US20230183334A1 - Therapeutic antibody formulation - Google Patents

Therapeutic antibody formulation Download PDF

Info

Publication number
US20230183334A1
US20230183334A1 US18/176,844 US202318176844A US2023183334A1 US 20230183334 A1 US20230183334 A1 US 20230183334A1 US 202318176844 A US202318176844 A US 202318176844A US 2023183334 A1 US2023183334 A1 US 2023183334A1
Authority
US
United States
Prior art keywords
pharmaceutical formulation
amino acid
seq
acid sequence
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/176,844
Inventor
Vincent John Corvari
Karthik Pisupati
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eli Lilly and Co
Original Assignee
Eli Lilly and Co
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 Eli Lilly and Co filed Critical Eli Lilly and Co
Priority to US18/176,844 priority Critical patent/US20230183334A1/en
Assigned to ELI LILLY AND COMPANY reassignment ELI LILLY AND COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CORVARI, VINCENT JOHN, PISUPATI, KARTHIK
Publication of US20230183334A1 publication Critical patent/US20230183334A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0021Intradermal administration, e.g. through microneedle arrays, needleless injectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • 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
    • C07K16/244Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]

Definitions

  • the present invention is in the field of medicine. More particularly, the present invention relates to aqueous pharmaceutical formulations comprising therapeutic antibodies that are suitable for subcutaneous (“SQ”), intramuscular (“IM”), and/or intraperitoneal (“IP”) administration.
  • SQL subcutaneous
  • IM intramuscular
  • IP intraperitoneal
  • Administration of therapeutic antibodies via SQ, IP and/or IM administration is both common and advantageous.
  • Such routes of administration allow the therapeutic antibody to be delivered in a short period of time and allow patients to self-administer therapeutic antibodies without visiting a medical practitioner.
  • formulating therapeutic antibodies into aqueous pharmaceutical formulations suitable for SQ, IM and/or IP administration is both challenging and unpredictable. Additionally, undesirable injection-associated pain, even after a syringe needle is removed, has been reported with such routes of administration and can impair patient compliance with therapy.
  • Aqueous pharmaceutical formulations must provide stability to the therapeutic antibody in solution while, at the same time, maintaining the therapeutic antibody's functional characteristics essential for therapeutic efficacy such as target affinity, selectivity and potency.
  • the aqueous pharmaceutical formulation must also be safe for administration to, and well tolerated by, patients as well as being suitable for manufacturing and storage.
  • Formulating high concentrations of therapeutic antibodies is even more complex. For example, increased rates of antibody degradation, cleavage, clipping, high molecular weight aggregation, dimerization, trimerization, precipitation pH shift, turbidity, solution color change, changes in charge, isomerization, oxidation and/or deamination (all of which affect the therapeutic antibody concentration, functionality and efficacy) have been reported for aqueous formulations of highly concentrated therapeutic antibodies.
  • Another known challenge when formulating high concentrations of therapeutic antibodies is an increase in viscosity which can negatively affect SQ, IM and/or IP administration of an aqueous pharmaceutical formulation. Additionally, injection-associated pain has been reported with formulations having increased viscosity.
  • some therapeutic antibodies such as ixekizumab possess charge distributions leading to high levels of intermolecular interactions (e.g., as may be shown by Dynamic Light Scattering), phase separation, gelation and precipitation, making solubility of the molecule in aqueous solution, especially at high concentrations, very challenging to balance.
  • Charge distribution of such antibodies may also manifest in an isoelectric point preventing formulation at neutral pH.
  • some therapeutic antibodies have a polarity, or dipole moment, such that they are only stable in aqueous formulations within narrow, non-neutral, pH windows. Injection-associated pain has been reported, however, for acidic (e.g., ⁇ pH 6.5) pharmaceutical formulations of therapeutic antibodies.
  • Such therapeutic antibodies such as ixekizumab which possesses an isoelectric point of 8.1 (requiring acidic pH formulation)
  • Ixekizumab is a highly specific anti-IL17A antagonistic antibody, as described, for example, in U.S. Pat. No. 7,838,638.
  • Commercially marketed under the tradename TALTZ® ixekizumab is administered subcutaneously to patients in a highly concentrated (about 80 mg/mL) pharmaceutical formulation having an acidic pH (about 5.7).
  • the commercial pharmaceutical formulation of ixekizumab as described in U.S. Pat. No. 9,376,491, also includes high concentrations of citrate buffer (about 20 mM) and NaCl (about 200 mM).
  • citrate buffer about 20 mM
  • NaCl about 200 mM
  • pharmaceutical formulations having acidic pH and high concentrations of NaCl and/or citrate buffer have been associated with injection-associated pain and patients have reported injection-associated pain after injecting the commercial pharmaceutical formulation of ixekizumab.
  • Injection-associated pain of aqueous pharmaceutical formulations comprising therapeutic antibodies is a complex, multifactorial issue.
  • each individual component, and/or concentration, ratio and characteristic thereof, of an aqueous pharmaceutical formulation can impact injection-associated pain associated with a therapeutic.
  • individual components (and/or concentrations, ratios and characteristics thereof) can impact the stability, functional characteristics, manufacturability and/or tolerability of a formulated therapeutic antibody in an aqueous pharmaceutical formulation.
  • a specific formulation adjustment may provide a beneficial impact to a given aspect of the formulation, the same adjustment may also negatively impact other aspects of the formulation.
  • an aqueous pharmaceutical formulation of therapeutic antibodies suitable for SQ, IM and/or IP administration and which is well tolerated by patients, exhibiting a therapeutically beneficial level of injection-associated pain More particularly, there is a need for such aqueous pharmaceutical formulation for highly concentrated therapeutic antibodies possessing an isoelectric point not compatible with neutral pH in solution, requiring aqueous formulation at an acidic pH. Even more particularly, there is a need for an aqueous pharmaceutical formulation of ixekizumab suitable for SQ, IM and/or IP administration and which is well tolerated by patients, exhibiting an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab (as described in U.S. Pat. No. 9,376,491). Such aqueous pharmaceutical formulation must also provide stability for the therapeutic antibody and preserve the properties of the therapeutic antibody essential for therapeutic efficacy. Such aqueous pharmaceutical formulations must also be amendable to manufacturing, preferably having an extended shelf life.
  • aqueous pharmaceutical formulations provided herein satisfy the aforementioned needs in a surprising and unexpected way. More particularly, the aqueous pharmaceutical formulations provided herein are bufferless aqueous pharmaceutical formulations, suitable for SQ, IM and/or IP administration of high concentrations of ixekizumab, while also preserving the functional characteristics of ixekizumab essential for therapeutic efficacy. Additionally, the aqueous pharmaceutical formulations provided herein are well tolerated by patients, exhibiting an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and providing a therapeutically favorable level of injection-associated pain.
  • the present disclosure provides a bufferless, aqueous pharmaceutical formulation for administering SQ, IM or IP a high concentration of a therapeutic antibody to a patient with a therapeutically favorable level of injection-associated pain
  • the aqueous pharmaceutical formulation comprising a therapeutic antibody at a concentration of greater than 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL or 120 mg/mL; sucrose in a concentration of 234 mM+/ ⁇ 10%; and a surfactant in a concentration between 0.005% w/v+/ ⁇ 10% to 0.05% w/v+/ ⁇ 10%, wherein, the pharmaceutical formulation is an aqueous solution at a pH between 5.2 to 6.5.
  • the surfactant is polysorbate 20 or polysorbate 80. In further specific embodiments, the surfactant is polysorbate 80.
  • the bufferless aqueous pharmaceutical formulation is substantially free of an ionic tonicity excipient. In some embodiments, the pharmaceutical formulation is substantially free of L-amino acid excipients.
  • the antibody possesses an isoelectric point not compatible with neutral pH in solution. In some such embodiments, the antibody possesses an isoelectric point of ⁇ 7.5 and in even further embodiments, the antibody possesses an isoelectric point of ⁇ 8.0.
  • the therapeutic antibody is an anti-IL-17A antibody comprising a LCVR having the amino acid sequence of SEQ ID NO.7 and a HCVR having the amino acid sequence of SEQ ID NO.8.
  • the anti-IL17A antibody comprises a light chain (LC) having the amino acid sequence of SEQ ID NO.9 and a heavy chain (HC) having the amino acid sequence of SEQ ID NO.10.
  • an aqueous pharmaceutical formulation of the present disclosure wherein the aqueous pharmaceutical formulation upon SQ, IP and/or IM administration to a patient exhibits a reduced risk of, and/or a, therapeutically favorable level of injection-associated pain.
  • the anti-ILIA antibody comprises a light chain variable region (LCVR) comprising complementarity determining regions (CDRs) LCDR1, LCDR2, and LCDR3 and a heavy chain variable region (HCVR) comprising CDRs HCDR1, HCDR2, and HCDR3, wherein LCDR1 has the amino acid sequence of SEQ ID NO.1, LCDR2 has the amino acid sequence of SEQ ID NO.2, LCDR3 has the amino acid sequence of SEQ ID NO.3, HCDR1 has the amino acid sequence of SEQ ID NO.4, HCDR2 has the amino acid sequence of SEQ ID NO.5, and HCDR3 has the amino acid sequence of SEQ ID NO.6.
  • LCVR light chain variable region
  • CDRs complementarity determining regions
  • HCVR heavy chain variable region
  • the aqueous pharmaceutical formulation is an aqueous solution at a pH of between 5.2 to 6.5, and comprises the anti-IL17A antibody in a concentration of greater than 60 mg/mL+/ ⁇ 10%,70 mg/mL+/ ⁇ 10%, 80 mg/mL+/ ⁇ 10%, 88 mg/mL+/ ⁇ 10%, 100 mg/mL+/ ⁇ 10%, 120 mg/mL+/ ⁇ 10% or 160 mg/mL+/ ⁇ 10%; sucrose in a concentration of 234 mM+/ ⁇ 10%; and a surfactant in a concentration of 0.005+/ ⁇ 10% to 0.05+/ ⁇ 10% % w/v.
  • the bufferless aqueous pharmaceutical formulation is substantially free of an ionic tonicity excipient. In some embodiments, the pharmaceutical formulation is substantially free of L-amino acid excipients.
  • the surfactant is one of polysorbate 20 or 80. In more specific embodiments, the surfactant is polysorbate 80. In even more specific embodiments, the polysorbate 80 is at a concentration of 0.03% w/v+/ ⁇ 10%. According to such embodiments, the bufferless aqueous pharmaceutical formulation is suitable for SQ, IP and/or IM administration to a patient and exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • the aqueous pharmaceutical formulations provided herein comprise an antibody in a concentration of about 80 mg/mL (e.g., +/ ⁇ 10%); sucrose in a concentration of about 234 mM (e.g., +/ ⁇ 10%); and polysorbate 80 in a concentration of about 0.03% w/v (e.g., +/ ⁇ 10%), and the pharmaceutical formulation is substantially free of an ionic tonicity excipient, substantially free of L-amino acid excipients, and is at a pH of about 5.7 (e.g., +/ ⁇ 10%), and the antibody is an anti-IL17A antibody comprising a LCVR having the amino acid sequence of SEQ ID NO.7 and a HCVR having the amino acid sequence of SEQ ID NO.8.
  • the anti-IL17A antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO. 10 and a light chain having the amino acid sequence of SEQ ID NO. 9.
  • the aqueous pharmaceutical formulation is suitable for SQ, IP and/or IM administration to a patient and exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • a system for subcutaneously delivering an aqueous pharmaceutical formulation to a patient in need of treatment includes a device having a chamber, a drive mechanism operatively coupled to the chamber, and a needle, the chamber being capable of storing a liquid, the needle having a bore in fluid communication with an outlet of the chamber to receive a liquid from the chamber, and the drive mechanism being operative to force the transfer of a liquid from the chamber into the bore of the needle.
  • a pharmaceutical formulation of the present disclosure disposed within the chamber and the inner wall of the chamber having a silicone oil coating at an amount of less than about 0.4 mg.
  • the inner wall of the chamber has a silicone oil coating at an amount of about 0.2 mg or an amount of less than about 0.2 mg.
  • the patient is in need of treatment of RA, Ps, GenPs, Pruritus, AS, PA, PPP, HS or MM.
  • the present disclosure provides a method for reducing injection-associated pain and/or providing a therapeutically favorable level of injection-associated pain experienced by a patient at the time of, or shortly after, SQ, IM and/or IP injection of an aqueous pharmaceutical formulation comprising a therapeutic antibody, the method comprising administering to a patient an aqueous pharmaceutical formulation of the present disclosure.
