30 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 10 mM to about 30 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 9 mM to about 29 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 10 mM to about 28 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 11 mM to about 27 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 12 mM to about 26 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 13 mM to about 25 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 14 mM to about 24 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 15 mM to about 23 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 15 mM to about 21 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 15 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 16 mM to about 22 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 16 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 17 mM to about 21 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 17 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 20 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 19 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 18 mM to about 23 mM of glacial acetic acid. In some aspects, the buffer may comprise from about 19 mM to about 20 mM of glacial acetic acid. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 19 mM of glacial acetic acid.
The buffer preferably includes minimal amounts of sodium chloride, and in some aspects, includes no sodium chloride. In some aspects, the buffer may comprise from about 15 mM to about 36 mM of sodium chloride. In some aspects, the buffer may comprise from about 16 mM to about 36 mM of sodium chloride. In some aspects, the buffer may comprise from about 18 mM to about 34 mM of sodium chloride. In some aspects, the buffer may comprise from about 20 mM to about 32 mM of sodium chloride. In some aspects, the buffer may comprise from about 22 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 23 mM to about 29 mM of sodium chloride. In some aspects, the buffer may comprise from about 23 mM to about 27 mM of sodium chloride. In some aspects, the buffer may comprise from about 24 mM to about 28 mM of sodium chloride. In some aspects, the buffer may comprise from about 24 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 25 mM to about 27 mM of sodium chloride. In some aspects, the buffer may comprise from about 25 mM to about 28 mM of sodium chloride. In some aspects, the buffer may comprise from about 25 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.5 mM to about 27.5 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.3 mM to about 27.3 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.4 mM to about 27.4 mM of sodium chloride. In some aspects, the buffer may comprise from about 25.35 mM to about 27.35 mM of sodium chloride. In some aspects, the buffer may comprise from about 26 mM to about 30 mM of sodium chloride. In some aspects, the buffer may comprise from about 26 mM to about 28 mM of sodium chloride. In some aspects, the buffer may comprise from about 26 mM to about 27 mM of sodium chloride. In some aspects, the buffer may comprise from about 26.3 mM to about 27.3 mM of sodium chloride. In some aspects, the buffer may comprise from about 26.4 mM to about 27.4 mM of sodium chloride. In some aspects, the buffer may comprise from about 26.3 mM to about 26.4 mM of sodium chloride. These ranges include the lower and upper amounts that define the range. In some aspects, the buffer comprises about 26 mM of sodium chloride. In some aspects, the buffer comprises about 27 mM of sodium chloride. In some aspects, the buffer comprises about 26.3 mM of sodium chloride. In some aspects, the buffer comprises about 26.4 mM of sodium chloride. In some aspects, the buffer comprises about 26.35 mM of sodium chloride.
The antibody formulation preferably comprises a non-ionic surfactant. More preferably, the non-ionic surfactant comprises polysorbate 80. The antibody formulation, including the antibody and the aqueous buffer, preferably comprises from about 0.01% to about 1% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.03% to about 0.7% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.05% to about 0.4% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.075% to about 0.3% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.25% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.2% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.15% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.07% to about 0.14% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.3% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.2% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.15% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.12% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.08% to about 0.1% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.15% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.2% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.18% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.11% (by volume) of polysorbate 80. In some aspects, the antibody formulation comprises from about 0.09% to about 0.1% (by volume) of polysorbate 80. These ranges include the lower and upper amounts that define the range. In some aspects, the antibody formulation comprises about 0.1% (by volume) of polysorbate 80.
The antibody formulation preferably is buffered to an acidic pH. The formulation preferably has a pH of about 4.8 to about 5.6. In some aspects, the formulation has a pH of about 4.9 to about 5.5. In some aspects, the formulation has a pH of about 5.0 to about 5.4. In some preferred aspects, the formulation has a pH of about 5.0 to about 5.3. In some preferred aspects, the formulation has a pH of about 5.0 to about 5.2. In some aspects, the formulation has a pH of about 5.1 to about 5.3. In some aspects, the formulation has a pH of about 5.1 to about 5.5. In some preferred aspects, the formulation has a pH of about 5.1 to about 5.2. In some preferred aspects, the formulation has a pH of about 5.1 to about 5.4 In some aspects, the formulation has a pH of about 5.2 to about 5.4. In some aspects, the formulation has a pH of about 5.2 to about 5.5. In some preferred aspects, the formulation has a pH of about 5.2 to about 5.3. These ranges include the lower and upper amounts that define the range. In some aspects, the formulation has a pH of about 5.2.
In some preferred aspects, the antibody formulation comprises about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.7 mM to about 1.3 mM of sodium acetate trihydrate, about 200 mM to about 206 mM of mannitol, about 16 mM to about 22 mM of glacial acetic acid, and about 24 mM to about 28 mM of sodium chloride, and about 0.07% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In some aspects, the antibody formulation consists essentially of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting essentially of about 0.7 mM to about 1.3 mM of sodium acetate trihydrate, about 200 mM to about 206 mM of mannitol, about 16 mM to about 22 mM of glacial acetic acid, and about 24 mM to about 28 mM of sodium chloride, and about 0.07% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In some aspects, the antibody formulation consists of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 0.7 mM to about 1.3 mM of sodium acetate trihydrate, about 200 mM to about 206 mM of mannitol, about 16 mM to about 22 mM of glacial acetic acid, and about 24 mM to about 28 mM of sodium chloride, and about 0.07% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In any such embodiments, the antibody may be present in the formulation at about 37 mg to about 43 mg, or about 38 mg to about 42 mg, or about 39 mg to about 41 mg, or about 40 mg.
In some preferred aspects, the antibody formulation comprises about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In some aspects, the antibody formulation consists essentially of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting essentially of about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In some aspects, the antibody formulation consists of about 35 mg to about 45 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In any such
embodiments, the antibody may be present in the formulation at about 37 mg to about 43 mg, or about 38 mg to about 42 mg, or about 39 mg to about 41 mg, or about 40 mg. The acetate salt may comprise any suitable acetate salt. Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
In some preferred aspects, the antibody formulation comprises about 39 mg to about 41 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about
18 mM to about 20 mM of glacial acetic acid, and about 25.35 mM to about 26.35 mM of sodium chloride, and about 0.09% to about 0.11% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In some aspects, the antibody formulation consists essentially of about 39 mg to about 41 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ. ID NO: 2, a buffer consisting essentially of about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about 18 mM to about 20 mM of glacial acetic acid, and about 25.35 mM to about 26.35 mM of sodium chloride, and about 0.09% to about 0.11% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In some aspects, the antibody formulation consists of about 39 mg to about 41 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about 18 mM to about 20 mM of glacial acetic acid, and about 25.35 mM to about 26.35 mM of sodium chloride, and about 0.09% to about 0.11% (by volume) of polysorbate 80, and has a pH of about 5.1 to about 5.3. In any such embodiments, the antibody may be present in the formulation at about 37 mg to about 43 mg, or about 38 mg to about 42 mg, or about 39 mg to about 41 mg, or about 40 mg. The acetate salt may comprise any suitable acetate salt. Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
In some preferred aspects, the antibody formulation comprises about 40 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 1 mM of an acetate salt, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, and has a pH of about 5.2. In some aspects, the antibody formulation consists essentially of about 40 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting essentially of about 1 mM of an acetate salt, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, and has a pH of about 5.2. In some aspects, the antibody formulation consists of about 40 mg of an antibody that specifically binds to tumor necrosis factor alpha and comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer consisting of about 1 mM of an acetate salt, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, and has a pH of about 5.2. The acetate salt may comprise any suitable acetate salt. Non-limiting examples of preferred acetate salts include magnesium acetate salts, potassium acetate salts, calcium acetate salts, zinc acetate salts, and sodium acetate salts. More preferred acetate salts include anhydrous sodium acetate and sodium acetate trihydrate. Sodium acetate trihydrate is highly preferred.
The formulation stabilizes the antibody for improved shelf storage, particularly over a period of months to years. When stored in the formulation, the antibody maintains thermal and colloidal stability during the period of storage. For example, when stored in the formulation, the antibody is stable and exhibits minimal aggregation, flocculation, fragmentation, and denaturation, and the antibody retains it tumor necrosis factor alpha binding activity.
It is preferred that the antibody formulation be stored under refrigerated conditions, and preferably at a temperature of from about 2° C to about 8° C, including from about 2° C to about 6° C, and including about 2° C, about 3° C, about 4° C, about 5° C, about 6° C, about 7° C, or about 8° C. The antibody formulation may be stored at such temperatures for at least about 3 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 6 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 9 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 12 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 15 months. In some aspects, the antibody formulation may be stored at such temperatures for at least about 18 months. During the storage period the antibody is stable and exhibits minimal aggregation, flocculation, fragmentation, and denaturation, and the antibody retains it tumor necrosis factor alpha binding activity such that the antibody formulation may be removed from storage, administered to a patient, and still exhibit therapeutic efficacy against the condition for which the formulation is administered.
The formulation comprises about 10 mg to about 70 mg of antibody. Among this amount of antibody protein is a percentage of antibody monomers in active, native form, as well as a percentage of antibody fragments, antibody aggregates, and denatured or partially denatured antibodies that have reduced or no tumor necrosis binding activity. It is highly preferred that the formulation include a maximal amount of functional antibody monomers and a minimal amount of antibody fragments, aggregates, and structurally altered forms of the antibody with reduced binding activity and/or therapeutic efficacy (relative to the unaltered monomer). For example, the antibody formulation preferably contains at least about 85% by weight of antibody monomers, and less than about 15% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months.