  • the present disclosure provides a method of delivering a therapeutic antibody to a patient with a therapeutically favorable level of injection-associated pain, wherein the method comprises administering to a patient a pharmaceutical formulation of the present disclosure, wherein the method provides a therapeutically favorable level of injection-associated pain.
  • the present disclosure provides an improved method of delivering a therapeutic antibody to a patient, wherein the improvement comprises a reduction in, and/or providing a therapeutically favorable level of, injection-associated pain with SQ, IM or IP administration of an aqueous pharmaceutical formulation, the method comprising administering to a patient an aqueous pharmaceutical formulation of the present disclosure.
  • the reduction in injection-associated pain comprises a reduction from commercially available formulations and/or providing a therapeutically favorable level of injection-associated pain.
  • a therapeutically favorable level of injection-associated pain may comprise a VAS score of less than 30 mm or a VAS score of less than 20 mm.
  • the present disclosure provides an improved method for administering an anti-IL17A antibody to a patient in need thereof, wherein the improvement comprises a reduction in the level of injection-associated pain upon the administration of a SQ, IM or IP injection of an aqueous pharmaceutical formulation, the method comprising administering to the patient an aqueous pharmaceutical formulation of the present disclosure, wherein said step of administering provides an improved level of injection-associated pain and/or provides a therapeutically favorable level of injection-associated pain.
  • the aqueous pharmaceutical formulation consists essentially of an aqueous pharmaceutical formulation of the present disclosure.
  • the reduction in the level of injection-associated pain comprises providing an improved level of injection-associated pain (for example, a reduction in VAS score compared to the commercial formulation of ixekizumab, i.e., the citrate and NaCl formulation exemplified by the control formulation of Table 2).
  • the method provides a therapeutically favorable level of injection-associated pain comprising a VAS score of less than 30 mm or less than 20 mm.
  • the anti-IL17A antibody is ixekizumab and, according to some such embodiments, the improved level of injection-associated pain comprises a reduction in VAS score compared to the commercial formulation of ixekizumab (the citrate and NaCl formulation exemplified by the control formulation of Table 2).
  • the aqueous pharmaceutical formulation is administered by SQ injection.
  • an improved method of treating at least one of PsO, PsA and AxSpa comprising a reduction in injection-associated pain upon the SQ administration of an aqueous pharmaceutical formulation comprising an anti-IL17A antibody, the method comprising administering an aqueous pharmaceutical formulation of the present disclosure, wherein said step of administering provides an improved level of injection-associated pain and/or provides a therapeutically favorable level of injection-associated pain.
  • a therapeutically favorable level of injection-associated pain is provided comprising a VAS score of less than 30 mm or less than 20 mm.
  • the anti-IL17A antibody is ixekizumab and, according to some such embodiments, the improved level of injection-associated pain comprises a reduction in VAS score compared to the commercial formulation of ixekizumab (the citrate and NaCl formulation exemplified by the control formulation of Table 2).
  • the present disclosure also provides an aqueous pharmaceutical formulation of the present disclosure for use in therapy.
  • the present disclosure provides an aqueous pharmaceutical formulation of the present disclosure for use in the treatment of rheumatoid arthritis (RA), psoriasis (Ps), genital psoriasis (GenPs), pruritus, ankylosing spondylitis (AS), psoriatic arthritis (PA), palmoplantar pustulosis (PPP), Hidradenitis suppurativa (HS) or multiple myeloma (MM).
  • RA rheumatoid arthritis
  • Ps psoriasis
  • GenPs genital psoriasis
  • AS ankylosing spondylitis
  • PA palmoplantar pustulosis
  • HS Hidradenitis suppurativa
  • MM multiple myeloma
  • a use of an aqueous pharmaceutical formulation of the present disclosure for the manufacturer of a medicament for the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM is provided.
  • use of such aqueous pharmaceutical formulations is suitable for SQ, IP and/or IM administration to a patient and exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • the present disclosure provides a method of treating RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM comprising administering to a patient in need thereof an effective amount of an aqueous pharmaceutical formulation of the present disclosure, wherein the aqueous pharmaceutical formulation comprises an anti-IL17A antibody.
  • such method of treating includes administering subcutaneously, to the patient, an initial dose of the aqueous pharmaceutical formulation, on day 0, followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient at every four week interval thereafter, wherein the aqueous pharmaceutical formulation administered to the patient at every four week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
  • such method of treating includes administering subcutaneously, to the patient, an initial dose of the aqueous pharmaceutical formulation, on day 0, followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient at every two week interval thereafter, wherein the aqueous pharmaceutical formulation administered to the patient at every two week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
  • such method of treating includes administering subcutaneously, to the patient, an initial dose of the aqueous pharmaceutical formulation, on day 0, followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient on each of days 14, 28, 42, 56, 70 and 84, and followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient at every four week interval thereafter, wherein the aqueous pharmaceutical formulation, administered to the patient at each of days 14, 28, 42, 56, 70 and 84, and every four week interval thereafter, comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
  • the initial dose of the aqueous pharmaceutical formulation comprises about 160 mg of the anti-IL17A antibody.
  • the about 160 mg initial dose of the aqueous pharmaceutical formulation comprises two doses of the aqueous pharmaceutical formulation, each dose comprising about 80 mg of the anti-IL17A antibody.
  • the aqueous pharmaceutical formulation exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • an aqueous pharmaceutical formulation comprising an anti-IL17A antibody for use in the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM wherein the pharmaceutical formulation is to be administered subcutaneously with an initial dose on day 0, followed by a dose every four weeks interval thereafter, wherein the pharmaceutical formulation to be administered at every four week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
  • compositions disclosed herein comprising an anti-IL17A antibody for use in the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM wherein the pharmaceutical formulation is to be administered subcutaneously with an initial dose on day 0, followed by a dose every two weeks interval thereafter, wherein the pharmaceutical formulation to be administered at every two week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
  • compositions disclosed herein comprising an anti-IL17A antibody for use in the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM wherein the pharmaceutical formulation is to be administered subcutaneously with an initial dose on day 0, followed by a dose on each of days 14, 28, 42, 56, 70 and 84, wherein the pharmaceutical formulation to be administered on each of days 14, 28, 42, 56, 70 and 84 after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
  • the initial dose of the aqueous pharmaceutical formulation comprises about 160 mg of the anti-IL17A antibody.
  • the about 160 mg initial dose of the aqueous pharmaceutical formulation comprises two doses of the aqueous pharmaceutical formulation, each dose comprising about 80 mg of the anti-IL17A antibody.
  • the aqueous pharmaceutical formulations provided herein exhibit an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provide a therapeutically favorable level of injection-associated pain.
  • aqueous pharmaceutical formulation or “pharmaceutical formulation” mean an aqueous solution having at least one therapeutic antibody capable of exerting a biological effect in a human, at least one inactive ingredient (e.g., excipient, surfactant, etc.) which, when combined with the therapeutic antibody, is suitable for therapeutic administration to a human.
  • inactive ingredient e.g., excipient, surfactant, etc.
  • the pharmaceutical formulations provided by the present disclosure are bufferless (i.e., do not comprise agents such as citrate buffer, histidine buffer, acetate buffer, or the like, or combinations thereof, which have acid-base conjugate components, for resisting pH change), aqueous, stable formulations wherein the degree of degradation, modification, aggregation, loss of biological activity and the like, of therapeutic antibodies therein, is acceptably controlled and does not increase unacceptably with time.
  • antibody refers to an immunoglobulin G (IgG) molecule comprising two heavy chains (“HC”) and two light chains (“LC”) inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (“HCVR”) and a heavy chain constant region (“CH”).
  • Each light chain is comprised of a light chain variable region (“LCVR”) and a light chain constant region (“CL”).
  • CDR complementarity determining regions
  • FR framework regions
  • Each HCVR and LCVR is composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of each HC and LC contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • the antibodies are anti-IL17A antibodies.
  • Interleukin 17A, or IL17A refers to cytokines of the IL17 cytokine family (also known as cytotoxic T-lymphocyte-associated antigen 8 (“CTLA8”)).
  • IL17A cytokines exist as homodimeric complexes (e.g., IL17A/A) or as heterodimeric complexes in complex with another IL17 cytokine family member such as IL17F (e.g., IL17A/F).
  • IL17A cytokines are believed to be produced primarily by effector T helper (Th17) cells and have been shown to induce secretion of pro-inflammatory cytokines such as IL-6, IL-8, IL-1 and TNF.
  • Th17 effector T helper
  • the homodimeric complex form of IL17A, IL17A/A has been shown to play a role in diseases such as psoriasis and psoriatic arthritis, both immune-related diseases associated with T cell dysregulation.
  • anti-IL17A antibodies are antibodies that specifically bind and antagonize human IL17A by way of specificity for the A subunit (e.g., the A subunit of IL17A/F or one or both of the A subunits of IL17A/A).
  • LCDR1 comprises the amino acid sequence of SEQ ID NO.1
  • LCDR2 comprises the amino acid sequence of SEQ ID NO.2
  • LCDR3 comprises the amino acid sequence of SEQ ID NO.3
  • HCDR1 comprises the amino acid sequence of SEQ ID NO.4
  • HCDR2 comprises the amino acid sequence of SEQ ID NO.5
  • HCDR3 comprises the amino acid sequence of SEQ ID NO.6.
  • the LCVR comprises the amino acid sequence of SEQ ID NO.7 and the HCVR comprises the amino acid sequence of SEQ ID NO.8.
  • the LC comprises the amino acid sequence of SEQ ID NO.9 and the HC comprises the amino acid sequence of SEQ ID NO.10.
  • An exemplary embodiment of an anti-IL17A antibody is ixekizumab, as described, for example, in U.S. Pat. No. 7,838,638.
  • An additional example of an anti-IL17A antibody is secukinumab (marketed under the tradename)COSENTYX®), as described, for example, in U.S. Pat. No. 7,807,155.
  • the terms “about” or “approximately”, when used in reference to a particular recited numerical value or range of values, means that the value may vary from the recited value by no more than 10% (e.g., +/ ⁇ 10%).
  • the expression “about 100” includes 90 and 110 and all values in between (e.g., 91, 92, 93, 94, etc.).
  • the terms “substantially free of” or “substantially devoid of” mean the presence of a given substance (e.g., ionic tonicity excipient) is below a limitation of detection for an assay used for detecting the presence of such substance.
  • ionic tonicity excipient means an excipient that comprises an ionic compound (e.g., an electrolyte such as sodium chloride, potassium chloride, magnesium chloride, calcium chloride, arginine hydrochloride, or the like), which is distinct from the antibody and surfactant comprising an aqueous pharmaceutical formulation.
  • an ionic tonicity excipient as is known in the field, may be used to adjust the osmotic pressure of a pharmaceutical formulation.
  • L-amino acid excipients refers to L-amino acids which are added as either a part of a buffer (e.g., L-histidine in a histidine buffer; L-arginine in an arginine buffer, etc.) or as an excipient component of an aqueous pharmaceutical formulation (but does not refer to components of the therapeutic antibody).
  • a buffer e.g., L-histidine in a histidine buffer; L-arginine in an arginine buffer, etc.
  • an excipient component of an aqueous pharmaceutical formulation but does not refer to components of the therapeutic antibody.
  • VAS visual analog scale
  • VAS refers to an evaluation tool for assessing injection-associated pain experienced by a patient.
  • VAS consists of a 100 mm contiguous scale, upon which a patient identifies their level of pain following injection.
  • the VAS scoring extremes are “no pain at all” (e.g., 0) and “worst pain imaginable” (e.g., 100).
  • Severity of pain may be categorized, according to the VAS tool, as mild pain ( ⁇ 30 mm); moderate pain (>30 mm- ⁇ 70 mm) and severe pain (>70 mm).
  • injection-associated pain is in reference to acute pain experienced by a patient at the time of, or shortly after, injection of an aqueous pharmaceutical formulation.
  • a desired property of a stable pharmaceutical formulation is being well tolerated by patients, for example, providing a therapeutically favorable level of injection-associated pain (e.g., a VAS score of ⁇ 30 mm and/or ⁇ 20 mm).
  • a therapeutically favorable level of injection-associated pain e.g., a VAS score of ⁇ 30 mm and/or ⁇ 20 mm.
  • the components, and concentrations and/or ratios thereof, of a pharmaceutical formulation may impact injection-associated pain experienced by the patient.
  • treatment and/or “treating” and/or “treat” are intended to refer to all processes wherein there may be a total elimination, slowing or delaying, reduction in severity or frequency (e.g., of flares or episodes), interruption or stopping of the progression of disease and/or symptoms thereof, but does not require a total elimination of all disease symptoms.
  • Treatment includes administration of an aqueous pharmaceutical formulation of the present disclosure for treatment of a disease in a human that would benefit from at least one of the above-listed processes, including: (a) inhibiting further progression of disease symptoms and effects, i.e., arresting its development; (b) relieving the disease, i.e., causing an elimination or regression of disease, disease symptoms or complications thereof; and (c) preventing or reducing the frequency of disease episodes or flares.
  • the pharmaceutical formulations provided herein may be used in the treatment of at least one of RA, Ps, GenPs, AS, PA, PPP, HS or MM.
  • the term “patient,” “subject” and “individual,” refers to a human. Unless otherwise noted, the subject is further characterized as having, being at risk of developing, or experiencing symptoms of a disease that would benefit from administration of a pharmaceutical formulation disclosed herein.
  • an “effective amount” or “therapeutically effective amount” of a pharmaceutical formulation of the instant disclosure refers to an amount necessary (at dosages, frequency of administration and for periods of time for a particular means of administration) to achieve the desired therapeutic result.
  • An effective amount of pharmaceutical formulation of the present disclosure may vary according to factors such as the disease state, age, sex, and weight of the subject and the ability of the pharmaceutical formulation of the present disclosure to elicit a desired response in the subject.
  • An effective amount is also one in which any toxic or detrimental effects of the pharmaceutical formulation of the present disclosure are outweighed by the therapeutically beneficial effects.
  • dose regimens for the treatment of a disease with a pharmaceutical formulation of the present disclosure.
  • dose refers to an amount of a pharmaceutical formulation that is administered to a subject.
  • dose regimen or “dosage regimen”, as generally known in the field and as may be referred to interchangeably herein, includes a treatment schedule for administering a set (i.e., series or sequence) of doses to be administered to a patient over a period of time.
  • a dose regimen of the present disclosure may include an initial dose of an aqueous pharmaceutical formulation (for example, comprising an anti-IL17A antibody) of the present disclosure administered to a patient on the first day of treatment (e.g., Day 0).
  • An initial dose may be referred to herein as a “loading dose”.
  • a dose regimen of the present disclosure may include an initial period of treatment, sometimes referred to herein as an “induction period”, which follows the loading dose.
  • a patient may be administered a dose (or doses) comprising a specific concentration of a therapeutic antibody (e.g., anti-IL17A antibody), at a given frequency of administration (e.g., every day, every 2 weeks, every 4 weeks, etc.), for a given duration of time (e.g., 4, 12 or 16 weeks).
  • dose regimens of the present disclosure may include a period following the induction period, sometimes referred to herein as the “maintenance period”, in which a patient is administered a dose comprising a specific concentration of the therapeutic antibody, at a given frequency of administration (e.g., every 2 or 4 weeks, etc.).
  • aqueous pharmaceutical formulations of the present disclosure may be administered to a patient via parenteral administration.
  • Parenteral administration refers to the injection of a dose into the body by a sterile syringe or some other drug delivery system including an autoinjector or an infusion pump.
  • Exemplary drug delivery systems for use with the aqueous pharmaceutical formulations of the present disclosure are described in the following references, the disclosures of which are expressly incorporated herein by reference in their entirety: U.S. Patent Publication No. 2014/0054883 to Lanigan et al., filed Mar. 7, 2013 and entitled “Infusion Pump Assembly”; U.S. Pat. No. 7,291,132 to DeRuntz et al., filed Feb.
  • anti-IL17A antibody 80 mg/mL PS-80 0.03% w/v (0.3 mg/mL) Sucrose 234 mM (8% w/v) pH 5.7 *The anti-IL17A antibody comprises an HCVR of SEQ ID NO: 8 and an LCVR of SEQ ID NO: 7.
  • the manufacturing process for the anti-IL17A antibody pharmaceutical formulation presented in Table 1 may be accomplished by weighing an appropriate quantity of water (e.g., at a temperature of 20+/ ⁇ 5° C.) into a tared empty vessel of appropriate size. The appropriate quantity of sucrose is added and mixed. Polysorbate 80 is accurately weighed out in a glass container and an appropriate quantity of water at a temperature of 20+/ ⁇ 5° C. is added into the glass container to give the desired concentration and the solution is mixed. The entire content of the polysorbate 80 solution is added to the other excipients. The polysorbate 80 solution container is rinsed with water to ensure the entire contents are transferred. After addition of the polysorbate 80 solution, the solution is mixed.
  • an appropriate quantity of water e.g., at a temperature of 20+/ ⁇ 5° C.
  • Polysorbate 80 is accurately weighed out in a glass container and an appropriate quantity of water at a temperature of 20+/ ⁇ 5° C. is added into the glass container to give the desired concentration and the solution is mixed
  • the pH of the solution is checked to be within 5.7+/ ⁇ 0.3; adjustment with HCl or NaOH solution is done if necessary.
  • the excipient composition is passed through a filter (polyvinylidene fluoride [PVDF]) for bioburden reduction.
  • PVDF polyvinylidene fluoride
  • the anti-1L17A antibody previously expressed in cells, purified, and concentrated, is mixed with an appropriate amount of the formulation excipient solution.
  • the pH of the solution is re-checked to be within 5.7+/ ⁇ 0.3.
  • the pharmaceutical formulation is passed through a PVDF filter for bioburden reduction and may then be stored at 5° C.
  • Both physical and chemical stability is essential for a pharmaceutical formulation of a therapeutic antibody to allow storage and transportation (e.g., 1 year, 18 months, or 2 years) and preserve safety and efficacy.
  • Exemplary evaluations to gauge the physical stability of a pharmaceutical formulation include solubility (phase-separation, gelation) assessments, molecular interactions (e.g., as measured by DLS), visual clarity (i.e., opalescence) characterization by turbidity assessment, and viscosity measurement.
  • chemical stability may be assessed using various analytical methods including size exclusion chromatography (SEC), cation exchange chromatography (CEX) HPLC, reduced and non-reduced capillary electrophoresis (CE-SDS R/NR) and particulate analysis.
  • SEC size exclusion chromatography
  • CEX cation exchange chromatography
  • CE-SDS R/NR reduced and non-reduced capillary electrophoresis
  • the exemplified anti-IL17A antibody pharmaceutical formulation of Table 1 demonstrates chemical and physical stability as well as solubility for the highly concentrated therapeutic antibody, ixekizumab, which possesses an isoelectric point of ⁇ 7.5, not compatible with formulation at neutral pH in solution.
  • aqueous pharmaceutical formulation Sufficiently high solubility is essential for an aqueous pharmaceutical formulation.
  • the aqueous pharmaceutical formulation must maintain the antibody in monomeric state, without high molecular weight (HMW) aggregation, at high concentration.
  • Solubility of an anti-IL17A antibody, having an isoelectric point ⁇ 8.0 (in solution), at high concentrations is analyzed under varying conditions.
  • Samples of each aqueous formulation provided in Table 2 are incubated at each of 5, 0 and ⁇ 5 degrees Celsius (e.g., samples of each formulation may be incubated, in parallel, at 5, 0 and ⁇ 5° C.) for one week. Following incubation samples are assessed for phase separation, gelation, turbidity and viscosity.
  • the exemplified anti-IL17A antibody (comprising two LCVRs having the amino acid sequence of SEQ ID NO: 7 and two HCVRs having the amino acid sequences of SEQ ID NO: 8) has a propensity to phase separate in solution below 0 degrees Celsius (° C.).
  • storage of drug product is at 5° C. and requires stability for periodic refrigeration temperature excursions below 0° C.
  • increasing citrate buffer and NaCl concentrations sufficiently lowers the temperature at which phase separation occurs.
  • Injection-associated pain has been reported to be associated with formulations comprising increased citrate buffer and NaCl concentrations and patients have reported injection-associated pain after injecting the commercial pharmaceutical formulation of ixekizumab.
  • Phase separation of formulations provided in Table 2 is assessed, following incubation at ⁇ 5° C. for one week, by visual monitoring for signs of phase separation (e.g., the formation of a dense, protein rich layer at the bottom of the vial). Results are provided in Table 3.
  • thermodynamic solid phase change e.g., gelation
  • gelation has been observed with the exemplified anti-IL17A antibody at high concentrations at temperatures of 5° C. and below.
  • U.S. Pat. No. 9,376,491 also shows that increasing citrate buffer and NaCl concentration sufficiently avoids gelation at lower temperatures.
  • injection-associated pain has been reported to be associated with formulations comprising increased citrate buffer and NaCl concentrations and patients have reported injection-associated pain after injecting the commercial pharmaceutical formulation of ixekizumab.
  • Turbidity i.e., loss of transparency due to particulate matter suspension
  • Turbidity is an inherent challenge for aqueous pharmaceutical formulations of therapeutic antibodies.
  • the challenge is exasperated at high concentrations of antibodies and at lower temperatures, which can lead to the formulation failing visual inspection.
  • turbidity is assessed (measurements taken at ambient temperature) both visually (e.g., light-based method using purified water as a comparator) and by a nephlometer (HACH Turbidimeter, according to manufacturer instructions) yielding quantitative measurements (NTUs). Lower NTUs are desired; more specifically NTUs values of less than 50 are desired with a failure cut-off at 80 NTUs. Results are provided in Table 3.
  • aqueous pharmaceutical formulation to be acceptable for manufacturing, administration to and tolerability by patients must possess appropriate viscosity. Less viscous (at least ⁇ 20 cP) aqueous solution is required in order to be subcutaneously delivered. Increased concentrations of therapeutic antibody present the challenge of increasing viscosity. It is known that pharmaceutical formulations with NaCl have decreased viscosity, but as noted, increasing NaCl concentration in a pharmaceutical formulation has been associated with injection-associated pain. Viscosity of formulation 1 and the control formulation of Table 2 is assessed following incubation at 20° C., by viscometer (Anton Paar AMVn Viscometer, according to manufacturer instructions) yielding centipoise (cP) measurements. Lower cP being desired, especially for example, ⁇ 20 cP. Results are provided in Table 3.
  • Chemical stability is essential for the development of an aqueous pharmaceutical formulation both for allowing storage (i.e., sufficient shelf-life) and preserving safety and efficacy.
  • Chemical stability comparing the control and formulation 1 (provided in Table 2) is assessed following an incubation period of four weeks at 25° C. or 40° C. in accelerated degradation studies. Change in % HMW aggregate is compared against % HMW aggregate at time 0.
  • HMW high molecular weight
  • SEC size-exclusion chromatography
  • formulation 1 of Table 2 provides unexpected stability comparable to (or improved over) the control formulation of Table 2.
  • a multivariate assessment of physical and chemical stability of formulation 1 of Table 2 is performed as set forth below.
  • formulation 1 of Table 2 is modified to assess physical and chemical stability response of each variable and/or interactions between the variables.
  • Formulation 1 of Table 2 is set as the center point formulation for such experiment.
  • Variant formulations are provided in Table 6.
  • Each variant formulation is assessed for phase separation, gelation and turbidity according to procedures described above. This multivariate assessment provides identification of tolerance limitations for the assessed variables. No phase separation or gelation was observed and acceptable turbidity values were observed.
  • Long-term stability of an aqueous pharmaceutical formulation is required to demonstrate storage capability and sufficient shelf life (e.g., 1 year, 2 years or greater).
  • Long-term stability of the center point formulation of Table 6 (which corresponds to the formulation provided in Table 1 and Formulation 1 of Table 2) is assessed following incubation of samples at: 5° C. for 1, 3 and 6 months; 25° C. for 1 and 3 months; and 35° C. for 1 and 3 months (assessment of sample prior to incubation is also performed).
  • the center point formulation of Table 6 demonstrates long-term stability for the therapeutic antibody, even under stressed conditions of extended periods at high temperatures.
  • a single-dose, subject blinded, randomized, cross-over study is performed in which subjects are randomized into one of two treatment groups.
  • Each treatment group receives subcutaneous injections of the pharmaceutical formulations comprising 80 mg/ml of ixekizumab, as set forth in Table 8, according to the following injection regimens.
  • Treatment group 1 receives a single dose of Formulation B, followed by a single dose of Formulation A seven days later.
  • Treatment group 2 receives a single dose, by SQ injection, of Formulation A followed by a single dose, by SQ injection, of Formulation B fourteen days later.
  • Injections are administered by medical personnel in the abdomen of the subject while the subject is in a sitting or reclining position. Subsequent injections may be alternated between abdominal quadrants.