In some aspects, the antibody formulation contains at least about 90% by weight of antibody monomers, and less than about 10% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 93% by weight of antibody monomers, and less than about 7% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 95% by weight of antibody monomers, and less than about 5% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 96% by weight of antibody monomers, and less than about 4% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 97% by weight of antibody monomers, and less than about 3% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 98% by weight of antibody monomers, and less than about 2% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. In some aspects, the antibody formulation contains at least about 99% by weight of antibody monomers, and less than about 1% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about six months. The amount of antibody monomers and antibody fragments, aggregates, and structurally altered forms may be determined according to any technique suitable in the art, including those described or exemplified herein, including any one or combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), size exclusion chromatography (SE-UPLC), non-reducing and reducing capillary electrophoresis SDS (NR CE- SDS and R CE-SDS), peptide mapping and particle counting (PC).
In some aspects, the antibody formulation contains at least about 90% by weight of antibody monomers, and less than about 10% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 93% by weight of antibody monomers, and less than about 7% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 95% by weight of antibody monomers, and less than about 5% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 96% by weight of antibody monomers, and less than about 4% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 97% by weight of antibody monomers, and less than about 3% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 98% by weight of antibody monomers, and less than about 2% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. In some aspects, the antibody formulation contains at least about 99% by weight of antibody monomers, and less than about 1% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about twelve months. The amount of antibody monomers and antibody fragments, aggregates, and structurally altered forms may be determined according to any technique suitable in the art, including those described or exemplified herein, including any one or combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), size exclusion chromatography (SE-UPLC), non-reducing and reducing capillary electrophoresis SDS (NR CE-SDS and R CE-SDS), peptide mapping and particle counting (PC).
In some aspects, the antibody formulation contains at least about 90% by weight of antibody monomers, and less than about 10% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 93% by weight of antibody monomers, and less than about 7% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 95% by weight of antibody monomers, and less than about 5% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 96% by weight of antibody monomers, and less than about 4% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 97% by weight of antibody monomers, and less than about 3% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 98% by weight of antibody monomers, and less than about 2% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. In some aspects, the antibody formulation contains at least about 99% by weight of antibody monomers, and less than about 1% by weight of antibody fragments, aggregates, and structurally altered forms with reduced tumor necrosis factor alpha binding activity and/or therapeutic efficacy when stored at about 2° C to about 8° C for at least about eighteen months. The amount of antibody monomers and antibody fragments, aggregates, and structurally altered forms may be determined according to any technique suitable in the art, including those described or exemplified herein, including any one or combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), size exclusion chromatography (SE-UPLC), non-reducing and reducing capillary electrophoresis SDS (NR CE-SDS and R CE-SDS), peptide mapping and particle counting (PC).
The invention also features methods for treating Rheumatoid Arthritis in a subject in need thereof by administering a therapeutically effective amount of any of the antibody formulations described or exemplified herein. The invention also features methods for treating Juvenile Idiopathic Arthritis, Psoriatic Arthritis, Ankylosing Spondylitis, Crohn's Disease, Plaque Psoriasis, Ulcerative Colitis, Inflammatory Bowel Disease, Hidradenitis Suppurativa, or Refractory Asthma by administering a therapeutically effective amount of any of the antibody formulations described or exemplified herein. Therapeutic efficacy is attained, for example, by the ONS-3010 antibody present in the administered formulation. Administration of the antibody formulation may be according to any suitable route, preferably by injection, and more preferably by subcutaneous injection. Administration may be carried out under the direction or supervision of a medical practitioner.
The antibody formulations described and exemplified herein may be for use as a medicament. The antibody formulations described and exemplified herein may be for use in the manufacture of a medicament. The formulations may be for use in the treatment of Rheumatoid Arthritis. The formulations may be for use in the treatment of Juvenile
Idiopathic Arthritis. The formulations may be for use in the treatment of Psoriatic Arthritis. The formulations may be for use in the treatment of Ankylosing Spondylitis. The
formulations may be for use in the treatment of Crohn's Disease. The formulations may be for use in the treatment of Plaque Psoriasis. The formulations may be for use in the treatment of Ulcerative Colitis. The formulations may be for use in the treatment of Inflammatory Bowel Disease. The formulations may be for use in the treatment of
Hidradenitis Suppurativa. The formulations may be for use in the treatment of Refractory Asthma.
The invention also features kits. The kits may be used, for example, to practice any of the methods described or exemplified herein. In some aspects, a kit comprises any antibody formulation described or exemplified herein, and instructions for using the antibody formulation in any of the methods or uses described or exemplified herein. The kit may comprise a device for injecting the antibody formulation into a subject, including but not limited to a syringe and needle, or catheter.
The instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Rheumatoid Arthritis, including instructions for injecting the antibody formulation into a Rheumatoid Arthritis patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Juvenile Idiopathic Arthritis, including instructions for injecting the antibody formulation into a Juvenile Idiopathic Arthritis patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Psoriatic Arthritis, including instructions for injecting the antibody formulation into a Psoriatic Arthritis patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Ankylosing Spondylitis, including instructions for injecting the antibody formulation into a Ankylosing Spondylitis patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Crohn's Disease, including instructions for injecting the antibody formulation into a Crohn's Disease patient in need thereof. In some aspects, the
instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Plaque Psoriasis, including instructions for injecting the antibody formulation into a Plaque Psoriasis patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Ulcerative Colitis, including instructions for injecting the antibody formulation into a Ulcerative Colitis patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Inflammatory Bowel Disease, including instructions for injecting the antibody formulation into an Inflammatory Bowel Disease patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Hidradenitis Suppurativa, including instructions for injecting the antibody formulation into a Hidradenitis Suppurativa patient in need thereof. In some aspects, the instructions included with the kit may include instructions for administering the antibody formulation in a method for treating Refractory Asthma, including instructions for injecting the antibody formulation into a Refractory Asthma patient in need thereof.
The following examples are provided to describe the invention in greater detail. They are intended to illustrate, not to limit, the invention.
Example 1
Materials and Methods
Introduction. Antibody ONS-3010 represents a biosimilar of adalimumab, and has been reformulated for enhanced storage stability. It is believed that the modifications to the buffer of the formulation composition may reduce the incidence of injection-site reaction, including injection pain and a burning sensation observed from subcutaneous administration of adalimumab (Kaiser C et al. (2012) Rheumatol. Int. 32:295-9, and
Fransson J et al. (1996) J. Pharm. Pharmacol. 48:1012-5). Current adalimumab formulations include (in addition to the antibody), sodium chloride, monobasic sodium phosphate dihydrate, dibasic sodium phosphate dihydrate, sodium citrate, citric acid monohydrate, mannitol, polysorbate 80, and sterile water for injection. The experimental approach described below included three experimental series of development work to reformulate adalimumab for therapeutic administration.
The first experimental series of studies focused on buffer composition, strength, and ability to achieve the desired pH of about 5.2. The second experimental series of experiments utilized stressed stability studies with refined set of formulation conditions based on results of experimental series 1. Sodium chloride concentration was probed in experimental series 2. The third experimental series of formulation development studies compared three conditions, including a control of the adalimumab reference product buffer (per 0.8 ml: 40 mg adalimumab, 4.93 mg sodium chloride, 0.69 mg monobasic sodium phosphate dihydrate, 1.22 mg dibasic sodium phosphate dihydrate, 0.24 mg sodium citrate, 1.04 mg citric acid monohydrate, 9.6 mg mannitol, 0.8 mg polysorbate 80, and Q..S. sterile water for injection, pH 5.2). For each buffer system, there was one condition for the adalimumab reference formulation level of NaCI and mannitol, and a condition where those levels were modified relative to the adalimumab reference formulation (lower NaCI, higher mannitol (LS/HM)). These modifications resulted in formulations of comparable osmolality to the adalimumab reference formulation, while maintaining isotonicity.
Dynamic Light Scattering (DLS). The DLS testing method used a Wyatt DynaPro™ Plate Reader to provide information on protein size distribution and overall colloidal stability in solution. Hydrodynamic radius provided information on the presence of aggregation and confirmation of the molecule's structure in solution. DLS testing provided an orthogonal measure of size distribution in solution under non-denaturing conditions.
Differential Scanning Calorimetry (DSC). Differential scanning calorimetry measured the melting transitions for the protein and, thus, provided information on protein thermal stability in solution. Calorimetry was performed using a GE VP Capillary DSC system. The protein was heated from 25° C to 95° C at an optimized scan rate allowing the melting transitions (Tm) to occur while the protein is unfolding. A buffer control was heated alongside the sample and used to calculate melting temperatures and transitions. The DSC profile was typical of antibodies and demonstrated that the protein folded into distinct domains.
Size Exclusion Chromatography (SE-UPLC). SE-UPLC was used to monitor ONS-3010 size variant distribution. The SE-UPLC testing method separates proteins based on size. The method is isocratic with a sodium phosphate running buffer, using a Waters Acquity UPLC BEH200 SEC column (1.7 μιη, 4.6 x 150 mm). Peaks were monitored using absorbance at 280 nm. Species eluting before the monomer peak were aggregates (HMWS) and peaks eluting after the monomer peak were degradants (LMWS).
NonReducing and Reducing Capillary Electrophoresis SDS (NR CE-SDS and R CE-SDS). CE-SDS analysis was used to compare ONS-3010 size variants under denaturing conditions, with both non-reducing and reducing conditions, using a Beckman PA800 plus instrument. Capillary gel electrophoresis provides automated analysis of reduced and non-reduced proteins by size to determine protein purity and/or heterogeneity. Samples were treated with either an alkylation or reducing agent and SDS was bound to all proteins via a sample buffer. A polymer matrix was filled into the capillary prior to sample analysis. Samples were electrokinetically introduced to the capillary by an applied voltage, then electrophoresis was performed by applying a constant voltage to the capillary. The SDS treated proteins have mass to charge properties that are proportional to the protein weights, which allows for the separation of the SDS-bound proteins by the differences in molecular weight. Test article proteins were quantified by UV detection at 220 nm.
Modulation of TNF-alpha activity: L929 Cell-Based Bioassay. The primary mechanism of action of adalimumab is the neutralization of circulating TNF-alpha. L929 cell-based bioassay measures cell death/viability. TNF-alpha induces cytotoxicity in L929 cells; relative potency of adalimumab was measured by monitoring live cells through a luminescent tag.
Peptide mapping. N-terminal sequence variants, C-terminal sequence variants, oxidation, deamidation, succinimide formation, isomerization are measured using peptide mapping LC-MS methodologies.
Particle count. The level of aggregates and particulates is a critical quality attribute to assess for liquid protein formulations. The presence of aggregates and particulates may negatively impact product quality. Example 2
Results
Experimental series 1. The first experimental series of studies focused on buffer composition, strength, and ability to achieve the desired pH of 5.2. Buffers tested included citrate and phosphate (which are used in the reference product formulation) and acetate (Table 1). Sodium chloride and mannitol concentrations (equivalent to those in adalimumab reference formulation) were added to conditions throughout experimental series 1 experiments. From this experimental series of experiments, it was observed that some buffers were better than others at achieving and maintaining the desired pH in the range of 4.9 - 5.5 (0.3 pH units outside of the adalimumab reference formulation). SE-UPLC purity, in particular, was highly correlated with pH, and the use of acetate buffer resulted in preferable profiles (Figures 1 and 2).
Table 1. Round 1 Formulation Conditions
Figure imgf000027_0001
Y: NaCI and mannitol included at Adalimumab reference buffer concentrations of 4.93 mg/0.08 mL and 9.6 mg/0.8 mL, respectively
DSC thermograms were helpful in assessing product stability toward thermal denaturation. All traces showed two dominant thermal transitions: a larger one after 72° C, and a smaller one after 80° C. Under certain conditions, an additional shoulder is seen after 60° C, which is believed to indicate the beginning of an unfolding process under these formulation conditions (Figure 3). These latter formulations were excluded from
subsequent experimental series of screening.
Dynamic light scattering (DLS) was used to monitor the hydrodynamic radius Rh (size) of protein molecules in solution. Hydrodynamic radius size in the 5 - 6 nanometer range under lower (~ Img/mL) protein concentration are typical for monomeric monoclonal antibodies (about 140 kDa in size); this size increases with protein concentration, possibly due to crowding, self-association, or aggregation. Such higher sizes should typically be avoided under formulation conditions since they are indicative of an inherently unstable condition. Hydrodynamic radii of ONS-3010 in experimental series 1 formulation conditions were monitored at two protein concentrations for more complete picture of colloidal stability. Rh was not dominated by pH (Figure 4): there was considerable variation in Rh even within a relatively narrow pH range, underscoring the impact of buffer composition on colloidal stability. The conditions that had Rh≤ adalimumab reference formulation of 8.0 nm were selected for further evaluation in experimental series 2.
Experimental series 2. The second experimental series utilized stressed stability studies with a refined set of formulation conditions based on results of experimental series 1. Acetate buffer is of particular interest at the 20 mM level. Sodium chloride
concentration was also evaluated. Some conditions matched the adalimumab reference formulation NaCI levels, while others did not contain NaCI (Table 2).
Table 2. Experimental Series 2 Formulation Conditions
Figure imgf000028_0001
Y: NaCI and mannitol included at Adalimumab reference (AR) buffer concentrations (4.93 mg / 0.8 mL and 9.6 mg / 0.8 mL, respectively). N: no NaCI added
Experimental series 2 formulation buffers containing NaCI were less stable upon incubation at 55° C for up to 14 days as compared to the buffers without NaCI. As shown in
Figure 5, time at elevated temperature caused both aggregation (increasing SE-UPLC high molecular weight species or HMWS), as well as degradation and fragmentation (increasing low molecular weight species or LMWS). Formulation conditions containing NaCI in these experiments showed comparable rates of aggregation and fragmentation relative to that of the adalimumab reference formulation. Formulations lacking NaCI, however, exhibited improved stability toward both mechanisms of aggregation and degradation. This is illustrated in Figure 6, which displays overlaid chromatograms of ONS-3010 formulated with the adalimumab reference buffer, formulated with a NaCI-containing acetate buffer, and formulated with an acetate buffer without NaCI. Figure 7 and Figure 8 highlight trends in
SE-UPLC aggregation and fragmentation, respectively. The removal of NaCI appeared to also correlate with improved colloidal stability as measured by DLS, and with improved stability in CEX-HPLC. For experimental series 3, lower NaCI conditions were designed which reduced (but did not eliminate) NaCI levels, while adjusting mannitol concentrations to result in osmolality levels close to that of reference product formulation.
Table 3 below summarizes the experimental series 2 conditions and their analytical results, highlighting reasons for their inclusion or exclusion from experimental series 3 investigations. In general, conditions selected for experimental series 3 showed comparable or improved stability toward thermal and chemical denaturation as monitored by a variety of orthogonal techniques (SE-UPLC, CEX-HPLC, CE-SDS). Relative potency was also assessed using the L929 cell-based potency assay, and colloidal stability was monitored with DLS. Finally, all samples were visually monitored throughout the study (and haziness upon dilution for testing became an exclusion criterion).
Table 3. Experimental Series 2 Data Summary
Figure imgf000029_0001
Experimental series 3. The final experimental series of formulation development studies compares three conditions including the adalimumab reference formulation as a control (Table 4). The other two reformulation conditions use acetate buffer. There is one acetate buffer matching the adalimumab reference level of NaCI and mannitol, and an acetate buffer where those levels are modified relative to the adalimumab reference (AR) formulation (lower NaCI, higher mannitol(LS/HM)). These modifications result in
formulations of comparable osmolality to the adalimumab reference, maintaining isotonicity.
Table 4. Experimental Series 3 Formulation Conditions.
Figure imgf000030_0001
Y: NaCI and mannitol included at Adalimumab reference (AR) buffer concentrations (4.93 mg / 0.8 mL and 9.6 mg / 0.8 mL, respectively). LS/HM : lower NaCI and higher mannitol levels.
A comprehensive series of stressed, accelerated, and real-time stability studies were executed as part of experimental series 3 formulation development (Table 5). Real-time studies were conducted in glass vials, both to provide a container contact similar to that in final drug product presentation (type 1 borosilicate glass) and to facilitate assessment of product appearance and particle formation. In addition to incubation of ONS-3010 under different temperatures as a liquid, the product will be stored under frozen conditions (both -20° C and -80° C) as well as exposed to repetitive cycles of frozen to liquid transitions. A forced oxidation study and a shaking/shear force study provide additional information to help inform final formulation selection.
Table 5. Components and Scope of Round 3 Experiments.
Study Element Condition Duration Analytical Testing Findings
SE-UPLC, CE-SDS,
Stressed CEX-H PLC, peptide Condition 3 best across all
55°C Up to 7 days
stability mapping, L929 assays
bioassay
SE-UPLC, CE-SDS, Acetate formulations CEX-H PLC, peptide comparable to Adalimumab
37°C Up to 28 days mapping, L929 reference for CEX-H PLC, SE-
Accelerated bioassay, UPLC, CE-SDS, peptide map stability appearance and bioassay.
SE-U PLC. CE-SDS, Acetate formulations
25°C Up to 28 days CEX-HPLC, peptide comparable to Adalimumab mapping, L929 reference for CEX-HPLC, SE- bioassay, UPLC, CE-SDS, peptide map appearance and bioassay.
All formulations comparable
SE-UPLC, CE-SDS, to Adalimumab reference. CEX-H PLC, peptide Lower salt condition 3
Real-time
2 - 8°C 0,lm,5m,12m,18m mapping, L929 reduces particles up to 28 stability
bioassay, particle days. Particle count at 28 count, appearance day better than
Adalimumab reference.
All formulations DLS comparable or better than Adalimumab reference.
DLS, DSC,
Additional At time Lower salt helps Rh.
At time zero osmolality, Particle
characterization zero Osmolality all isotonic.
count
Particle count T=0 comparable or better than Adalimumab reference.
No change in SE-U PLC, slight
1% t-butyl
changes in CEX (condition 3
Forced hydrogen SE-U PLC, CEX-H PLC,
Up to 8 hours better), clear oxidation in oxidation peroxide, peptide mapping
peptide map (condition 3 25°C
better)
SE-UPLC, CE-SDS,
CEX-H PLC, peptide All formulations comparable
Frozen stability -80°C 0,lm,5m,12m,18m
mapping, L929 to Adalimumab reference bioassay
-20°C shows some H MWS (noncovalent) with higher
Freeze-thaw -20°C and SE-UPLC, CE-SDS mannitol, all formulations
Up to 5 cycles
cycles -80°C (NR) comparable to Adalimumab reference at
-80°C
All conditions show comparable or better
SE-U PLC, CE-SDS, protection to shear relative
37°C, with
CEX-H PLC, L929 to Adalimumab reference and
Shaking study Up to 28 days bioassay, selected for SE-UPLC, CE-SDS and without
peptide map, bioassay. CEX showed Tween
appearance acetate conditions comparable to Adalimumab reference.
Example 3
Characterization of Material
Dynamic Light Scattering. Dynamic Light Scattering was used to measure the hydrodynamic radius (Rh) of ONS-3010 at four different protein concentrations (see Figure 9 for graphical representation of the results). The adalimumab reference formulation, at 50 mg/ml, has an average hydrodynamic radius of 8.0 nm. Condition 2 (acetate) shows similar values at the full 50 mg/ml concentration, while condition 3 (acetate LS/HM) with lower salt shows a smaller Rh value, correlated with better control of self-association at higher protein concentrations. Lower amounts of sodium chloride as used in condition 3 are enough to disrupt the crowding/association taking place at 50 mg/mL used under typical formulation conditions. At lower concentrations, Rh values converge on values in the 5 - 6 nm range, typical for monomeric monoclonal antibodies. Condition 3 (Acetate LS/H M) results in the lowest Rh values over the entire range of protein concentrations measured.
The presence of minor quantities of sodium chloride (26.35 mM Sodium Chloride vs. 105.45 mM Sodium Chloride) in the buffered formulation prevents crowding of the individual antibody molecules at high protein concentrations of 50mg/mL. This
phenomenon is further confirmed by a lower hydrodynamic radius for the low sodium chloride containing the buffered formulation (Figure 9 Acetate LS/HM buffer Condition 3 50mg/mL). This is a unique synergistic effect of sodium chloride (lower concentrations) with acetate buffer that prevents crowding and reduces aggregation (HMWS) (Figures 11 and 12). It is believed that lower concentrations of sodium chloride and the absence of citrate in the formulation could be associated with better patient acceptability (reduced irritation and pain at site of injection). Modification of the buffer composition (lower concentration or removal of citrate, specifically) may reduce the incidence of injection-site reaction (e.g., a "burning sensation") upon subcutaneous administration of the drug.
Another such ONS-3010 material from the 200 L pilot scale was tested for its colloidal properties at ~50mg/mL after storage in a glass vial (type I borosilicate) for 17 months at 2-8°C. The tested lot was formulated into both the acetate LS/HM buffer formulation (BDS-O) and the adalimumab reference formulation (BDS-H). As shown in Table 6, the hydrodynamic radius for BDS-0 is about 5.5 nm, which is significantly lower tha n that for BDS-H (7.9 nm). This difference in hydrodynamic radius (Rh) is indicative of increased colloidal stability in the buffered formulation compared to the adalimumab reference formulation. Moreover this important colloidal property (Rh) remains unchanged over the storage period of 17 months at 2-8°C, indicating greater storage stability for the buffered formulation.
Table 6. ONS-3010 DLS results.
Protein concentration
Sample Avg. Rh(nm)
(mg/mL)
BDS-0 49.9 5.5 53.7
Osmolarity. Osmolality values for the three conditions were measured using the Nova Flex instrument. All conditions were similar to one another and to the adalimumab reference formulation, and were in the isotonic range of 290 - 340 mOsm/kg (Table 7).
Table 7. ONS-3010 Experimental Series 3 Osmolality Values
Figure imgf000033_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Particle Count. Particle analysis was conducted using a HIAC Model 9703+ system following a modified USP method (allowing for detection of particles as small as 2 μιη). Cumulative results for all size ranges are shown in Table 8, with counts per container calculated based on the 0.8 ml pre-filled syringe presentation. The 10 and 25 μιη size bins are specifically tracked per the USP method <788>. Values for all conditions are well below the limits of≤ 600 cumulative counts per container for > 25 micron particles, and≤ 6000 cumulative counts per container for > 10 micron particles. Lower salt (formulation condition 3) appears to further reduce particles at 2-10 micron relative to the adalimumab reference formulation and the higher salt formulation (Table 8).
Table 8. Particle Count Results for Experimental Series 3 Time Zero and Day 28 Samples
Figure imgf000033_0002
100 2 2 0 0
2 808 646 1788 1430
3 396 317 1058 846
5 96 77 214 171
10 30 24 36 29
Acetate
15 10 8 22 18
25 6 5 6 5
50 4 3 0 0
100 0 0 0 0
2 356 285 290 232
3 240 192 178 142
5 112 90 72 58
Acetate 10 56 45 18 14
LS/HM 15 30 24 10 8
25 12 10 0 0
50 2 2 0 0
100 0 0 0 0
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Stressed Stability (55° C). To probe the behavior of ONS-3010 toward a stressed condition of elevated temperature, samples were incubated at 55° C for up to 7 days, a nd then tested by multiple analytical methods. While 55° C is well above storage conditions and any expected short-term handling conditions that would be encountered in the clinic, the stressed stability arm is extremely useful at highlighting formulation ability to protect from a myriad of forced degradation events that dominate at higher temperature. For both adalimumab and ONS-3010, 55° C is lower than the initial onset of thermal denaturation as monitored by differential scanning calorimetry.