  • Assessment for injection-associated pain based on VAS scale scoring is performed immediately after each injection (e.g., within 1 min.) and at 10 minutes post injection. Results are provided in Tables 9 and 10 below.
  • Formulation A provides a substantial decrease in VAS score over Formulation B (the commercially available formulation of Taltz®) both immediately after injection and at 10 minutes post-injection.
  • Formulation A provides a substantial improvement in patients experiencing no injection-associated pain immediately post-injection as well as a substantial benefit in the reduction of patients experiencing moderate-to-severe injection-associated pain immediately post-injection over Formulation B (the commercially available formulation of Taltz®).
  • Pharmacokinetic analysis of an aqueous pharmaceutical formulation of ixekizumab may be performed according to a study in which subjects receive a SQ injection of one of Formulation A or B (as provided in Table 8). Subjects are then assessed for pharmacokinetic analysis at various time points (e.g., prior to SQ injection and then post-SQ injection such as 1-24 hrs., 1-90 days post-injection).
  • a single-dose, subject blinded, randomized, parallel design study is performed in which, on day 1, subjects are randomized into one of two treatment groups. Prior to receiving a treatment (e.g., day 1, pre-dose) a pre-dose sample from patients of both treatment groups is taken for pharmacokinetic property assessment. On Day 1, treatment group 1 receives a single, SQ injection of Formulation A and treatment group 2 receives a single, subcutaneous injection of Formulation B (as described in Table 8). Injections may be administered by medical personnel in the abdomen of the subjects.
  • Formulation A demonstrates comparable PK parameters to Formulation B (the commercially available formulation of Taltz®). Also, no severe adverse events were reported for either formulation and overall safety is consistent and comparable to Formulation B.
  • Formulation A demonstrates levels of target neutralization comparable to Formulation B (the commercially available formulation of Taltz®) after extended periods of storage and under stressed conditions.
  • SEQ ID NO: 1 (LCD1 of Exemplary anti-IL17A antibody) RSSRSLVHSRGNTYLH (LCDR2 of Exemplary anti-IL17A antibody) SEQ ID NO: 2 KVSNRFI (LCDR3 of Exemplary anti-IL17A antibody) SEQ ID NO: 3 SQSTHLPFT (HCDR1 of Exemplary anti-IL17A antibody) SEQ ID NO: 4 GYSFTDYHIH (HCDR2 of Exemplary anti-IL17A antibody) SEQ ID NO: 5 VINPMYGTTDYNQRFKG (HCDR3 of Exemplary anti-IL17A antibody) SEQ ID NO: 6 YDYFTGTGVY (LCVR of Exemplary anti-IL17A antibody) SEQ ID NO: 7 DIVMTQTPLSLSVTPGQPASISCRSSRSLVHSRGN TYLHWYLQKPGQSPQLLIYKVSNRFIGVPDRFSGS GSGTDFTLKISRVEAEDVGVYYCSQSTHLPFTFGQ GTKLEI

Abstract

Stable aqueous pharmaceutical formulations for therapeutic antibodies and methods of using such stable aqueous pharmaceutical formulations.

Description

    REFERENCE TO AN ELECTRONIC SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ST.26 XML format and is hereby incorporated by reference in its entirety. Said ST.26 XML copy, created on Feb. 28, 2023, is named X22251_REPLACEMENTSEQUENCELISTING and is 14,222 bytes in size.
  • The present invention is in the field of medicine. More particularly, the present invention relates to aqueous pharmaceutical formulations comprising therapeutic antibodies that are suitable for subcutaneous (“SQ”), intramuscular (“IM”), and/or intraperitoneal (“IP”) administration.
  • Administration of therapeutic antibodies via SQ, IP and/or IM administration is both common and advantageous. Such routes of administration allow the therapeutic antibody to be delivered in a short period of time and allow patients to self-administer therapeutic antibodies without visiting a medical practitioner. However, formulating therapeutic antibodies into aqueous pharmaceutical formulations suitable for SQ, IM and/or IP administration is both challenging and unpredictable. Additionally, undesirable injection-associated pain, even after a syringe needle is removed, has been reported with such routes of administration and can impair patient compliance with therapy.
  • The challenge and unpredictability associated with formulating therapeutic antibodies into aqueous pharmaceutical formulations suitable for SQ, IM and/or IP administration is due, in part, to the numerous properties a pharmaceutical formulation must possess to be therapeutically viable. Aqueous pharmaceutical formulations must provide stability to the therapeutic antibody in solution while, at the same time, maintaining the therapeutic antibody's functional characteristics essential for therapeutic efficacy such as target affinity, selectivity and potency. In addition, the aqueous pharmaceutical formulation must also be safe for administration to, and well tolerated by, patients as well as being suitable for manufacturing and storage.
  • Formulating high concentrations of therapeutic antibodies is even more complex. For example, increased rates of antibody degradation, cleavage, clipping, high molecular weight aggregation, dimerization, trimerization, precipitation pH shift, turbidity, solution color change, changes in charge, isomerization, oxidation and/or deamination (all of which affect the therapeutic antibody concentration, functionality and efficacy) have been reported for aqueous formulations of highly concentrated therapeutic antibodies. Another known challenge when formulating high concentrations of therapeutic antibodies is an increase in viscosity which can negatively affect SQ, IM and/or IP administration of an aqueous pharmaceutical formulation. Additionally, injection-associated pain has been reported with formulations having increased viscosity.
  • Furthermore, some therapeutic antibodies such as ixekizumab possess charge distributions leading to high levels of intermolecular interactions (e.g., as may be shown by Dynamic Light Scattering), phase separation, gelation and precipitation, making solubility of the molecule in aqueous solution, especially at high concentrations, very challenging to balance. Charge distribution of such antibodies may also manifest in an isoelectric point preventing formulation at neutral pH. For example, some therapeutic antibodies have a polarity, or dipole moment, such that they are only stable in aqueous formulations within narrow, non-neutral, pH windows. Injection-associated pain has been reported, however, for acidic (e.g., ≤pH 6.5) pharmaceutical formulations of therapeutic antibodies. Thus, such therapeutic antibodies, such as ixekizumab which possesses an isoelectric point of 8.1 (requiring acidic pH formulation), pose additional, unpredictable challenges for formulating in a way that balances stability of the therapeutic antibody with functional properties required for efficacy, as well as tolerability by patients.
  • Ixekizumab is a highly specific anti-IL17A antagonistic antibody, as described, for example, in U.S. Pat. No. 7,838,638. Commercially marketed under the tradename TALTZ®, ixekizumab is administered subcutaneously to patients in a highly concentrated (about 80 mg/mL) pharmaceutical formulation having an acidic pH (about 5.7). The commercial pharmaceutical formulation of ixekizumab, as described in U.S. Pat. No. 9,376,491, also includes high concentrations of citrate buffer (about 20 mM) and NaCl (about 200 mM). However, pharmaceutical formulations having acidic pH and high concentrations of NaCl and/or citrate buffer have been associated with injection-associated pain and patients have reported injection-associated pain after injecting the commercial pharmaceutical formulation of ixekizumab.
  • Injection-associated pain of aqueous pharmaceutical formulations comprising therapeutic antibodies is a complex, multifactorial issue. For example, each individual component, and/or concentration, ratio and characteristic thereof, of an aqueous pharmaceutical formulation can impact injection-associated pain associated with a therapeutic. Likewise, individual components (and/or concentrations, ratios and characteristics thereof) can impact the stability, functional characteristics, manufacturability and/or tolerability of a formulated therapeutic antibody in an aqueous pharmaceutical formulation. Thus, while a specific formulation adjustment may provide a beneficial impact to a given aspect of the formulation, the same adjustment may also negatively impact other aspects of the formulation. Even further adding to the complexity, a nearly limitless number of different formulation components (e.g., buffers and excipients), as well as concentrations and ratios thereof, have been reported. However, there remains little-to-no correlation for predicting the impact of a specific formulation on the various properties and characteristics of a given therapeutic antibody.
  • Accordingly, there is a need for an aqueous pharmaceutical formulation of therapeutic antibodies suitable for SQ, IM and/or IP administration and which is well tolerated by patients, exhibiting a therapeutically beneficial level of injection-associated pain. More particularly, there is a need for such aqueous pharmaceutical formulation for highly concentrated therapeutic antibodies possessing an isoelectric point not compatible with neutral pH in solution, requiring aqueous formulation at an acidic pH. Even more particularly, there is a need for an aqueous pharmaceutical formulation of ixekizumab suitable for SQ, IM and/or IP administration and which is well tolerated by patients, exhibiting an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab (as described in U.S. Pat. No. 9,376,491). Such aqueous pharmaceutical formulation must also provide stability for the therapeutic antibody and preserve the properties of the therapeutic antibody essential for therapeutic efficacy. Such aqueous pharmaceutical formulations must also be amendable to manufacturing, preferably having an extended shelf life.
  • The aqueous pharmaceutical formulations provided herein satisfy the aforementioned needs in a surprising and unexpected way. More particularly, the aqueous pharmaceutical formulations provided herein are bufferless aqueous pharmaceutical formulations, suitable for SQ, IM and/or IP administration of high concentrations of ixekizumab, while also preserving the functional characteristics of ixekizumab essential for therapeutic efficacy. Additionally, the aqueous pharmaceutical formulations provided herein are well tolerated by patients, exhibiting an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and providing a therapeutically favorable level of injection-associated pain. Accordingly, the present disclosure provides a bufferless, aqueous pharmaceutical formulation for administering SQ, IM or IP a high concentration of a therapeutic antibody to a patient with a therapeutically favorable level of injection-associated pain, the aqueous pharmaceutical formulation comprising a therapeutic antibody at a concentration of greater than 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL or 120 mg/mL; sucrose in a concentration of 234 mM+/−10%; and a surfactant in a concentration between 0.005% w/v+/−10% to 0.05% w/v+/−10%, wherein, the pharmaceutical formulation is an aqueous solution at a pH between 5.2 to 6.5. According to specific embodiments, the surfactant is polysorbate 20 or polysorbate 80. In further specific embodiments, the surfactant is polysorbate 80. According to some embodiments, the bufferless aqueous pharmaceutical formulation is substantially free of an ionic tonicity excipient. In some embodiments, the pharmaceutical formulation is substantially free of L-amino acid excipients. In further embodiments, the antibody possesses an isoelectric point not compatible with neutral pH in solution. In some such embodiments, the antibody possesses an isoelectric point of ≥7.5 and in even further embodiments, the antibody possesses an isoelectric point of ≥8.0. In further, specific embodiments of the aqueous pharmaceutical formulations provided herein, the therapeutic antibody is an anti-IL-17A antibody comprising a LCVR having the amino acid sequence of SEQ ID NO.7 and a HCVR having the amino acid sequence of SEQ ID NO.8. In even further specific embodiments, the anti-IL17A antibody comprises a light chain (LC) having the amino acid sequence of SEQ ID NO.9 and a heavy chain (HC) having the amino acid sequence of SEQ ID NO.10. According to embodiments of the present disclosure, an aqueous pharmaceutical formulation of the present disclosure is provided, wherein the aqueous pharmaceutical formulation upon SQ, IP and/or IM administration to a patient exhibits a reduced risk of, and/or a, therapeutically favorable level of injection-associated pain.