SE-UPLC. Exposure of ONS-3010 to 55° C generated both higher and lower molecular weight species (HMWS and LMWS) (Table 9), both of which can be monitored by SE-UPLC. From time zero to day 7, there were marked increases in dimer (with a retention time of approximately 3.1 minutes) and species larger-than-dimer with earlier retention times, both counted as HMWS. For fragmentation, a distinct peak was formed off the backside of the monomer peak at ~ 3.9 minutes, plus an additional peak at 4.5 minutes (Figure 10). After 7 days of incubation, there were clear differences between the SE-UPLC chromatograms of the formulation conditions (Figure 11). I n particular, the Acetate LS/H M condition illustrated protection toward H MWS formation relative to the adalimumab reference formulation and to the other formulation conditions. This is also displayed in Figure 12. Table 9. SE-UPLC data for Stressed Stability (55° C) Day 1 to Day 7
Figure imgf000035_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
CEX-HPLC. Cation exchange chromatography of samples treated at 55° C provides a broad view of many physicochemical changes that can manifest themselves as cha nges in molecular charge. This includes specific charge based modifications such as deamidation, isomerization, and pyroglutamine formation, but can also reveal more subtle
conformational shifts that can begin to occur at elevated temperatures. CEX-HPLC profiles were monitored for time zero through day 2 sam ples (Table 10).
Table 10. CEX data for Stressed Stability (55° C)
Figure imgf000035_0002
Adalimumab reference T=0 2-8° C 15.5 64.4 18.5 1.7
Acetate T=0 2-8° C 15.7 64.1 18.8 1.5
Acetate LS/H M T=0 2-8° C 15.5 64.0 18.6 1.8
Adalimumab reference 55° C Day 1 19.6 58.5 21.0 0.9
Acetate 55° C Day 1 19.4 58.8 20.8 1.1
Acetate LS/H M 55° C Day 1 20.4 58.1 19.6 2.0
Adalimumab reference 55° C Day 2 24.9 53.7 20.8 0.6
Acetate 55° C Day 2 24.4 54.3 20.5 0.9
Acetate LS/H M 55° C Day 2 26.2 53.5 19.4 0.9
Refer to Table 4 for buffer components. LS/HM: Lower NaCI and higher mannitol levels
Representative chromatograms for samples in the adalimumab reference formulation are shown in Figure 13, with an overlay of 55° C day 2 samples. Trends in CEX % Main, Acidic and Basic species are shown in Figure 14, Figure 15, and Figure 16. The acetate LS/HM buffer formulation had a similar CEX profile to that of the adalimumab reference formulation after treatment at 55 degrees C for two days. Minor differences are within assay variability.
CE-SDS R/NR. To provide an orthogonal view of size variants (denatured vs. the non- denatured SE-UPLC methodology), CE-SDS was used under both non-reducing and reducing conditions. The strong differentiation between formulation conditions observed with SE- UPLC is not seen under analysis by denaturing techniques, indicating that the size variants formed are largely noncovalent in nature.
Bioassay. In general, all samples tested showed comparable relative potency in the bioassay within method variability. There was no measurable change in potency after the 7 day treatment at 55 degrees C, demonstrating stability toward thermal denaturation.
Peptide Map. Peptide mapping allowed a more specific view of modifications in ONS-3010. The 55° C incubation temperature fostered certain chemical modifications that are not commonly observed at lower temperatures, such as pyroglutamic acid formation of the N-terminal heavy chain and specific isomerization events. Condition 3 (Acetate LS/HM) appeared to have the most protection against this assortment of chemical modifications (Figure 17, Figure 18, Figure 19, Figure 20, and Table 11). The oxidation of methionine 256 in the CH2 domain in condition 3 day 7 stressed sample, for example, was kept to a level within the unstressed range. C-terminal variants and glycosylation levels remained constant throughout the treatment. Table 11. Selected ONS-3010 post-translational modifications monitored by peptide mapping in stressed stability samples
Figure imgf000037_0001
Refer to Table 4 for buffer components. LS/HM: Lower NaCI and higher mannitol levels
Accelerated Stability (37° C and 25° C). Accelerated (37° C and 25° C) and real-time stability studies were conducted in glass vials. Samples were pulled for analysis by SE-UPLC, CEX-HPLC, CE-SDS, Appearance, Bioassay, and peptide mapping (at selected timepoints). At 25° C up to 28 days, acetate formulations were comparable to adalimumab reference formulations for CEX-HPLC, SE-UPLC, CE-SDS, peptide map and Bioassay. SE-UPLC. An overlay of 37° C incubated chromatograms for the adalimumab reference formulation is shown in Figure 21, with a zoomed overlay of all three conditions at day 28 in Figure 22. All three conditions have similar SE-UPLC profiles after 28 days at 37° C, with main peak/monomer purity levels ~ 96%. The formation of a back shoulder on the monomer peak (4 minutes retention time) is observed in all formulation conditions, a nd was at the integration threshold for some earlier time points (leading to some variation in qua ntitating LMWS, see Table 12 for numerical values, noting the punctuated increase between day 21 and 28).
Table 12. SE-UPLC for 37° C accelerated stability samples
Figure imgf000038_0001
Refer to Table 4 for buffer components. LS/H M : Lower NaCI and higher mannitol levels
CEX-HPLC. CEX-HPLC analysis of accelerated stability samples at 14 day, 21 day and 28 days of treatment at 37° C (Table 13) showed acetate conditions comparable to the adalimumab reference formulation.
Table 13. CEX-HPLC data for Stressed Stability (37° C)
Figure imgf000038_0002
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels CE-SDS (R/NR). For the 37° C incubated samples, CE-SDS testing reveals similar trends in NR denatured size variants between the three conditions after 28 days. For 37° C R CE-SDS, all conditions relative to the adalimumab reference formulation are comparable after 28 days.
Bioassay. All formulation conditions displayed full potency in the L929 bioassay after 28 days incubation at 37° C.
Appearance. Regular visual inspection was conducted for the accelerated and realtime stability samples. Specifically, the samples were monitored for any change in color, clarity, and the presence of particles, proteinaceous or not. In general, the ONS-3010 samples displayed visual appearance as desired for a protein product. In 37° C incubated samples, all formulations except condition 3 displayed some particle formation by day 20, while condition 3 (acetate LS/HM) remained free from particles at day 28 (Table 14). All formulations showed some visible particles at 26 days at 2-8° C (Table 30).
Table 14. Appearance data for Stressed Stability (37° C)
Figure imgf000039_0001
21 IP IP C
22 IP IP C
25 IP IP C
26 IP IP c
27 IP IP c
28 IP IP c
C = Clear, IP to 5P scale: IP = particle visible, 5P = many particles visible
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Forced Oxidation. A forced oxidation study utilized 1% t-butyl hydrogen peroxide treatment to induce oxidation in ONS-3010 formulation candidates. SE-UPLC, CEX-HPLC, and tryptic peptide map methodologies were used to monitor changes in product quality attributes and specific amino acid residues that are susceptible to oxidation. Oxidative modification is one of the major chemical degradation pathways. Sites of oxidation damage on backbone or side-chain can change hydrophobicity of protein surfaces. The fingerprint of oxidation by using LC-MS peptide mapping enables a fast and reliable approach for formulation selection.
There were five methionine residues distributed along the sequence of ONS-3010: at residue M4 in the light chain, and residues M34, M83, M256, and M432 in the heavy chain. M34 is within the CDR.
Based on the peptide mapping data, the Fc region methionine residues M256 and M432 were the dominant residues modified by oxidation (Figure 23 and Figure 24).
Formulation condition #3 provided the most protection overall to oxidation. Oxidized species of residues M4 and M83 were below the method LOO even in stressed conditions. Upon stress, oxidized M34 was at the LOO, with the three conditions comparable within method variability.
For CEX-HPLC, the oxidative stress treatment results in a decrease in main peak and a corresponding increase in basic peak percentages of several percent (Table 15). Condition 3 (Acetate LS/HM) appeared more protective to the oxidation than the other conditions. SE-UPLC values are essentially unchanged upon treatment.
Table 15. CEX-HPLC data for Forced Oxidation 37° C % Acidic % Main % Basic % Extra Basic
Sample Description
Species Species Species Species
Adalimumab Reference T=0 2-8° C 15.3 65.2 18.6 0.8
Acetate T=0 2-8° C 15.1 65.4 18.7 0.8
Acetate LS/HM T=0 2-8° C 15.5 65.1 18.6 0.8
Adalimumab Reference 1% TBHP 14.0 63.4 20.9 1.7
Acetate 1% TBHP 14.2 64.1 20.5 1.3
Acetate LS/HM 1% TBHP 14.6 64.6 19.9 0.9
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Freeze-Thaw Cycling. Freeze-thaw cycling was conducted for sam ples in the candidate formulations at two temperatures: -20° C and -80° C. Samples were placed in freezers set to the appropriate temperature and allowed to freeze thoroughly (for at least one hour). Samples were then removed from the freezer and allowed to thaw at 25° C (approximately 1 hour). This freezing step plus the thawing step constituted a single cycle. Samples were subjected to up to 5 freeze-thaw cycles, and then a nalyzed together by SE- UPLC, with a subset of samples also tested by NR CE-SDS.
All formulation conditions appeared to be stable to multiple freeze-thaw cycles at -80° C, with main peak purity values after five cycles equivalent to time zero values. With -20° C cycling, some increases in % H MWS were observed, more prevalent in the higher mannitol formulation condition 3 (Table 16). It is believed that this may be indicative of mannitol crystallization fostered at this lower frozen tem perature. The adalimumab reference formulation and condition 3 exhibited similar patterns upon cycling as monitored by N R CE-SDS.
Table 16. SE-UPLC data for Freeze/Thaw ("C" = freeze/thaw cycle 1-5)
Figure imgf000041_0001
Adalimumab Reference F/T C2 25/-200 C 0.71 99.22 0.07
Acetate F/T C2 25/-200 C 0.71 99.14 0.14
Acetate LS/HM F/T C2 25/-200 C 1.42 98.44 0.14
Adalimumab Reference F/T C2 25/-800 C 0.73 99.16 0.1
Acetate F/T C2 25/-800 C 0.70 99.18 0.12
Acetate LS/HM F/T C2 25/-800 C 0.66 99.20 0.14
Adalimumab Reference F/T C3 25/-200 C 0.73 99.15 0.1
Acetate F/T C3 25/-200 C 0.72 99.13 0.15
Acetate LS/HM F/T C3 25/-200 C 1.40 98.48 0.12
Adalimumab Reference F/T C3 25/-800 C 0.73 99.15 0.13
Acetate F/T C3 25/-800 C 0.70 99.15 0.14
Acetate LS/HM F/T C3 25/-800 C 0.65 99.26 0.09
Adalimumab Reference F/T C4 25/-200 C 0.73 99.17 0.11
Acetate F/T C4 25/-200 C 0.71 99.14 0.14
Acetate LS/HM F/T C4 25/-200 C 1.37 98.56 0.06
Adalimumab Reference F/T C4 25/-800 C 0.73 99.14 0.12
Acetate F/T C4 25/-800 C 0.69 99.24 0.07
Acetate LS/HM F/T C4 25/-800 C 0.65 99.21 0.14
Adalimumab Reference F/T C5 25/-200 C 0.72 99.17 0.11
Acetate F/T C5 25/-200 C 0.70 99.15 0.14
Acetate LS/HM F/T C5 25/-200 C 1.37 98.57 0.06
Adalimumab Reference F/T C5 25/-800 C 0.72 99.16 0.12
Acetate F/T C5 25/-800 C 0.70 99.15 0.15
Acetate LS/HM F/T C5 25/-800 C 0.64 99.24 0.12
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Shaking Studies. In order to assess the protective ability of the formulation toward shear forces, a shaking study was conducted in glass vials (0.5 mL fills) placed in an orbital shaking incubator set to 150 rpm at 37° C. A second arm contained formulations without the addition of polysorbate 80. Samples were tested with the following methods: SE-UPLC, CEX-HPLC, CE-SDS (R/NR), L929 BioAssay, Peptide Map, and Appearance. For both shaking studies with and without polysorbate 80, based on SE-UPLC, conditions 2 and 3 (with main peak purities ~ 96% at day 28) were comparable or slightly better to the adalimumab reference formulation (Tables 17-24). CEX-HPLC results showed no significant difference in the acetate formulations compared to the adalimumab reference formulation (Tables 25-27). Day 28 samples displayed full potency in the L929 bioassay, and CE-SDS (R/NR) values are similar across conditions. At day 28, all conditions showed some visible particulate formation: condition 3 showed more protection than the other conditions (Tables 28 and 29).
Table 17. SE-UPLC data for Shaking Study with Polysorbate 80 - Day 7
Figure imgf000043_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 18. SE-UPLC data for Shaking Study with Polysorbate 80 - Day 14
Figure imgf000043_0002
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 19. SE-UPLC data for Shaking Study with Polysorbate 80 - Day 21
Figure imgf000043_0003
Refer to Table 4 for buffer components. LS/HM: Lower NaCI and higher mannitol levels Table 20. SE-UPLC data for Shaking Study with Polysorbate 80 - Day 28
Figure imgf000043_0004
Adalimumab Reference Shaking D28 1.51 95.94 2.56
Acetate Shaking D28 1.78 95.81 2.41
Acetate LS/HM Shaking D28 1.58 96.07 2.35
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 21. SE-UPLC data for Shaking Study without Polysorbate 80 - Day 6
without Polysorbate 80 (Tw)
Figure imgf000044_0001
Refer to Table 4 for buffer components. LS/H M : Lower NaCI and higher mannitol levels Table 22. SE-UPLC data for Shaking Study without Polysorbate 80 (Tw) - Day 13
Figure imgf000044_0002
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 23. SE-UPLC data for Shaking Study without Polysorbate 80 (Tw) - Day 20
Figure imgf000044_0003
Refer to Table 4 for buffer components. LS/H M : Lower NaCI and higher mannitol levels Table 24. SE-UPLC data for Shaking Study without Polysorbate 80 - Day 28
Figure imgf000044_0004