  • According to particular embodiments of the present disclosure, a bufferless aqueous pharmaceutical formulation for an anti-IL7A antibody is provided. In embodiments, the anti-ILIA antibody comprises a light chain variable region (LCVR) comprising complementarity determining regions (CDRs) LCDR1, LCDR2, and LCDR3 and a heavy chain variable region (HCVR) comprising CDRs HCDR1, HCDR2, and HCDR3, wherein LCDR1 has the amino acid sequence of SEQ ID NO.1, LCDR2 has the amino acid sequence of SEQ ID NO.2, LCDR3 has the amino acid sequence of SEQ ID NO.3, HCDR1 has the amino acid sequence of SEQ ID NO.4, HCDR2 has the amino acid sequence of SEQ ID NO.5, and HCDR3 has the amino acid sequence of SEQ ID NO.6. According to such embodiments, the aqueous pharmaceutical formulation is an aqueous solution at a pH of between 5.2 to 6.5, and comprises the anti-IL17A antibody in a concentration of greater than 60 mg/mL+/−10%,70 mg/mL+/−10%, 80 mg/mL+/−10%, 88 mg/mL+/−10%, 100 mg/mL+/−10%, 120 mg/mL+/−10% or 160 mg/mL+/−10%; sucrose in a concentration of 234 mM+/−10%; and a surfactant in a concentration of 0.005+/−10% to 0.05+/−10% % w/v. According to some embodiments, the bufferless aqueous pharmaceutical formulation is substantially free of an ionic tonicity excipient. In some embodiments, the pharmaceutical formulation is substantially free of L-amino acid excipients. In some embodiments, the surfactant is one of polysorbate 20 or 80. In more specific embodiments, the surfactant is polysorbate 80. In even more specific embodiments, the polysorbate 80 is at a concentration of 0.03% w/v+/−10%. According to such embodiments, the bufferless aqueous pharmaceutical formulation is suitable for SQ, IP and/or IM administration to a patient and exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • In particular embodiments, the aqueous pharmaceutical formulations provided herein comprise an antibody in a concentration of about 80 mg/mL (e.g., +/−10%); sucrose in a concentration of about 234 mM (e.g., +/−10%); and polysorbate 80 in a concentration of about 0.03% w/v (e.g., +/−10%), and the pharmaceutical formulation is substantially free of an ionic tonicity excipient, substantially free of L-amino acid excipients, and is at a pH of about 5.7 (e.g., +/−10%), and the antibody is an anti-IL17A antibody comprising a LCVR having the amino acid sequence of SEQ ID NO.7 and a HCVR having the amino acid sequence of SEQ ID NO.8. In further such embodiments, the anti-IL17A antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO. 10 and a light chain having the amino acid sequence of SEQ ID NO. 9. According to such embodiments, the aqueous pharmaceutical formulation is suitable for SQ, IP and/or IM administration to a patient and exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • In further embodiments, a system for subcutaneously delivering an aqueous pharmaceutical formulation to a patient in need of treatment is provided. Such system includes a device having a chamber, a drive mechanism operatively coupled to the chamber, and a needle, the chamber being capable of storing a liquid, the needle having a bore in fluid communication with an outlet of the chamber to receive a liquid from the chamber, and the drive mechanism being operative to force the transfer of a liquid from the chamber into the bore of the needle. Such system also includes a pharmaceutical formulation of the present disclosure disposed within the chamber and the inner wall of the chamber having a silicone oil coating at an amount of less than about 0.4 mg. According to some more specific embodiments, the inner wall of the chamber has a silicone oil coating at an amount of about 0.2 mg or an amount of less than about 0.2 mg. According to some embodiments of the system, the patient is in need of treatment of RA, Ps, GenPs, Pruritus, AS, PA, PPP, HS or MM.
  • In further embodiments, the present disclosure provides a method for reducing injection-associated pain and/or providing a therapeutically favorable level of injection-associated pain experienced by a patient at the time of, or shortly after, SQ, IM and/or IP injection of an aqueous pharmaceutical formulation comprising a therapeutic antibody, the method comprising administering to a patient an aqueous pharmaceutical formulation of the present disclosure. According to embodiments, the present disclosure provides a method of delivering a therapeutic antibody to a patient with a therapeutically favorable level of injection-associated pain, wherein the method comprises administering to a patient a pharmaceutical formulation of the present disclosure, wherein the method provides a therapeutically favorable level of injection-associated pain. According to further embodiments, the present disclosure provides an improved method of delivering a therapeutic antibody to a patient, wherein the improvement comprises a reduction in, and/or providing a therapeutically favorable level of, injection-associated pain with SQ, IM or IP administration of an aqueous pharmaceutical formulation, the method comprising administering to a patient an aqueous pharmaceutical formulation of the present disclosure. According to embodiments, the reduction in injection-associated pain comprises a reduction from commercially available formulations and/or providing a therapeutically favorable level of injection-associated pain. According to embodiments, a therapeutically favorable level of injection-associated pain may comprise a VAS score of less than 30 mm or a VAS score of less than 20 mm.
  • According to embodiments, the present disclosure provides an improved method for administering an anti-IL17A antibody to a patient in need thereof, wherein the improvement comprises a reduction in the level of injection-associated pain upon the administration of a SQ, IM or IP injection of an aqueous pharmaceutical formulation, the method comprising administering to the patient an aqueous pharmaceutical formulation of the present disclosure, wherein said step of administering provides an improved level of injection-associated pain and/or provides a therapeutically favorable level of injection-associated pain. According to some embodiments, the aqueous pharmaceutical formulation consists essentially of an aqueous pharmaceutical formulation of the present disclosure. According to embodiments, the reduction in the level of injection-associated pain comprises providing an improved level of injection-associated pain (for example, a reduction in VAS score compared to the commercial formulation of ixekizumab, i.e., the citrate and NaCl formulation exemplified by the control formulation of Table 2). According to some embodiments, the method provides a therapeutically favorable level of injection-associated pain comprising a VAS score of less than 30 mm or less than 20 mm. According to embodiments, the anti-IL17A antibody is ixekizumab and, according to some such embodiments, the improved level of injection-associated pain comprises a reduction in VAS score compared to the commercial formulation of ixekizumab (the citrate and NaCl formulation exemplified by the control formulation of Table 2). According to some embodiments, the aqueous pharmaceutical formulation is administered by SQ injection.
  • According to further embodiments of the present disclosure, an improved method of treating at least one of PsO, PsA and AxSpa is provided, wherein the improvement comprises a reduction in injection-associated pain upon the SQ administration of an aqueous pharmaceutical formulation comprising an anti-IL17A antibody, the method comprising administering an aqueous pharmaceutical formulation of the present disclosure, wherein said step of administering provides an improved level of injection-associated pain and/or provides a therapeutically favorable level of injection-associated pain. According to some embodiments, a therapeutically favorable level of injection-associated pain is provided comprising a VAS score of less than 30 mm or less than 20 mm. In some more specific embodiments, the anti-IL17A antibody is ixekizumab and, according to some such embodiments, the improved level of injection-associated pain comprises a reduction in VAS score compared to the commercial formulation of ixekizumab (the citrate and NaCl formulation exemplified by the control formulation of Table 2).
  • The present disclosure also provides an aqueous pharmaceutical formulation of the present disclosure for use in therapy. In particular embodiments, the present disclosure provides an aqueous pharmaceutical formulation of the present disclosure for use in the treatment of rheumatoid arthritis (RA), psoriasis (Ps), genital psoriasis (GenPs), pruritus, ankylosing spondylitis (AS), psoriatic arthritis (PA), palmoplantar pustulosis (PPP), Hidradenitis suppurativa (HS) or multiple myeloma (MM). According to further embodiments of the present disclosure, a use of an aqueous pharmaceutical formulation of the present disclosure for the manufacturer of a medicament for the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM is provided. According to such embodiments, use of such aqueous pharmaceutical formulations is suitable for SQ, IP and/or IM administration to a patient and exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • According to particular embodiments, the present disclosure provides a method of treating RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM comprising administering to a patient in need thereof an effective amount of an aqueous pharmaceutical formulation of the present disclosure, wherein the aqueous pharmaceutical formulation comprises an anti-IL17A antibody. In a more particular embodiment, such method of treating includes administering subcutaneously, to the patient, an initial dose of the aqueous pharmaceutical formulation, on day 0, followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient at every four week interval thereafter, wherein the aqueous pharmaceutical formulation administered to the patient at every four week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL. In another particular embodiment, such method of treating includes administering subcutaneously, to the patient, an initial dose of the aqueous pharmaceutical formulation, on day 0, followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient at every two week interval thereafter, wherein the aqueous pharmaceutical formulation administered to the patient at every two week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL. In yet another particular embodiment, such method of treating includes administering subcutaneously, to the patient, an initial dose of the aqueous pharmaceutical formulation, on day 0, followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient on each of days 14, 28, 42, 56, 70 and 84, and followed by administering subcutaneously the aqueous pharmaceutical formulation to the patient at every four week interval thereafter, wherein the aqueous pharmaceutical formulation, administered to the patient at each of days 14, 28, 42, 56, 70 and 84, and every four week interval thereafter, comprises the anti-IL17A antibody at a concentration of about 80 mg/mL. According to some of the methods of treating provided by the instant disclosure, the initial dose of the aqueous pharmaceutical formulation comprises about 160 mg of the anti-IL17A antibody. In some such embodiments, the about 160 mg initial dose of the aqueous pharmaceutical formulation comprises two doses of the aqueous pharmaceutical formulation, each dose comprising about 80 mg of the anti-IL17A antibody. According to such methods, the aqueous pharmaceutical formulation exhibits an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provides a therapeutically favorable level of injection-associated pain.
  • According to particular embodiments, there is provided herein an aqueous pharmaceutical formulation comprising an anti-IL17A antibody for use in the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM wherein the pharmaceutical formulation is to be administered subcutaneously with an initial dose on day 0, followed by a dose every four weeks interval thereafter, wherein the pharmaceutical formulation to be administered at every four week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL. In another particular embodiment, there is provided pharmaceutical formulations disclosed herein comprising an anti-IL17A antibody for use in the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM wherein the pharmaceutical formulation is to be administered subcutaneously with an initial dose on day 0, followed by a dose every two weeks interval thereafter, wherein the pharmaceutical formulation to be administered at every two week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL. In yet another particular embodiment, there is provided pharmaceutical formulations disclosed herein comprising an anti-IL17A antibody for use in the treatment of RA, Ps, GenPs, pruritus, AS, PA, PPP, HS or MM wherein the pharmaceutical formulation is to be administered subcutaneously with an initial dose on day 0, followed by a dose on each of days 14, 28, 42, 56, 70 and 84, wherein the pharmaceutical formulation to be administered on each of days 14, 28, 42, 56, 70 and 84 after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL. According to some embodiments, the initial dose of the aqueous pharmaceutical formulation comprises about 160 mg of the anti-IL17A antibody. In some such embodiments, the about 160 mg initial dose of the aqueous pharmaceutical formulation comprises two doses of the aqueous pharmaceutical formulation, each dose comprising about 80 mg of the anti-IL17A antibody. According to such embodiments, the aqueous pharmaceutical formulations provided herein exhibit an improved level of injection-associated pain over the commercial pharmaceutical formulation of ixekizumab and/or provide a therapeutically favorable level of injection-associated pain.
  • As used interchangeably herein, the expressions “aqueous pharmaceutical formulation” or “pharmaceutical formulation” mean an aqueous solution having at least one therapeutic antibody capable of exerting a biological effect in a human, at least one inactive ingredient (e.g., excipient, surfactant, etc.) which, when combined with the therapeutic antibody, is suitable for therapeutic administration to a human. The pharmaceutical formulations provided by the present disclosure are bufferless (i.e., do not comprise agents such as citrate buffer, histidine buffer, acetate buffer, or the like, or combinations thereof, which have acid-base conjugate components, for resisting pH change), aqueous, stable formulations wherein the degree of degradation, modification, aggregation, loss of biological activity and the like, of therapeutic antibodies therein, is acceptably controlled and does not increase unacceptably with time.
  • As used herein, the term “antibody” refers to an immunoglobulin G (IgG) molecule comprising two heavy chains (“HC”) and two light chains (“LC”) inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (“HCVR”) and a heavy chain constant region (“CH”). Each light chain is comprised of a light chain variable region (“LCVR”) and a light chain constant region (“CL”). Each HCVR and LCVR are further sub-dividable into regions of hypervariability, termed complementarity determining regions (“CDR”), interspersed with regions that are more conserved, termed framework regions (“FR”). Each HCVR and LCVR is composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of each HC and LC contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • According to particular embodiments of aqueous pharmaceutical formulations provided herein, the antibodies are anti-IL17A antibodies. Interleukin 17A, or IL17A, as used herein refers to cytokines of the IL17 cytokine family (also known as cytotoxic T-lymphocyte-associated antigen 8 (“CTLA8”)). IL17A cytokines exist as homodimeric complexes (e.g., IL17A/A) or as heterodimeric complexes in complex with another IL17 cytokine family member such as IL17F (e.g., IL17A/F). IL17A cytokines are believed to be produced primarily by effector T helper (Th17) cells and have been shown to induce secretion of pro-inflammatory cytokines such as IL-6, IL-8, IL-1 and TNF. The homodimeric complex form of IL17A, IL17A/A, has been shown to play a role in diseases such as psoriasis and psoriatic arthritis, both immune-related diseases associated with T cell dysregulation.
  • When referred to herein, such anti-IL17A antibodies are antibodies that specifically bind and antagonize human IL17A by way of specificity for the A subunit (e.g., the A subunit of IL17A/F or one or both of the A subunits of IL17A/A). According to specific embodiments of anti-IL17A antibodies, LCDR1 comprises the amino acid sequence of SEQ ID NO.1, LCDR2 comprises the amino acid sequence of SEQ ID NO.2, LCDR3 comprises the amino acid sequence of SEQ ID NO.3, HCDR1 comprises the amino acid sequence of SEQ ID NO.4, HCDR2 comprises the amino acid sequence of SEQ ID NO.5, and HCDR3 comprises the amino acid sequence of SEQ ID NO.6.
  • According to some such embodiments, the LCVR comprises the amino acid sequence of SEQ ID NO.7 and the HCVR comprises the amino acid sequence of SEQ ID NO.8. In even more specific embodiments of such anti-IL17 antibodies, the LC comprises the amino acid sequence of SEQ ID NO.9 and the HC comprises the amino acid sequence of SEQ ID NO.10. An exemplary embodiment of an anti-IL17A antibody is ixekizumab, as described, for example, in U.S. Pat. No. 7,838,638. An additional example of an anti-IL17A antibody is secukinumab (marketed under the tradename)COSENTYX®), as described, for example, in U.S. Pat. No. 7,807,155.
  • As may be used herein, the terms “about” or “approximately”, when used in reference to a particular recited numerical value or range of values, means that the value may vary from the recited value by no more than 10% (e.g., +/−10%). For example, as used herein, the expression “about 100” includes 90 and 110 and all values in between (e.g., 91, 92, 93, 94, etc.).
  • As referred to herein, the terms “substantially free of” or “substantially devoid of” mean the presence of a given substance (e.g., ionic tonicity excipient) is below a limitation of detection for an assay used for detecting the presence of such substance.
  • The term “ionic tonicity excipient”, as referred to herein, means an excipient that comprises an ionic compound (e.g., an electrolyte such as sodium chloride, potassium chloride, magnesium chloride, calcium chloride, arginine hydrochloride, or the like), which is distinct from the antibody and surfactant comprising an aqueous pharmaceutical formulation. An ionic tonicity excipient, as is known in the field, may be used to adjust the osmotic pressure of a pharmaceutical formulation. However (and as provided in the examples provided herein), adjustment of pH with HCl or NaOH, as necessary, following dissolution and mixing of the aqueous pharmaceutical formulation is not within the meaning of the term ionic tonicity excipient as used herein (as HCl or NaOH, added for pH adjustment are not acting in the formulation as an ionic tonicity excipient).
  • As referred to herein, the term L-amino acid excipients refers to L-amino acids which are added as either a part of a buffer (e.g., L-histidine in a histidine buffer; L-arginine in an arginine buffer, etc.) or as an excipient component of an aqueous pharmaceutical formulation (but does not refer to components of the therapeutic antibody).
  • As referred to interchangeably herein, the “visual analog scale” or “VAS”, refers to an evaluation tool for assessing injection-associated pain experienced by a patient. VAS consists of a 100 mm contiguous scale, upon which a patient identifies their level of pain following injection. The VAS scoring extremes are “no pain at all” (e.g., 0) and “worst pain imaginable” (e.g., 100). Severity of pain may be categorized, according to the VAS tool, as mild pain (≤30 mm); moderate pain (>30 mm-≤70 mm) and severe pain (>70 mm). When referred to herein, “injection-associated pain” is in reference to acute pain experienced by a patient at the time of, or shortly after, injection of an aqueous pharmaceutical formulation. A desired property of a stable pharmaceutical formulation is being well tolerated by patients, for example, providing a therapeutically favorable level of injection-associated pain (e.g., a VAS score of <30 mm and/or <20 mm). As is known, the components, and concentrations and/or ratios thereof, of a pharmaceutical formulation may impact injection-associated pain experienced by the patient.
  • As used interchangeably herein, “treatment” and/or “treating” and/or “treat” are intended to refer to all processes wherein there may be a total elimination, slowing or delaying, reduction in severity or frequency (e.g., of flares or episodes), interruption or stopping of the progression of disease and/or symptoms thereof, but does not require a total elimination of all disease symptoms. Treatment includes administration of an aqueous pharmaceutical formulation of the present disclosure for treatment of a disease in a human that would benefit from at least one of the above-listed processes, including: (a) inhibiting further progression of disease symptoms and effects, i.e., arresting its development; (b) relieving the disease, i.e., causing an elimination or regression of disease, disease symptoms or complications thereof; and (c) preventing or reducing the frequency of disease episodes or flares. According to specific embodiments, the pharmaceutical formulations provided herein may be used in the treatment of at least one of RA, Ps, GenPs, AS, PA, PPP, HS or MM.
  • As used interchangeably herein, the term “patient,” “subject” and “individual,” refers to a human. Unless otherwise noted, the subject is further characterized as having, being at risk of developing, or experiencing symptoms of a disease that would benefit from administration of a pharmaceutical formulation disclosed herein.
  • As used interchangeably herein, an “effective amount” or “therapeutically effective amount” of a pharmaceutical formulation of the instant disclosure refers to an amount necessary (at dosages, frequency of administration and for periods of time for a particular means of administration) to achieve the desired therapeutic result. An effective amount of pharmaceutical formulation of the present disclosure may vary according to factors such as the disease state, age, sex, and weight of the subject and the ability of the pharmaceutical formulation of the present disclosure to elicit a desired response in the subject. An effective amount is also one in which any toxic or detrimental effects of the pharmaceutical formulation of the present disclosure are outweighed by the therapeutically beneficial effects.
  • The instant disclosure also relates to dose regimens for the treatment of a disease with a pharmaceutical formulation of the present disclosure. As referred to herein and as generally known in the art, the term “dose” refers to an amount of a pharmaceutical formulation that is administered to a subject. A “dose regimen” or “dosage regimen”, as generally known in the field and as may be referred to interchangeably herein, includes a treatment schedule for administering a set (i.e., series or sequence) of doses to be administered to a patient over a period of time.
  • By way of example, a dose regimen of the present disclosure may include an initial dose of an aqueous pharmaceutical formulation (for example, comprising an anti-IL17A antibody) of the present disclosure administered to a patient on the first day of treatment (e.g., Day 0). An initial dose may be referred to herein as a “loading dose”. Additionally, a dose regimen of the present disclosure may include an initial period of treatment, sometimes referred to herein as an “induction period”, which follows the loading dose. During an induction period, for example, a patient may be administered a dose (or doses) comprising a specific concentration of a therapeutic antibody (e.g., anti-IL17A antibody), at a given frequency of administration (e.g., every day, every 2 weeks, every 4 weeks, etc.), for a given duration of time (e.g., 4, 12 or 16 weeks). Additionally, dose regimens of the present disclosure may include a period following the induction period, sometimes referred to herein as the “maintenance period”, in which a patient is administered a dose comprising a specific concentration of the therapeutic antibody, at a given frequency of administration (e.g., every 2 or 4 weeks, etc.).
  • The aqueous pharmaceutical formulations of the present disclosure may be administered to a patient via parenteral administration. Parenteral administration, as understood in the medical field, refers to the injection of a dose into the body by a sterile syringe or some other drug delivery system including an autoinjector or an infusion pump. Exemplary drug delivery systems for use with the aqueous pharmaceutical formulations of the present disclosure are described in the following references, the disclosures of which are expressly incorporated herein by reference in their entirety: U.S. Patent Publication No. 2014/0054883 to Lanigan et al., filed Mar. 7, 2013 and entitled “Infusion Pump Assembly”; U.S. Pat. No. 7,291,132 to DeRuntz et al., filed Feb. 3, 2006 and entitled “Medication Dispensing Apparatus with Triple Screw Threads for Mechanical Advantage”; U.S. Pat. No. 7,517,334 to Jacobs et al., filed Sep. 18, 2006 and entitled “Medication Dispensing Apparatus with Spring-Driven Locking Feature Enabled by Administration of Final Dose”; and U.S. Pat. No. 8,734,394 to Adams et al., filed Aug. 24, 2012 and entitled “Automatic Injection Device with Delay Mechanism Including Dual Functioning Biasing Member.” Parenteral routes include IM, SQ and IP routes of administration.
  • EXAMPLES Exemplary Aqueous Pharmaceutical Formulation
  • TABLE 1
    Exemplary Aqueous Pharmaceutical Formulation
    Concentration
    anti-IL17A antibody* 80 mg/mL
    PS-80 0.03% w/v (0.3 mg/mL)
    Sucrose 234 mM (8% w/v)
    pH 5.7
    *The anti-IL17A antibody comprises an HCVR of SEQ ID NO: 8 and an LCVR of SEQ ID NO: 7.
  • The manufacturing process for the anti-IL17A antibody pharmaceutical formulation presented in Table 1 may be accomplished by weighing an appropriate quantity of water (e.g., at a temperature of 20+/−5° C.) into a tared empty vessel of appropriate size. The appropriate quantity of sucrose is added and mixed. Polysorbate 80 is accurately weighed out in a glass container and an appropriate quantity of water at a temperature of 20+/−5° C. is added into the glass container to give the desired concentration and the solution is mixed. The entire content of the polysorbate 80 solution is added to the other excipients. The polysorbate 80 solution container is rinsed with water to ensure the entire contents are transferred. After addition of the polysorbate 80 solution, the solution is mixed. After dissolution and mixing has been completed, the pH of the solution is checked to be within 5.7+/−0.3; adjustment with HCl or NaOH solution is done if necessary. The excipient composition is passed through a filter (polyvinylidene fluoride [PVDF]) for bioburden reduction.
  • The anti-1L17A antibody, previously expressed in cells, purified, and concentrated, is mixed with an appropriate amount of the formulation excipient solution. The pH of the solution is re-checked to be within 5.7+/−0.3. The pharmaceutical formulation is passed through a PVDF filter for bioburden reduction and may then be stored at 5° C.
  • Physical-Chemical Properties
  • Both physical and chemical stability is essential for a pharmaceutical formulation of a therapeutic antibody to allow storage and transportation (e.g., 1 year, 18 months, or 2 years) and preserve safety and efficacy. Exemplary evaluations to gauge the physical stability of a pharmaceutical formulation include solubility (phase-separation, gelation) assessments, molecular interactions (e.g., as measured by DLS), visual clarity (i.e., opalescence) characterization by turbidity assessment, and viscosity measurement. Additionally, chemical stability may be assessed using various analytical methods including size exclusion chromatography (SEC), cation exchange chromatography (CEX) HPLC, reduced and non-reduced capillary electrophoresis (CE-SDS R/NR) and particulate analysis. As demonstrated herein, the exemplified anti-IL17A antibody pharmaceutical formulation of Table 1 demonstrates chemical and physical stability as well as solubility for the highly concentrated therapeutic antibody, ixekizumab, which possesses an isoelectric point of ≥7.5, not compatible with formulation at neutral pH in solution.
  • Solubility Assessments:
  • Sufficiently high solubility is essential for an aqueous pharmaceutical formulation. The aqueous pharmaceutical formulation must maintain the antibody in monomeric state, without high molecular weight (HMW) aggregation, at high concentration. Solubility of an anti-IL17A antibody, having an isoelectric point ≥8.0 (in solution), at high concentrations is analyzed under varying conditions.
  • Samples of each aqueous formulation provided in Table 2 are incubated at each of 5, 0 and −5 degrees Celsius (e.g., samples of each formulation may be incubated, in parallel, at 5, 0 and −5° C.) for one week. Following incubation samples are assessed for phase separation, gelation, turbidity and viscosity.
  • TABLE 2
    Formulations
    Anti-IL17A
    Non-Buffer Antibody*
    Sample ID Buffer Excipients Concentration pH
    Control 20 mM Citrate 200 mM NaCl 80 mg/mL 5.7
    (commercial 0.03% PS-80
    formulation as
    described in
    U.S. Patent No.
    9,376,491)
    1 None 234 mM sucrose 80 mg/mL 5.7
    (formulation of 0.03% PS-80
    Table 1)
    2 10 mM Citrate 274 mM mannitol 80 mg/mL 5.7
    3 10 mM Citrate 274 mM mannitol 80 mg/mL 5.7
    0.03% PS80
    4 10 mM Citrate 234 mM sucrose 80 mg/mL 5.7
    0.03% PS-80
    5  5 mM Citrate 175 mM NaCl 80 mg/mL 5.7
    0.03% PS80
    6 2.69 mM L-histidine 150 mM NaCl 80 mg/mL 6.5
    6.28 mM L-histidine
    hydrochloride
    monohydrate
    7 2.69 mM L-histidine 150 mM NaCl 80 mg/mL 6.5
    6.28 mM L-histidine 0.03% PS80
    hydrochloride
    monohydrate
    8 2.69 mM L-histidine 150 mM NaCl 80 mg/mL 5.7
    6.28 mM L-histidine 0.03% PS80
    hydrochloride
    monohydrate
    9 None 130 mM NaCl 80 mg/mL 5.7
    10  None 100 mM NaCl 80 mg/mL 5.7
    11  None  65 mM NaCl 50 mg/mL 5.7
    12  10 mM Citrate None 80 mg/mL 5.7
    *The anti-IL17A antibody comprises two HCVRs having the amino acid sequence of SEQ ID NO: 8 and two LCVRs having the amino acid sequence of SEQ ID NO: 7.