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 25. CEX-HPLC data for Shaking Study with Polysorbate 80 - Day 14
Figure imgf000045_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 26. CEX-HPLC data for Shaking Study with Polysorbate 80 - Day 28
Figure imgf000045_0002
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 27. CEX-HPLC data for Shaking Study without Polysorbate 80 (Tw) - Day 28
Figure imgf000045_0003
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 28. Appearance data for Shaking Study with Polysorbate 80
Figure imgf000045_0004
10 2P IP IP
11 2P IP IP
14 2P 2P IP
15 2P 2P IP
16 2P 2P IP
17 2P 2P IP
18 2P 2P IP
21 2P 2P IP
22 2P 2P IP
23 2P 2P IP
24 2P 2P IP
25 2P 2P IP
28 2P 2P IP
C = Clear, IP to 5P scale: IP = particle visible, 5P = many particles visible
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 29. Appearance data for Shaking Study without Polysorbate 80
Figure imgf000046_0001
21 C C IP
22 C C IP
23 C C IP
24 C C IP
27 C C IP
28 C C IP
C = Clear, IP to 5P scale: IP = particle visible, 5P = many particles visible
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Real-time Stability at 2 - 8° C and -80° C. Prior to the long term real-time stability study, formulation studies including stressed and accelerated temperature, forced oxidation, exposure high shear and freeze-thaw have been conducted to evaluate formulation candidates. The relationship between thermal stability, aggregation, fragmentation, degradation pathways and potency has been observed as a result of these treatments. Experimental series 3, Condition 3 (acetate LS/HM) has shown more protection than the others in several extreme conditions tested.
Storage at 2 - 8° C and -80° C is anticipated for ONS-3010 drug product. Long term real-time stability studies at 2 - 8° C and -80° C have been conducted with timepoints at: 1 month, 5 month, 12 month and 18 month. Samples have been tested with the following methods: SE-UPLC (Tables 32-34), CEX-HPLC (Tables 35-37), CE-SDS (R/NR) (Tables 38-42), L929 BioAssay (Table 43), Peptide Map (Tables 44-46), Particle Count (day 28 2-8° C Table 8) and Appearance (Tables 30-31).
Testing results on ONS-3010 samples formulated with the three different conditions show no significant biochemical change according to SE-UPLC (Figures 25-28) and bioassay results are equivalent and within method variability. There were no significant differences in the visual appearance for formulation conditions 1 and 3 (Tables 30 and 31). Sub-visible particle counts for day 28 timepoint are recorded in Table 8. At the day 28 timepoint, lower salt (formulation condition 3 Acetate LS/HM) appears to further reduce particles at 2-10 micron relative to the adalimumab reference formulation and the higher salt formulation. Stability evaluation for Condition 2 was discontinued after the 5 month time point and stability sample was subsequently frozen at -80°C for future testing if required.
Table 30. Appearance for 2-8° C Real time incubation Adalimumab Acetate
Day Reference Acetate LS/HM
0 C c C
1 C c c
4 c c c
5 c c c
6 c c c
7 c c c
8 c c c
11 c c c
12 c c c
13 c c c
14 c c c
15 c c c
18 c c c
19 c c c
20 c c c
21 c c c
22 c c c
25 c c c
26 IP IP IP
27 IP IP IP
28 IP IP IP
5 month IP IP IP
12 month IP 2P IP
18 month IP 2P IP
C = Clear, IP to 5P scale: IP = particle visi ble, 5P = mar
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Table 31. Appearance for -80°C Real time incubation
Figure imgf000048_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 32. SE-UPLC % HMWS for 2 - 8°C and -80°C incubation
Figure imgf000049_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 35. CEX-HPLC % Acidic Species for 2 - 8°C and -80°C incubation
Figure imgf000049_0002
2. Not Not Not Not
15.7 15.2 15.0 14.5
Acetate tested tested tested tested
3.
Acetate 15.5 15.0 15.7 16.8 17.0 15.2 16.5 15.3 LS/H M
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 36. CEX-HPLC % Main Species for 2 - 8°C and -80°C incubation
% Main Species
2 - 8°C 2 - 8°C 2 - 8°C 2 - 8°C -80°C -80°C -80°C
Sample 2 - 8°C 28 d
TO 5 month 12 month 18 month 5 month 12 month 18 month
1.
Adalimu
64.4 64.8 64.4 63.2 62.5 65.4 63.1 65.0 mab
reference
2. Not Not Not Not
64.1 64.9 64.5 65.6
Acetate tested tested tested tested
3.
Acetate 64.0 65.2 64.1 63.3 63.1 64.6 64.3 65.1 LS/H M
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 37. CEX-HPLC % Basic Species for 2 - 8°C and -80°C incubation
Figure imgf000050_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 38. CE-SDS Reduced Light Chain % area for 2 - 8°C and -80°C incubation
Figure imgf000050_0002
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 39. CE-SDS Reduced Heavy Chain % area for 2 - 8°C and -80°C incubation
Heavy Chain % area 2 - 8°C 2 - 8°C 2 - 8°C 2 - 8°C -80°C -80°C -80°C
Sample 2 - 8°C 28 d
TO 5 month 12 month 18 month 5 month 12 month 18 month
1.
Adalimu
67.5 67.4 67.5 65.8 65.5 67.7 65.9 66.0 mab
reference
2. Not Not Not Not
67.4 67.9 67.5 67.5
Acetate tested tested tested tested
3.
Acetate 66.4 67.6 67.4 65.7 66.9 67.2 65.8 66.0 LS/H M
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 40. CE-SDS Reduced Intermediate Species % area for 2 - 8°C and -80°C incubation
Intermediate Species % area
2 - 8°C 2 - 8°C 2 - 8°C 2 - 8°C -80°C -80°C -80°C
Sample 2 - 8°C 28 d
TO 5 month 12 month 18 month 5 month 12 month 18 month
1.
Adalimu
2.1 2.0 1.6 0.2 0.6 1.1 0.2 0.9 mab
reference
2. Not Not Not Not
2.0 2.0 1.5 1.2
Acetate tested tested tested tested
3.
Acetate 1.4 2.3 1.7 0.3 0.5 1.4 0.2 0.5 LS/H M
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Table 41. CE-SDS Non-Reduced Main Peak % Area for 2 - 8°C and -80°C incubation
Figure imgf000051_0001
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Table 42. CE-SDS Non-Reduced % Area of Pre-Main Peak Species for 2 - 8°C and -80°C incubation
% Area of pre-main peak species
2 to 8°C 2 to 8°C 2 to 8°C 2 to 8°C 2 - 8°C -80°C -80°C -80°C
Sample
TO 28 d 5 month 12 month 18 month 5 month 12 month 18 month
1.
Adalimu
8.6 5.8 7.8 6.9 6.1 7.6 6.7 6.1 mab
reference
2.
8.2 5.9 7.7 Not tested Not tested 7.8 Not tested Not tested
Acetate
3. 8.2 6.9 7.3 6.6 6.1 7.6 6.9 6.1 Acetate
LS/HM
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels Table 43. Bioassay L929 for 2 - 8°C and -80°C incubation
Figure imgf000052_0001
Refer to Table 4 for buffer components. LS/H M : Lower NaCI and higher mannitol levels
Table 44. N-terminal PyroE for 2 - 8°C and -80°C incubation
Figure imgf000052_0002
*samples at 2 - 8°C for 28 days were tested alone with a separate set of samples at time zero.
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Table 45. Oxidation for 2 - 8°C and -80°C incubation
Figure imgf000052_0003
Refer to Table 4 for buffer components. LS/H M : Lower NaCI and higher mannitol levels
Table 46. Isomerization for 2 - 8°C and -80°C incubation
Sum of Isomerization
2 - 8°C 2 - 8°C 28 2 - 8°C 2 - 8°C 2 - 8°C -80°C -80°C -80°C
Sample
TO d 5 month 12 month 18 month 5 month 12 month 18 month 1.
Adalimu
LOD LOD LOD 0.24 0.23 LOD 0.15 0.11 mab
reference
2. Not Not Not Not
LOD LOD LOD LOD
Acetate tested tested tested tested
3.
Acetate LOD LOD LOD 0.21 0.24 LOD 0.15 0.15 LS/H M
Refer to Table 4 for buffer components. LS/HM : Lower NaCI and higher mannitol levels
Example 4
Forced Degradation Evaluation
The data presented in this Example stem from forced degradation testing, and illustrate the enhanced stability with the acetate LS/HM buffer composition for ONS-3010. Test Sample as follows:
A. 51.0 mg/mL (acetate LS/HM buffer formulation) and 52.6 mg/mL (Adalimumab reference formulation)
B. 48.5 mg/mL (acetate LS/HM buffer formulation) and 50.3 mg/mL (Adalimumab reference formulation)
Purity SE-UPLC: SE-UPLC monitors ONS-3010 size homogeneity under non- denaturing conditions. The SE-UPLC testing method separates proteins based on size. The method is isocratic with a sodium phosphate running buffer, using a Waters Acquity UPLC BEH200 SEC column (1.7μιη, 4.6x150 mm). Peaks are monitored using absorbance at 280 nm. Species eluting before the monomer peak are aggregates (HMWS) and peaks eluting after the monomer peak are degradants (LMWS).
Purity CE-SDS (NR): CE-SDS analysis is used to monitor ONS-3010 size homogeneity under denaturing conditions, with non-reducing conditions, using a Beckman PA800 plus instrument. Samples are treated with an alkylation agent and SDS is bound to all proteins via a sample buffer. A polymer matrix is filled into the capillary prior to sample analysis. Samples are electrokinetically introduced to the capillary by an applied voltage, then electrophoresis is performed by applying a constant voltage to the capillary. The SDS treated proteins have a mass to charge properties that are proportional to the protein weights which allows for the separation of the SDS-bound proteins by the differences in molecular weight. Test article proteins are quantified by UV detection at 220 nm. Peptide Map: UPLC Peptide mapping is used to characterize a protein's primary structure. ONS-3010 is digested with trypsin and the resulting peptides are separated by RP-UPLC. The characteristic peptide map fingerprint is analyzed for peak relative retention time and overall peak pattern compared with the reference.
55°C Treatment: A 55°C study was conducted on 2 lots of ONS-3010 in solution. The results indicated the ONS-3010 antibody in the acetate LS/HM buffer formulation demonstrated a slower rate of degradation compared to the same antibody in the
Adalimumab reference formulation, displaying enhanced stability at 55°C for aggregation and % intact protein on non-reduced CE as well as peptide map. There was also a visual difference due to the antibody in the Adalimumab reference buffer becoming opalescent by day 10 of treatment compared to the antibody in the acetate LS/HM buffer formulation, which remained clear throughout 10 days of treatment.
Tables 48-50 show the longest timepoints with significant differences. There consistent trend throughout the treatment, however.
Table 47. Summary of Forced Temperature Test for ONS-3010 at 55°C for 10 Days
Figure imgf000054_0001
Note: No change in potency for treated samples in both formulations
Table 48. Summary of Forced Temperature Test for ONS-3010 and Adalimumab at 55°C for 10 Days
Figure imgf000054_0002
Oxidation Isomerization
Total
Product Buffer Treatment M256 M432 D31 D284 Pyro-E
Deamidation
Untreated 29 3.1 1.2 <LOD 0.1 1.2
55°C Dl 29.4 3.3 1.0 0.2 0.9 1.9
ONS-3010- Acetate
55°C D2 30.5 3.8 1.0 0.5 2.3 2.6 BDS LS/H M buffer
55°C D7 37.7 5.0 1.7 2.9 8.4 7.0
55°C D10 38.8 5.5 1.9 4.9 12.5 8.5
Untreated 26.9 2.6 1.1 <LOD 0.1 1.2
55°C Dl 29.7 4.0 1.2 0.1 1.3 2.2
ONS-3010- Adalimumab
55°C D2 31.9 4.6 1.5 0.5 3.7 3.6 BDS reference
55°C D7 30.7 5.7 2.6 2.2 11.0 8.8
55°C D10 39.2 6.6 2.4 3.8 14.4 12.1
Note: No change in potency for treated samples in both formulations. pH 3.0 Treatment: Treatment at pH 3.0 was conducted on 2 lots of ONS-3010 antibody. The ONS-3010 BDS in the acetate LS/HM buffer formulation demonstrated a higher level of stability based on aggregation compared to the same antibody in the Adalimumab reference formulation. There was also a visual difference, with the antibody in the adalimumab reference buffer becoming cloudy after 12 hours of treatment compared to the antibody in the acetate LS/HM buffer formulation, which remained clear throughout 12 hours of treatment at pH 3.
Table 49. Summary of Low pH Test for ONS-3010 at pH 3 for up to 12 Hours*
Figure imgf000055_0001
SEC
% % %
Product Buffer Treatment Visual
Aggregate Monomer Degradant
1 Hrs. clear 11.1 88.7 0.2
2 Hrs. clear 15.8 84.0 0.2
3 Hrs. clear 18.3 81.5 0.3
12 H rs. cloudy 30.5 69.0 0.5
Note: No change in potency for treated samples in both formulations.
*There is a 2-3 fold difference in amount of aggregates formed in the adalimumab reference (citrate-phosphate buffer) vs. the acetate LS/HM buffer over 1-3 Hours at pH 3.0. These data suggest that acetate LS/HM buffer offers greater stability against aggregation at pH 3.0. The acetate LS/HM buffer formulation shows greater protection at low pH compared to the adalimumab reference formulation.
Example 5
Summary
Results based on stressed and accelerated stability studies indicate promising reformulation conditions with comparable and/or improved degradation rates relative to that of the antibody in the adalimumab reference formulation. Formulation condition #3 (acetate LS/HM) used in experimental series 3, provides a protective effect to ONS-3010 relative to the adalimumab reference buffer. This observation is consistent in forced oxidation and shaking studies, as well as at elevated temperatures. The acetate LS/HM buffer formulation provides improved thermal, conformational and colloidal stability to ONS-3010, which indeed translates into comparable shelf life and/or improved product quality.
After 18 months of storage at 2 - 8°C and -80°C, Condition 1 (adalimumab reference) and Condition 3 (acetate LS/HM buffer formulation) were stable and showed no significant change.
Adalimumab Reference Formulation Buffer Components:
105.45 mM Sodium Chloride
5.53 mM Sodium Phosphate, Monobasic Dihydrate
8.57 mM Sodium Phosphate, Dibasic Dihydrate
1.02 mM Sodium Citrate, Dihydrate 6.19 mM Citric Acid, Monohydrate
65.87 mM Mannitol
0.1 % Polysorbate-80
pH 5.20 (adjust with sodium hydroxide as needed)
Q..S. with Sterile water for injection
ONS-3010 Acetate LS/H M Formulation Buffer Components:
26.35 mM Sodium Chloride
1.00 mM Sodium Acetate Trihydrate
19.00 mM Glacial Acetic Acid
203.00 mM Mannitol
0.1% Polysorbate-80
pH 5.20 (adjust with sodium hydroxide as needed)
Q..S. with Sterile Water for injection
Table 50. Comparison Adalimumab Reference and ONS-3010 Formulation Compositions
Figure imgf000057_0001
Polysorbate 80 0.8 mg 0.8 mg
PH 5.2 5.2
The invention is not limited to the embodiments described and exemplified above, but is capable of variation and modification within the scope of the appended claims.

Claims

We claim:
1. A buffered antibody formulation, comprising an antibody comprising a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising from about 0.7 mM to about 1.3 mM of an acetate salt, from about 200 mM to about 206 mM of mannitol, from about 16 mM to about 22 mM of glacial acetic acid, and from about 24 mM to about 28 mM of sodium chloride, and about 0.07% (v/v) to about 0.15% (v/v) of polysorbate 80, wherein the antibody formulation has a pH of from about 5.1 to about 5.3.
2. The buffered antibody formulation of claim 1, wherein the formulation comprises from about 30 mg to about 50 mg of the antibody.
3. The buffered antibody formulation of claim 1 or 2, wherein the formulation comprises from about 35 mg to about 45 mg of the antibody.
4. The buffered antibody formulation of any of claims 1-3, wherein the formulation comprises from about 37 mg to about 43 mg of the antibody.
5. The buffered antibody formulation of any of claims 1-4, wherein the formulation comprises about 40 mg of the antibody.
6. The buffered antibody formulation of any of claims 1-5, wherein the buffer comprises from about 0.8 mM to about 1.2 mM of the acetate salt.
7. The buffered antibody formulation of any of claims 1-6, wherein the buffer comprises from about 0.9 mM to about 1.1 mM of the acetate salt.
8. The buffered antibody formulation of any of claims 1-7, wherein the buffer comprises about 1 mM of the acetate salt.
9. The buffered antibody formulation of any of claims 1-8, wherein the buffer comprises from about 201 mM to about 205 mM of mannitol.
10. The buffered antibody formulation of any of claims 1-9, wherein the buffer comprises from about 202 mM to about 204 mM of mannitol.
11. The buffered antibody formulation of any of claims 1-10, wherein the buffer comprises about 203 mM of mannitol.
12. The buffered antibody formulation of any of claims 1-11, wherein the buffer comprises from about 17 mM to about 21 mM of glacial acetic acid.
13. The buffered antibody formulation of any of claims 1-12, wherein the buffer comprises from about 18 mM to about 20 mM of glacial acetic acid.
14. The buffered antibody formulation of any of claims 1-13, wherein the buffer comprises about 19 mM of glacial acetic acid.
15. The buffered antibody formulation of any of claims 1-14, wherein the buffer comprises from about 25 mM to about 27 mM of sodium chloride.
16. The buffered antibody formulation of any of claims 1-15, wherein the buffer comprises about 26 mM of sodium chloride.
17. The buffered antibody formulation of any of claims 1-16, wherein the buffer comprises about 26.35 mM of sodium chloride.
18. The buffered antibody formulation of any of claims 1-17, wherein the formulation comprises from about 0.08% (v/v) to about 0.12% (v/v) of polysorbate 80.
19. The buffered antibody formulation of any of claims 1-18, wherein the formulation comprises from about 0.09% (v/v) to about 0.11% (v/v) of polysorbate 80.
20. The buffered antibody formulation of any of claims 1-19, wherein the formulation comprises about 0.1% (v/v) of polysorbate 80.
21. The buffered antibody formulation of any of claims 1-19, wherein the formulation has a pH of about 5.2.
22. A buffered antibody formulation, comprising an antibody comprising a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.8 mM to about 1.2 mM of an acetate salt, about 201 mM to about 205 mM of mannitol, about 17 mM to about 21 mM of glacial acetic acid, and about 25 mM to about 27 mM of sodium chloride, and about 0.08% to about 0.15% (by volume) of polysorbate 80, wherein the antibody formulation has a pH of from about 5.1 to about 5.3.
23. The buffered antibody formulation of claim 22, wherein the formulation comprises from about 30 mg to about 50 mg of the antibody.
24. The buffered antibody formulation of claim 22 or 23, wherein the formulation comprises from about 35 mg to about 45 mg of the antibody.
25. The buffered antibody formulation of any of claims 22-24, wherein the formulation comprises from about 37 mg to about 43 mg of the antibody.
26. The buffered antibody formulation of any of claims 22-25, wherein the formulation comprises about 40 mg of the antibody.
27. The buffered antibody formulation of any of claims 22-26, wherein the buffer comprises from about 0.9 mM to about 1.1 mM of the acetate salt.
28. The buffered antibody formulation of any of claims 22-27, wherein the buffer comprises about 1 mM of the acetate salt.
29. The buffered antibody formulation of any of claims 22-28, wherein the buffer comprises from about 202 mM to about 204 mM of mannitol.
30. The buffered antibody formulation of any of claims 22-29, wherein the buffer comprises about 203 mM of mannitol.
31. The buffered antibody formulation of any of claims 22-30, wherein the buffer comprises from about 18 mM to about 20 mM of glacial acetic acid.
32. The buffered antibody formulation of any of claims 22-31, wherein the buffer comprises about 19 mM of glacial acetic acid.
33. The buffered antibody formulation of any of claims 22-32, wherein the buffer comprises about 26 mM of sodium chloride.
34. The buffered antibody formulation of any of claims 22-32, wherein the buffer comprises about 27 mM of sodium chloride.
35. The buffered antibody formulation of any of claims 22-32, wherein the buffer comprises about 26.35 mM of sodium chloride.
36. The buffered antibody formulation of any of claims 22-35, wherein the formulation comprises from about 0.09% (v/v) to about 0.11% (v/v) of polysorbate 80.
37. The buffered antibody formulation of any of claims 22-36, wherein the formulation comprises about 0.1% (v/v) of polysorbate 80.
38. The buffered antibody formulation of any of claims 22-37, wherein the formulation has a pH of about 5.2.
39. A buffered antibody formulation, comprising an antibody comprising a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 0.9 mM to about 1.1 mM of an acetate salt, about 202 mM to about 204 mM of mannitol, about 18 mM to about 20 mM of glacial acetic acid, and about 25.35 mM to about 26.35 mM of sodium chloride, and about
0.09% to about 0.11% (by volume) of polysorbate 80, wherein the antibody formulation has a pH of from about 5.1 to about 5.3.
40. The buffered antibody formulation of claim 39, wherein the formulation comprises from about 30 mg to about 50 mg of the antibody.
41. The buffered antibody formulation of claim 39 or 40, wherein the formulation comprises from about 35 mg to about 45 mg of the antibody.
42. The buffered antibody formulation of any of claims 39-41, wherein the formulation comprises from about 37 mg to about 43 mg of the antibody.
43. The buffered antibody formulation of any of claims 39-42, wherein the formulation comprises about 40 mg of the antibody.
44. The buffered antibody formulation of any of claims 39-43, wherein the buffer comprises about 1 mM of the acetate salt.
45. The buffered antibody formulation of any of claims 39-44, wherein the buffer comprises about 203 mM of mannitol.
46. The buffered antibody formulation of any of claims 39-45, wherein the buffer comprises about 19 mM of glacial acetic acid.
47. The buffered antibody formulation of any of claims 39-46, wherein the buffer comprises about 26 mM of sodium chloride.
48. The buffered antibody formulation of any of claims 39-46, wherein the buffer comprises about 26.35 mM of sodium chloride.
49. The buffered antibody formulation of any of claims 39-48, wherein the formulation comprises about 0.1% (v/v) of polysorbate 80.
50. The buffered antibody formulation of any of claims 39-49, wherein the formulation has a pH of about 5.2.
51. A buffered antibody formulation, comprising an antibody comprising a heavy chain comprising the amino acid sequence of SEQ. ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO: 2, a buffer comprising about 1 mM of sodium acetate trihydrate, about 203 mM of mannitol, about 19 mM of glacial acetic acid, and about 26.35 mM of sodium chloride, and about 0.1% (by volume) of polysorbate 80, wherein the antibody formulation has a pH of about 5.2.
52. The buffered antibody formulation of claim 51, wherein the formulation comprises from about 30 mg to about 50 mg of the antibody.
53. The buffered antibody formulation of claim 51 or 52, wherein the formulation comprises from about 35 mg to about 45 mg of the antibody.
54. The buffered antibody formulation of any of claims 51-53, wherein the formulation comprises from about 37 mg to about 43 mg of the antibody.
55. The buffered antibody formulation of any of claims 51-54, wherein the formulation comprises about 40 mg of the antibody.
56. A method for treating Rheumatoid Arthritis, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95-97 in an amount effective to treat Rheumatoid Arthritis.
57. The method of claim 56, wherein the subject is a human being.
58. The method of claim 56 or 57, wherein the administering step comprises subcutaneously injecting the antibody formulation.
59. A method for treating Juvenile Idiopathic Arthritis, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95- 97 in an amount effective to treat Juvenile Idiopathic Arthritis.
60. The method of claim 59, wherein the subject is a human being.