    **In addition to the tested aqueous pharmaceutical formulations set forth in Table 2, an aqueous pharmaceutical formulation comprising 10 mM acetate buffer, 150 mM NaCl and 80 mg/mL of the anti-IL17A antibody, at pH 5.0, was assessed following incubation, wherein unacceptable levels of antibody clipping were observed by non-reduced CD-SDS.
    ***Additionally, as set forth in U.S. Patent No. 9,376,491, unacceptable cloud point was observed for the anti-IL17A antibody with concentrations below either of 20 mM citrate buffer and 150 mM NaCl.
  • Phase Separation:
  • As detailed in U.S. Pat. No. 9,376,491, the exemplified anti-IL17A antibody (comprising two LCVRs having the amino acid sequence of SEQ ID NO: 7 and two HCVRs having the amino acid sequences of SEQ ID NO: 8) has a propensity to phase separate in solution below 0 degrees Celsius (° C.). However, storage of drug product is at 5° C. and requires stability for periodic refrigeration temperature excursions below 0° C. As provided in U.S. Pat. No. 9,376,491, increasing citrate buffer and NaCl concentrations sufficiently lowers the temperature at which phase separation occurs. Injection-associated pain, however, has been reported to be associated with formulations comprising increased citrate buffer and NaCl concentrations and patients have reported injection-associated pain after injecting the commercial pharmaceutical formulation of ixekizumab.
  • Phase separation of formulations provided in Table 2 is assessed, following incubation at −5° C. for one week, by visual monitoring for signs of phase separation (e.g., the formation of a dense, protein rich layer at the bottom of the vial). Results are provided in Table 3.
  • Gelation:
  • Events such as thermodynamic solid phase change (e.g., gelation) can occur at lower temperatures (5° C. or lower), negatively impacting stability. As detailed in U.S. Pat. No. 9,376,491, gelation has been observed with the exemplified anti-IL17A antibody at high concentrations at temperatures of 5° C. and below. U.S. Pat. No. 9,376,491 also shows that increasing citrate buffer and NaCl concentration sufficiently avoids gelation at lower temperatures. However, as noted, injection-associated pain has been reported to be associated with formulations comprising increased citrate buffer and NaCl concentrations and patients have reported injection-associated pain after injecting the commercial pharmaceutical formulation of ixekizumab.
  • Gelation assessment of formulations provided in Table 2 are provided in Table 3. Briefly, following incubation as described above, each vial is agitated (e.g., inverted and then returned upright) and then visually inspected for solidification or lack of liquid flow.
  • Turbidity:
  • Turbidity (i.e., loss of transparency due to particulate matter suspension) is an inherent challenge for aqueous pharmaceutical formulations of therapeutic antibodies. The challenge is exasperated at high concentrations of antibodies and at lower temperatures, which can lead to the formulation failing visual inspection.
  • Briefly, following incubation as described above, turbidity is assessed (measurements taken at ambient temperature) both visually (e.g., light-based method using purified water as a comparator) and by a nephlometer (HACH Turbidimeter, according to manufacturer instructions) yielding quantitative measurements (NTUs). Lower NTUs are desired; more specifically NTUs values of less than 50 are desired with a failure cut-off at 80 NTUs. Results are provided in Table 3.
  • Viscosity:
  • An aqueous pharmaceutical formulation, to be acceptable for manufacturing, administration to and tolerability by patients must possess appropriate viscosity. Less viscous (at least <20 cP) aqueous solution is required in order to be subcutaneously delivered. Increased concentrations of therapeutic antibody present the challenge of increasing viscosity. It is known that pharmaceutical formulations with NaCl have decreased viscosity, but as noted, increasing NaCl concentration in a pharmaceutical formulation has been associated with injection-associated pain. Viscosity of formulation 1 and the control formulation of Table 2 is assessed following incubation at 20° C., by viscometer (Anton Paar AMVn Viscometer, according to manufacturer instructions) yielding centipoise (cP) measurements. Lower cP being desired, especially for example, <20 cP. Results are provided in Table 3.
  • TABLE 3
    Solubility Assessment of the Formulations of Table 2
    Phase Separation Gelation Turbidity Viscosity
    Sample ID Assessment Assessment (NTUs) (cPs)
    Control No No 63 3
    1 No No 10 5
    2 Yes ND ND ND
    3 Yes ND ND ND
    4 Yes ND ND ND
    5 No No 85 ND
    6 No Yes ND ND
    7 No Yes ND ND
    8 No No 95 ND
    9 Yes ND ND ND
    10 Yes ND ND ND
    11 Yes ND ND ND
    12 Yes ND ND ND
  • As shown in Table 3, unacceptable phase separation or gelation was observed for all formulations lacking at least 150 mM NaCl (as well as the NaCl bufferless formulations), with the exception of formulation 1 which did not demonstrate phase separation. Phase separation results for formulation 1 are comparable to the control formulation (high citrate, high NaCl formulation). Also, unacceptable gelation was observed for formulations comprising histidine buffer and NaCl at pH 6.5. Formulation 1 did not demonstrate gelation and was comparable to the control formulation (high citrate, high NaCl formulation). Additionally, unacceptable turbidity was observed for both formulation 5 (citrate (5 mM), NaCl (175 mM)) and formulation 8 (histidine (9 mM) and NaCl (150 mM)). Formulation 1 demonstrated acceptable levels of turbidity and provided unexpected improved levels of turbidity compared to the control formulation (high citrate, high NaCl formulation). Further, as shown, both formulation 1 and the control formulation exhibit acceptable and comparable viscosity.
  • Chemical Stability:
  • Chemical stability is essential for the development of an aqueous pharmaceutical formulation both for allowing storage (i.e., sufficient shelf-life) and preserving safety and efficacy. Chemical stability comparing the control and formulation 1 (provided in Table 2) is assessed following an incubation period of four weeks at 25° C. or 40° C. in accelerated degradation studies. Change in % HMW aggregate is compared against % HMW aggregate at time 0.
  • In one assessment, the change in high molecular weight (HMW) aggregate in the formulations is assessed using size-exclusion chromatography (SEC) according to standard procedures. Results are provided in Table 4.
  • TABLE 4
    Summary of change in % HMW aggregates measured by SEC
    Change in Change in
    % HMW % HMW
    Formulation # aggregates aggregates
    (of Table 2) 25° C. 40° C.
    Control 0.25 0.05
    1 0.49 0.43
  • As shown, both the control formulation and formulation 1 of Table 2 demonstrate acceptable and comparable chemical stability in accelerated degradation studies.
  • Additional accelerated chemical stability of the control and formulation 1 of Table 2 is studied using Cation Exchange (CEX) HPLC. Briefly, samples are incubated at 25° C. for four weeks. Following incubation, samples are analyzed for increase in total % acid variants (% AV) using CEX HPLC. Increase in total % acid variants (% AV) provides an indicator of degradation of the therapeutic antibody in the aqueous formulation. Results are provided in Table 5.
  • TABLE 5
    Increase in % AV over 4 Weeks at 25° C.
    Formulation #
    (of Table 2) Increase in % AV
    Control 2.0
    1 2.3
  • As shown, both the control and formulation 1 of Table 2 demonstrate acceptable, and comparable, levels of chemical stability in the further accelerated degradation studies.
  • Multivariate Assessment of Formulation 1 of Table 2.
  • As demonstrated herein, formulation 1 of Table 2 provides unexpected stability comparable to (or improved over) the control formulation of Table 2. A multivariate assessment of physical and chemical stability of formulation 1 of Table 2 is performed as set forth below.
  • Briefly, four variables (antibody concentration; pH; sucrose concentration; and PS-80 concentration) of formulation 1 of Table 2 are modified to assess physical and chemical stability response of each variable and/or interactions between the variables. Formulation 1 of Table 2 is set as the center point formulation for such experiment. Variant formulations are provided in Table 6.
  • TABLE 6
    Variant Formulations
    Anti-IL17A*
    Sample ID Sucrose PS-80** Antibody pH
    Center Point 234 mM 0.03% 80 mg/mL 5.7
    (formulation 1 of Table 2)
    13 205 mM 0.05% 72 mg/mL 5.2
    14 205 mM 0.005% 72 mg/mL 6.2
    15 205 mM 0.005% 88 mg/mL 5.2
    16 205 mM 0.05% 88 mg/mL 6.2
    17 263 mM 0.005% 72 mg/mL 5.2
    18 263 mM 0.05% 72 mg/mL 6.2
    19 263 mM 0.05% 88 mg/mL 5.2
    20 263 mM 0.005% 88 mg/mL 6.2
    *The anti-IL17A antibody comprises two HCVRs having the amino acid sequence of SEQ ID NO: 8 and two LCVRs having the amino acid sequence of SEQ ID NO: 7.
    **Polysorbate tolerance for the ranges set forth in Table 6 are confirmed by accelerated freeze-thaw studies.
  • Each variant formulation is assessed for phase separation, gelation and turbidity according to procedures described above. This multivariate assessment provides identification of tolerance limitations for the assessed variables. No phase separation or gelation was observed and acceptable turbidity values were observed.
  • Long-term Stability Assessment
  • Long-term stability of an aqueous pharmaceutical formulation is required to demonstrate storage capability and sufficient shelf life (e.g., 1 year, 2 years or greater). Long-term stability of the center point formulation of Table 6 (which corresponds to the formulation provided in Table 1 and Formulation 1 of Table 2) is assessed following incubation of samples at: 5° C. for 1, 3 and 6 months; 25° C. for 1 and 3 months; and 35° C. for 1 and 3 months (assessment of sample prior to incubation is also performed).
  • Following incubation, samples are analyzed for percent monomer and percent high molecular weight (HMW) aggregate using size-exclusion chromatography (SEC) according to standard procedures. Results are provided in Table 7.
  • TABLE 7
    Long Term Stability Assessment of Center Point Formulation
    Incubation Temp Incubation Period Monomer HMW Aggregate
    (° C.) (months) (%) (%)
    Control NA 98.61 1.27
    (pre-incubation)
    5 1 98.83 1.10
    5 3 98.57 1.39
    5 6 98.61 1.27
    5 12 98.67 1.28
    25 1 98.59 1.32
    25 3 98.01 1.85
    35 1 97.93 1.70
    35 3 95.54 3.30
  • As provided, the center point formulation of Table 6 demonstrates long-term stability for the therapeutic antibody, even under stressed conditions of extended periods at high temperatures.
  • In Vivo Tolerability Study
  • Assessment of injection-associated pain from subcutaneous injection of an aqueous pharmaceutical formulation of ixekizumab, at a high concentration (80 mg/mL), is performed according to a study in which subjects receive a SQ injection of one of Formulation A or B (as provided in Table 8), followed by a SQ injection of the other of Formulation A or B some period of time (e.g., 1, 5, 7, 10, 14, etc., days) later. Subjects are then assessed for injection-associated pain based on the VAS scale scoring at specified time points (e.g., within 1 minute (i.e., immediately after injection), within 10 minutes, within 1 hour, within 4 hours within 1 day) after each injection.
  • TABLE 8
    Ixekizumab Pharmaceutical Formulation
    Formulation A
    (corresponds to center point Formulation B
    formulation of Table 6) (commercial formulation of Taltz ®)
    ixekizumab 80 mg/mL ixekizumab 80 mg/mL
    pH 5.7 pH 5.7
    PS-80 0.3 mM PS-80 0.03% w/v
    sucrose 80 mM NaCl 200 mM
    / / / citrate buffer 20 mM
  • Accordingly, a single-dose, subject blinded, randomized, cross-over study is performed in which subjects are randomized into one of two treatment groups. Each treatment group receives subcutaneous injections of the pharmaceutical formulations comprising 80 mg/ml of ixekizumab, as set forth in Table 8, according to the following injection regimens.
  • Treatment group 1 receives a single dose of Formulation B, followed by a single dose of Formulation A seven days later. Treatment group 2 receives a single dose, by SQ injection, of Formulation A followed by a single dose, by SQ injection, of Formulation B fourteen days later. Injections are administered by medical personnel in the abdomen of the subject while the subject is in a sitting or reclining position. Subsequent injections may be alternated between abdominal quadrants. Assessment for injection-associated pain based on VAS scale scoring is performed immediately after each injection (e.g., within 1 min.) and at 10 minutes post injection. Results are provided in Tables 9 and 10 below.