61. The method of claim 59 or 60, wherein the administering step comprises subcutaneously injecting the antibody formulation.
62. A method for treating Psoriatic Arthritis, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95-97 in an amount effective to treat Psoriatic Arthritis.
63. The method of claim 62, wherein the subject is a human being.
64. The method of claim 62 or 63, wherein the administering step comprises subcutaneously injecting the antibody formulation.
65. A method for treating Ankylosing Spondylitis, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95-97 in an amount effective to treat Ankylosing Spondylitis.
66. The method of claim 65, wherein the subject is a human being.
67. The method of claim 65 or 66, wherein the administering step comprises
subcutaneously injecting the antibody formulation.
68. A method for treating Crohn's Disease, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95-97 in an amount effective to treat Crohn's Disease.
69. The method of claim 68, wherein the subject is a human being.
70. The method of claim 68 or 69, wherein the administering step comprises
subcutaneously injecting the antibody formulation.
71. A method for treating Plaque Psoriasis, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95-97 in an amount effective to treat Plaque Psoriasis.
72. The method of claim 71, wherein the subject is a human being.
73. The method of claim 71 or 72, wherein the administering step comprises
subcutaneously injecting the antibody formulation.
74. A method for treating Ulcerative Colitis, comprising administering to a subject in need thereof the buffered antibody formulation of any one of claims 1-55 or 95-97 in an amount effective to treat Ulcerative Colitis.
75. The method of claim 74, wherein the subject is a human being.
76. The method of claim 74 or 75, wherein the administering step comprises
subcutaneously injecting the antibody formulation.
77. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Rheumatoid Arthritis.
78. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Juvenile Idiopathic Arthritis.
79. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Psoriatic Arthritis.
80. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Ankylosing Spondylitis.
81. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Crohn's Disease.
82. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Plaque Psoriasis.
83. The buffered antibody formulation of any of claims 1-55 or 95-97 for use in the treatment of Ulcerative Colitis.
84. The buffered antibody formulation of any of claims 1-55 or 95-97 for use as a medicament.
85. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Rheumatoid Arthritis.
86. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Juvenile Idiopathic Arthritis.
87. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Psoriatic Arthritis.
88. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Ankylosing Spondylitis.
89. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Crohn's Disease.
90. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Plaque Psoriasis.
91. A kit, comprising the buffered antibody formulation of any of claims 1-55 or 95-97 and instructions for using the antibody formulation in a method for treating Ulcerative Colitis.
92. The kit of any of claims 85-91, further comprising a device for injecting the antibody formulation into a subject.
93. The kit of claim 92, wherein the device comprises a syringe and a needle.
94. The kit of claim 92, wherein the device comprises a catheter.
95. The buffered antibody formulation of any of claims 1-21, wherein the acetate salt comprises sodium acetate trihydrate.
96. The buffered antibody formulation of any of claims 22-38, wherein the acetate salt comprises sodium acetate trihydrate.
97. The buffered antibody formulation of any of claims 39-50, wherein the acetate salt comprises sodium acetate trihydrate.
98. A buffer formulation for antibody storage, comprising a buffer comprising from about 0.7 mM to about 1.3 mM of an acetate salt, from about 200 mM to about 206 mM of mannitol, from about 16 mM to about 22 mM of glacial acetic acid, and from about 24 mM to about 28 mM of sodium chloride, and about 0.07% (v/v) to about 0.15% (v/v) of polysorbate 80, wherein the formulation has a pH of from about 5.1 to about 5.3.
99. The buffer formulation of claim 98, wherein the buffer comprises from about 0.8 mM to about 1.2 mM of the acetate salt.
100. The buffer formulation of claim 98 or 99, wherein the buffer comprises from about 0.9 mM to about 1.1 mM of the acetate salt.
101. The buffer formulation of any of claims 98-100, wherein the buffer comprises about 1 mM of the acetate salt.
102. The buffer formulation of any of claims 98-101, wherein the buffer comprises from about 201 mM to about 205 mM of mannitol.
103. The buffer formulation of any of claims 98-102, wherein the buffer comprises from about 202 mM to about 204 mM of mannitol.
104. The buffer formulation of any of claims 98-103, wherein the buffer comprises about 203 mM of mannitol.
105. The buffer formulation of any of claims 98-104, wherein the buffer comprises from about 17 mM to about 21 mM of glacial acetic acid.
106. The buffer formulation of any of claims 98-105, wherein the buffer formulation comprises from about 18 mM to about 20 mM of glacial acetic acid.
107. The buffer formulation of any of claims 98-106, wherein the buffer comprises about 19 mM of glacial acetic acid.
108. The buffer formulation of any of claims 98-107, wherein the buffer comprises from about 25 mM to about 27 mM of sodium chloride.
109. The buffer formulation of any of claims 98-108, wherein the buffer comprises about 26 mM of sodium chloride.
110. The buffer formulation of any of claims 98-109, wherein the buffer comprises about 26.35 mM of sodium chloride.
111. The buffer formulation of any of claims 98-110, wherein the formulation comprises from about 0.08% (v/v) to about 0.12% (v/v) of polysorbate 80.
112. The buffer formulation of any of claims 98-111, wherein the formulation comprises from about 0.09% (v/v) to about 0.11% (v/v) of polysorbate 80.
113. The buffer formulation of any of claims 98-112, wherein the formulation comprises about 0.1% (v/v) of polysorbate 80.
114. The buffer formulation of any of claims 98-113, wherein the formulation has a pH of about 5.2.
115. The buffer formulation of any of claims 98-114, wherein the acetate salt comprises sodium acetate trihydrate.
116. The buffer formulation of any of claims 98-115, further comprising an antibody.
PCT/US2014/060810 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability WO2015057910A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2016549199A JP6526025B2 (en) 2013-10-16 2014-10-16 Buffer solution formulation for enhancing antibody stability
EP14854535.3A EP3057616B1 (en) 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability
AU2014337263A AU2014337263B2 (en) 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability
CA2926588A CA2926588C (en) 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability
MX2016004926A MX2016004926A (en) 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability.
CN201480068235.2A CN106170298B (en) 2013-10-16 2014-10-16 Buffer formulations for improving antibody stability
US15/025,088 US10376582B2 (en) 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361891485P 2013-10-16 2013-10-16
US61/891,485 2013-10-16

Publications (1)

Publication Number Publication Date
WO2015057910A1 true WO2015057910A1 (en) 2015-04-23

Family

ID=52828678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/060810 WO2015057910A1 (en) 2013-10-16 2014-10-16 Buffer formulations for enhanced antibody stability

Country Status (8)

Country Link
US (1) US10376582B2 (en)
EP (1) EP3057616B1 (en)
JP (2) JP6526025B2 (en)
CN (1) CN106170298B (en)
AU (1) AU2014337263B2 (en)
CA (1) CA2926588C (en)
MX (1) MX2016004926A (en)
WO (1) WO2015057910A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017097407A1 (en) * 2015-12-07 2017-06-15 Merck Patent Gmbh Aqueous pharmaceutical formulation comprising anti-pd-1 antibody avelumab
CN107537036A (en) * 2016-06-27 2018-01-05 江苏泰康生物医药有限公司 A kind of pharmaceutical preparation of human vessel endothelium growth factor resisting monoclonal antibody and its application
US10376582B2 (en) 2013-10-16 2019-08-13 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US11951207B2 (en) 2016-06-30 2024-04-09 Celltrion Inc. Stable liquid pharmaceutical preparation

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8946395B1 (en) * 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US20100278822A1 (en) 2009-05-04 2010-11-04 Abbott Biotechnology, Ltd. Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US20110059079A1 (en) * 2009-09-04 2011-03-10 Xoma Technology Ltd. Antibody Coformulations
US20110076273A1 (en) * 2009-09-11 2011-03-31 Genentech, Inc. Highly Concentrated Pharmaceutical Formulations
US20110312000A1 (en) * 2006-09-11 2011-12-22 Immuno-Biological Laboratories Co., Ltd. Monoclonal antibody and use thereof
US20130243764A1 (en) 2011-07-19 2013-09-19 Jonathan Henry Ellis Antigen-binding proteins with increased fcrn binding
WO2013186230A1 (en) * 2012-06-12 2013-12-19 Boehringer Ingelheim International Gmbh Pharmaceutical formulation for a therapeutic antibody

Family Cites Families (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5955953A (en) * 1982-09-22 1984-03-31 大成道路株式会社 Synthetic paving body and method
EP0569525A1 (en) 1991-02-01 1993-11-18 Coulter Corporation METHOD OF PRODUCING F(ab') 2? FRAGMENTS OF IMMUNOGLOBULINS
SI9720020B (en) 1996-02-09 2001-12-31 Basf Ag Human antibodies that bind human TNF alpha
CA2868614A1 (en) 2001-06-08 2002-12-08 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
NZ555692A (en) 2002-07-19 2009-02-28 Abbott Biotech Ltd Treatment of TNF alpha related disorders
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
MY150740A (en) 2002-10-24 2014-02-28 Abbvie Biotechnology Ltd Low dose methods for treating disorders in which tnf? activity is detrimental
TW201705980A (en) 2004-04-09 2017-02-16 艾伯維生物技術有限責任公司 Multiple-variable dose regimen for treating TNF[alpha]-related disorders
WO2006096490A2 (en) 2005-03-08 2006-09-14 Pharmacia & Upjohn Company Llc ANTI-MAdCAM ANTIBODY COMPOSITIONS
CN101500607B (en) 2005-05-16 2013-11-27 阿布维生物技术有限公司 Use of TNFalpha inhibitor for treatment of erosive polyarthritis
NZ563580A (en) 2005-06-07 2010-09-30 Esbatech An Alcon Biomedical R Stable and soluble antibodies inhibiting TNFaplha
US20070041905A1 (en) 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
PE20070796A1 (en) 2005-10-24 2007-08-15 Wyeth Corp PROTEIN PRODUCTION METHOD USING ANTI-SENESCENCE COMPOUNDS
TW201337266A (en) 2005-11-01 2013-09-16 Abbott Biotech Ltd Methods and compositions for diagnosing ankylosing spondylitis using biomarkers
SG170837A1 (en) 2006-04-05 2011-05-30 Abbott Biotech Ltd Antibody purification
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
WO2007120626A2 (en) 2006-04-10 2007-10-25 Abbott Biotechnology Ltd. Uses and compositions for treatment of ankylosing spondylitis
CA2564435A1 (en) 2006-04-10 2007-10-10 Abbott Biotechnology Ltd. Methods for monitoring and treating intestinal disorders
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
US20080118496A1 (en) 2006-04-10 2008-05-22 Medich John R Uses and compositions for treatment of juvenile rheumatoid arthritis
EP2666478A3 (en) 2006-04-10 2014-10-22 AbbVie Biotechnology Ltd Uses and compositions for treatment of psoriasis
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
US20100021451A1 (en) 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20080311043A1 (en) 2006-06-08 2008-12-18 Hoffman Rebecca S Uses and compositions for treatment of psoriatic arthritis
TWI527603B (en) 2006-06-30 2016-04-01 艾伯維生物技術有限責任公司 Automatic injection device
AU2007294731B2 (en) 2006-09-13 2014-04-17 Abbvie Inc. Cell culture improvements
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
EP2500415A1 (en) 2006-09-13 2012-09-19 Abbott Laboratories Cell culture improvements
MX2009004351A (en) 2006-10-27 2009-05-12 Abbott Biotech Ltd Crystalline anti-htnfalpha antibodies.
WO2008121324A2 (en) 2007-03-30 2008-10-09 Abbott Laboratories Recombinant expression vector elements (reves) for enhancing expression of recombinant proteins in host cells
EP2165194A4 (en) 2007-05-31 2010-09-08 Abbott Lab BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNF-alpha INHIBITORS IN AUTOIMMUNE DISORDERS
EP2171451A4 (en) 2007-06-11 2011-12-07 Abbott Biotech Ltd Methods for treating juvenile idiopathic arthritis
SG183709A1 (en) 2007-08-08 2012-09-27 Abbott Lab Compositions and methods for crystallizing antibodies
EP2205280B1 (en) 2007-09-27 2019-09-04 Amgen Inc. Pharmaceutical formulations
NZ709704A (en) 2007-11-30 2017-03-31 Abbvie Biotechnology Ltd Protein formulations and methods of making same
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
WO2009073805A2 (en) * 2007-12-04 2009-06-11 Verdezyne, Inc. Aglycosylated therapeutic antibodies and therapeutic antibody-encoding nucleotide sequences
SG2013054218A (en) 2008-01-15 2014-10-30 Abbott Gmbh & Co Kg Powdered protein compositions and methods of making same
DE602008000796D1 (en) 2008-01-15 2010-04-22 Univ Utrecht Holding Bv Method for determining the amino acid sequence of peptides
KR20100113112A (en) 2008-01-15 2010-10-20 아보트 러보러터리즈 Improved mammalian expression vectors and uses thereof
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
MX2010010503A (en) 2008-03-24 2010-11-09 Abbott Biotech Ltd Methods and compositions for treating bone loss.
NZ592097A (en) 2008-10-20 2013-01-25 Abbott Lab Viral inactivation during purification of il-12 and il-18 antibodies
US20100111853A1 (en) 2008-10-20 2010-05-06 Abbott Laboratories, Inc. Antibodies that bind to il-12 and methods of purifying the same
CA2911256A1 (en) 2008-10-20 2010-12-09 Robert K. Hickman Isolation and purification of antibodies using protein a affinity chromatography
CN102271707B (en) 2008-10-29 2015-04-08 阿布林克斯公司 Formulations of single domain antigen binding molecules
CN102301235B (en) * 2008-11-28 2014-11-19 Abbvie公司 Stable Antibody Compositions And Methods For Stabilizing Same
US20120277165A1 (en) 2009-06-05 2012-11-01 Collins Brian E Methods of modulating fucosylation of glycoproteins
EP3708190A1 (en) * 2010-02-26 2020-09-16 Novo Nordisk A/S Stable antibody containing compositions
US9072668B2 (en) 2010-03-09 2015-07-07 Janssen Biotech, Inc. Non-aqueous high concentration reduced viscosity suspension formulations of antibodies
MX354845B (en) 2010-05-18 2018-03-22 Abbvie Inc Star APPARATUS and PROCESS OF PURIFICATION OF PROTEINS.
CA2799915C (en) 2010-05-25 2023-09-26 Genentech, Inc. Methods of purifying polypeptides
SI2575884T1 (en) 2010-06-03 2018-10-30 Abbvie Biotechnology Ltd Uses and compositions for treatment of hidradenitis suppurativa (hs)
CN105753933A (en) 2010-09-20 2016-07-13 Abbvie 公司 Purification Of Antibodies By Using Simulated Moving Bed Chromatography
US20120258496A1 (en) 2010-09-27 2012-10-11 Boehringer Ingelheim International Gmbh Production of low fucose antibodies in h4-ii-e rat cells
JP6023715B2 (en) 2010-10-11 2016-11-09 アッヴィ・バハマズ・リミテッド Protein purification method
MX344727B (en) 2010-11-11 2017-01-05 Abbvie Biotechnology Ltd IMPROVED HIGH CONCENTRATION ANTI-TNFa ANTIBODY LIQUID FORMULATIONS.
RU2615448C2 (en) 2010-12-28 2017-04-04 Чугаи Сейяку Кабусики Кайся Description of animal cell cultivation method
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2012151199A1 (en) 2011-05-02 2012-11-08 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
KR20140084253A (en) 2011-10-24 2014-07-04 애브비 인코포레이티드 Immunobinders directed against tnf
EP2773439A4 (en) 2011-10-31 2015-07-01 Merck Sharp & Dohme Chromatography process for resolving heterogeneous antibody aggregates
WO2013114164A1 (en) 2012-01-30 2013-08-08 Dr. Reddy's Laboratories Limited Method for obtaining glycoprotein composition with increased afucosylation content
WO2013114165A1 (en) 2012-01-30 2013-08-08 Dr Reddy's Laboratories Limited Process of obtaining glycoprotein composition with increased afucosylation content
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
PE20191815A1 (en) 2012-09-07 2019-12-27 Coherus Biosciences Inc STABLE AQUEOUS FORMULATIONS OF ADALIMUMAB
NO2760138T3 (en) 2012-10-01 2018-08-04
WO2014099636A1 (en) 2012-12-18 2014-06-26 Merck Sharp & Dohme Corp. Liquid formulations for an anti-tnf alpha antibody
CN105007896B (en) 2012-12-28 2019-04-09 雅培心血管系统公司 Therapeutic combination comprising antibody
SG11201507230PA (en) 2013-03-12 2015-10-29 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
WO2014151901A1 (en) 2013-03-14 2014-09-25 Abbvie Inc. Improvement of mammalian cell culture performance through surfactant supplementation of feed media
WO2014158231A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. Low acidic species compositions and methods for producing and using the same
WO2014175164A1 (en) 2013-04-25 2014-10-30 株式会社カネカ Fd chain gene or l chain gene each capable of increasing secretion amount of fab-type antibody
US9416181B2 (en) 2013-05-06 2016-08-16 Abbvie Inc. Compositions for cell culture and methods of using the same
AR096713A1 (en) 2013-06-25 2016-01-27 Cadila Healthcare Ltd PURIFICATION PROCESS FOR MONOCLONAL ANTIBODIES
WO2015051310A2 (en) 2013-10-03 2015-04-09 Bioanalytix, Inc. Mass spectrometry-based method for identifying and maintaining quality control factors during the development and manufacture of a biologic
AU2014337263B2 (en) 2013-10-16 2019-12-12 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
WO2015140700A1 (en) 2014-03-19 2015-09-24 Dr. Reddy's Laboratories Limited Cell culture process
EP2946765B1 (en) 2014-05-23 2016-08-31 Ares Trading S.A. Liquid pharmaceutical composition
HUP1400510A1 (en) 2014-10-28 2016-05-30 Richter Gedeon Nyrt Pharmaceutical anti-tnfalpha antibody formulation
EP3247718B1 (en) 2015-01-21 2021-09-01 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
EP3250598A1 (en) 2015-01-28 2017-12-06 Mabxience S.A. Pharmaceutical formulations for anti-tnf-alpha antibodies
CN105779394B (en) 2015-03-20 2020-03-24 广东东阳光药业有限公司 Cell culture method for reducing acid peak content of antibody and improving glycoform of antibody
CA3010600A1 (en) 2016-01-06 2017-07-13 Oncobiologics, Inc. Reduction of high molecular weight species, acidic charge species, and fragments in a monoclonal antibody composition
CA3010598A1 (en) 2016-01-06 2017-07-13 Oncobiologics, Inc. Modulation of afucosylated species in a monoclonal antibody composition
EP3411401A1 (en) 2016-02-03 2018-12-12 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
CN109073655A (en) 2016-02-04 2018-12-21 安口生物公司 The method of the amino acid sequence of identification and analysis albumen

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US20110312000A1 (en) * 2006-09-11 2011-12-22 Immuno-Biological Laboratories Co., Ltd. Monoclonal antibody and use thereof
US20100278822A1 (en) 2009-05-04 2010-11-04 Abbott Biotechnology, Ltd. Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US20110059079A1 (en) * 2009-09-04 2011-03-10 Xoma Technology Ltd. Antibody Coformulations
US20110076273A1 (en) * 2009-09-11 2011-03-31 Genentech, Inc. Highly Concentrated Pharmaceutical Formulations
US20130243764A1 (en) 2011-07-19 2013-09-19 Jonathan Henry Ellis Antigen-binding proteins with increased fcrn binding
WO2013186230A1 (en) * 2012-06-12 2013-12-19 Boehringer Ingelheim International Gmbh Pharmaceutical formulation for a therapeutic antibody

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3057616A4

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10376582B2 (en) 2013-10-16 2019-08-13 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
WO2017097407A1 (en) * 2015-12-07 2017-06-15 Merck Patent Gmbh Aqueous pharmaceutical formulation comprising anti-pd-1 antibody avelumab
EP3747466A1 (en) * 2015-12-07 2020-12-09 Merck Patent GmbH Aqueous pharmaceutical formulation
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
CN107537036A (en) * 2016-06-27 2018-01-05 江苏泰康生物医药有限公司 A kind of pharmaceutical preparation of human vessel endothelium growth factor resisting monoclonal antibody and its application
US11951207B2 (en) 2016-06-30 2024-04-09 Celltrion Inc. Stable liquid pharmaceutical preparation

Also Published As

Publication number Publication date
AU2014337263B2 (en) 2019-12-12
JP2016535085A (en) 2016-11-10
US10376582B2 (en) 2019-08-13
CA2926588C (en) 2020-07-21
EP3057616B1 (en) 2020-03-11
JP6526025B2 (en) 2019-06-05
EP3057616A1 (en) 2016-08-24
JP2019123718A (en) 2019-07-25
CN106170298B (en) 2024-01-09
US20160235845A1 (en) 2016-08-18
EP3057616A4 (en) 2017-06-14
CN106170298A (en) 2016-11-30
AU2014337263A1 (en) 2016-04-28
MX2016004926A (en) 2017-01-18
CA2926588A1 (en) 2015-04-23

Similar Documents

Publication Publication Date Title
US11285210B2 (en) Buffer formulations for enhanced antibody stability
CA2926588C (en) Buffer formulations for enhanced antibody stability
US10772960B2 (en) Stable aqueous formulations of adalimumab
JP2019056013A (en) Treatment of pediatric growth hormone deficiency with human growth hormone analogue
HUE029849T2 (en) Liquid pharmaceutical composition
AU2021216945A1 (en) Treatment of menorrhagia in patients with severe von Willebrand Disease by administration of recombinant VWF
KR20220008303A (en) How to improve lower urinary tract symptoms

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: 14854535

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2926588

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016549199

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/004926

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2014854535

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014854535

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2014337263

Country of ref document: AU

Date of ref document: 20141016

Kind code of ref document: A