  • TABLE 9
    Injection-Associated Pain Comparability Data
    VAS Score VAS Score
    Formulation time post-injection (w/in 1 min.) time post-injection (10 mins.)
    A (N = 63) 3.52 0.68
    B (N = 61) 25.21 5.15
  • As shown in Table 9, Formulation A provides a substantial decrease in VAS score over Formulation B (the commercially available formulation of Taltz®) both immediately after injection and at 10 minutes post-injection.
  • TABLE 10
    Patient Tolerability Analysis
    Formulation A Formulation B
    time post-injection time post-injection
    VAS Score (w/in 1 min.) (w/in 1 min.)
    No pain 26 (of 63 patients): 41.3% 5 (of 61 patients): 8.2%
    (VAS = 0)
    Mild Pain 36 (of 63 patients): 57.1% 36 (of 61 patients): 59.0%
    (VAS ≤ 30)
    Moderate-to- 1 (of 63 patients): 1.6% 20 (of 61 patients): 32.8%
    Severe Pain
    (VAS > 30)
  • As shown in Table 10, Formulation A provides a substantial improvement in patients experiencing no injection-associated pain immediately post-injection as well as a substantial benefit in the reduction of patients experiencing moderate-to-severe injection-associated pain immediately post-injection over Formulation B (the commercially available formulation of Taltz®).
  • In Vivo Pharmacokinetic Analysis
  • Pharmacokinetic analysis of an aqueous pharmaceutical formulation of ixekizumab may be performed according to a study in which subjects receive a SQ injection of one of Formulation A or B (as provided in Table 8). Subjects are then assessed for pharmacokinetic analysis at various time points (e.g., prior to SQ injection and then post-SQ injection such as 1-24 hrs., 1-90 days post-injection).
  • Accordingly, a single-dose, subject blinded, randomized, parallel design study is performed in which, on day 1, subjects are randomized into one of two treatment groups. Prior to receiving a treatment (e.g., day 1, pre-dose) a pre-dose sample from patients of both treatment groups is taken for pharmacokinetic property assessment. On Day 1, treatment group 1 receives a single, SQ injection of Formulation A and treatment group 2 receives a single, subcutaneous injection of Formulation B (as described in Table 8). Injections may be administered by medical personnel in the abdomen of the subjects. Post-dosing, samples are taken on study days 3, 5 (±1 day), 8 (±1 day), 11 (±1 day), 15 (±2 days), 22 (±2 days), 29 (±2 days), 43 (±2 days), 57 (±3 days), 71 (±3 days) and 85 (±3 days) to assess pharmacokinetic parameters including Cmax (maximum observed drug concentration), AUC[0-∞] (area under the concentrations versus time curve from time zero to infinity), AUC[0 tlast] (area under the concentrations versus time curve from time zero on study Day 1 to time of last measurable concentration), and Tmax (time of the maximum observed drug concentration). Results are provided in Table 11.
  • TABLE 11
    In Vivo Pharmacokinetic Analysis
    Value
    (geometric least Ratio
    Formulation PK Parameter squares mean) (Form. A / Form B)
    Formulation A (N = 33) AUC[0-∞] 159 1.05
    Formulation B (N = 32) (ug*day/mL) 152
    Formulation A (N = 33) AUC[0-tlast] 153 1.04
    Formulation B (N = 32) (ug*day/mL) 146
    Formulation A (N = 33) Cmax 6.29 1.00
    Formulation B (N = 33) (ug/mL) 6.31
    Formulation A (N = 33) Tmax 4.09 (median of
    Formulation B (N = 33) (days) 3.95 differences)
    0
  • As shown in Table 11, Formulation A demonstrates comparable PK parameters to Formulation B (the commercially available formulation of Taltz®). Also, no severe adverse events were reported for either formulation and overall safety is consistent and comparable to Formulation B.
  • Target Neutralization Assessment
  • Following incubation of samples of Formulation A at 5° C. for 1, 6 and 12 months; 25° C. for 1 month; and 35° C. for 1 month, potency of Formulation A is assessed in comparison to Formulation B (of Table 8) by way of a cell-based bioassay. Briefly, murine osteoblast cell line MC3T3-E1, which endogenously expresses IL-17A receptor and stably expresses firefly luciferase gene, is cultured such that when IL-17A is present transcription of luciferase is induced at levels proportional to IL-17A activity. Previously incubated samples of Formulation A and B are introduced to culture wells of the cell-based bioassay, respectively, and following measurement of luciferase expression, inhibition dose curves are generated. Data is analyzed using a four parameter logistic curve fit. Relative potency is determined by calculating the ratio of the EC50 for Formulation A in comparison to the EC50 of Formulation B (e.g., the reference standard). Results are provided in Table 12.
  • TABLE 12
    Relative Potency Assessment of Formulation
    A (% relative to Formulation B)
    Incubation Incubation Temp.
    Period (° C.)
    (Months) 5° C. 25° C. 35° C.
    1 101% 98% 101%
    6 103% ND ND
    12  98% ND ND
  • As shown in Table 12, Formulation A demonstrates levels of target neutralization comparable to Formulation B (the commercially available formulation of Taltz®) after extended periods of storage and under stressed conditions.
  • Sequences
    SEQ ID NO: 1
    (LCD1 of Exemplary anti-IL17A antibody)
    RSSRSLVHSRGNTYLH
    (LCDR2 of Exemplary anti-IL17A antibody)
    SEQ ID NO: 2
    KVSNRFI
    (LCDR3 of Exemplary anti-IL17A antibody)
    SEQ ID NO: 3
    SQSTHLPFT
    (HCDR1 of Exemplary anti-IL17A antibody)
    SEQ ID NO: 4
    GYSFTDYHIH
    (HCDR2 of Exemplary anti-IL17A antibody)
    SEQ ID NO: 5
    VINPMYGTTDYNQRFKG
    (HCDR3 of Exemplary anti-IL17A antibody)
    SEQ ID NO: 6
    YDYFTGTGVY
    (LCVR of Exemplary anti-IL17A antibody)
    SEQ ID NO: 7
    DIVMTQTPLSLSVTPGQPASISCRSSRSLVHSRGN
    TYLHWYLQKPGQSPQLLIYKVSNRFIGVPDRFSGS
    GSGTDFTLKISRVEAEDVGVYYCSQSTHLPFTFGQ
    GTKLEIK
    (HCVR of Exemplary anti-IL17A antibody)
    SEQ ID NO: 8
    QVQLVQSGAEVKKPGSSVKVSCKASGYSFTDYHIH
    WVRQAPGQGLEWMGVINPMYGTTDYNQRFKGRVTI
    TADESTSTAYMELSSLRSEDTAVYYCARYDYFTGT
    GVYWGQGTLVTVSS
    (light chain of Exemplary anti-IL17A
    antibody)
    SEQ ID NO: 9
    DIVMTQTPLSLSVTPGQPASISCRSSRSLVHSRGN
    TYLHWYLQKPGQSPQLLIYKVSNRFIGVPDRFSGS
    GSGTDFTLKISRVEAEDVGVYYCSQSTHLPFTFGQ
    GTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCL
    LNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKD
    STYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV
    TKSFNRGEC
    (heavy chain of Exemplary anti-IL17A
    antibody)
    SEQ ID NO: 10
    QVQLVQSGAEVKKPGSSVKVSCKASGYSFTDYHIH
    WVRQAPGQGLEWMGVINPMYGTTDYNQRFKGRVTI
    TADESTSTAYMELSSLRSEDTAVYYCARYDYFTGT
    GVYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSES
    TAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAV
    LQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSN
    TKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPK
    PKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDG
    VEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNG
    KEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTL
    PPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQ
    PENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGN
    VFSCSVMHEALHNHYTQKSLSLSLG

Claims (20)

1-9. (canceled)
10. A method of treating RA, Ps, GenPs, Pruritus, AS, PA, PPP, HS or MM comprising administering to a patient in need thereof an effective amount of a bufferless aqueous pharmaceutical formulation comprising:
(i) an anti-IL-17A antibody at a concentration of 80 mg/mL+/−10%;
(ii) sucrose in a concentration of 234 mM+/−10%; and
(iii) a surfactant in a concentration of between 0.005% w/v+/−10% to 0.05% w/v+/−10%,
wherein, the pharmaceutical formulation is an aqueous solution at a pH between 5.2 to 6.5 and the anti-IL17A antibody comprises a light chain variable region (LCVR) and a heavy chain variable region (HCVR), wherein the LCVR comprises complementarity determining regions (CDRs) LCDR1, LCDR2, and LCDR3 and the HCVR comprises CDRs HCDR1, HCDR2, and HCDR3, wherein:
LCDR1 comprises the amino acid sequence of SEQ ID NO. 1,
LCDR2 comprises the amino acid sequence of SEQ ID NO. 2,
LCDR3 comprises the amino acid sequence of SEQ ID NO. 3,
HCDR1 comprises the amino acid sequence of SEQ ID NO. 4,
HCDR2 comprises the amino acid sequence of SEQ ID NO. 5, and
HCDR3 comprises the amino acid sequence of SEQ ID NO. 6.
11. (canceled)
12. The method of claim 10, comprising:
administering subcutaneously, to the patient, an initial dose of the pharmaceutical formulation, on day 0, followed by administering subcutaneously the pharmaceutical formulation to the patient at every four week interval thereafter,
wherein the initial dose of the pharmaceutical formulation comprises the anti-IL17A antibody at a concentration of about 160 mg, and the pharmaceutical formulation administered to the patient at every four week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
13. The method of claim 12, wherein the initial dose of the pharmaceutical formulation, comprising the anti-IL-17A antibody at a concentration of about 160 mg, comprises two doses of the pharmaceutical formulation wherein each of the two doses comprises about 80 mg of the anti-IL17A antibody.
14. The method of claim 10, comprising:
administering subcutaneously, to the patient, an initial dose of the pharmaceutical formulation, on day 0, followed by administering subcutaneously the pharmaceutical formulation to the patient at every two week interval thereafter,
wherein the initial dose of the pharmaceutical formulation comprises the anti-IL17A antibody at a concentration of about 160 mg, and the pharmaceutical formulation administered to the patient at every two week interval after the initial dose comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
15. The method of claim 14, wherein the initial dose of the pharmaceutical formulation, comprising the anti-IL-17A antibody at a concentration of about 160 mg, comprises two doses of the pharmaceutical formulation wherein each of the two doses comprises about 80 mg of the anti-IL17A antibody.
16. The method of claim 10, comprising:
administering subcutaneously, to the patient, an initial dose of the pharmaceutical formulation, on day 0, followed by administering subcutaneously the pharmaceutical formulation to the patient on each of days 14, 28, 42, 56, 70 and 84, and followed by administering subcutaneously the pharmaceutical formulation to the patient at every four week interval thereafter,
wherein the initial dose of the pharmaceutical formulation comprises the anti-IL17A antibody at a concentration of about 160 mg and,
wherein the pharmaceutical formulation, administered to the patient at each of days 14, 28, 42, 56, 70 and 84, and every four week interval thereafter, comprises the anti-IL17A antibody at a concentration of about 80 mg/mL.
17. The method of claim 16, wherein the initial dose of the pharmaceutical formulation, comprising the anti-IL-17A antibody at a concentration of about 160 mg, comprises two doses of the pharmaceutical formulation wherein each of the two doses comprises about 80 mg of the anti-IL17A antibody.
18. A method of reducing injection-associated pain experienced by a patient at the time of, or shortly after, SQ, IP and/or IM administration of an aqueous pharmaceutical formulation comprising an anti-IL17A antibody, the method comprising administering to a patient a bufferless aqueous pharmaceutical formulation comprising:
(i) an anti-IL-17A antibody at a concentration of 80 mg/mL+/−10%;
(ii) sucrose in a concentration of 234 mM+/−10%; and
(iii) a surfactant in a concentration of between 0.005% w/v+/−10% to 0.05% w/v+/−10%,
wherein, the pharmaceutical formulation is an aqueous solution at a pH between 5.2 to 6.5 and the anti-IL17A antibody comprises a light chain variable region (LCVR) and a heavy chain variable region (HCVR), wherein the LCVR comprises complementarity determining regions (CDRs) LCDR1, LCDR2, and LCDR3 and the HCVR comprises CDRs HCDR1, HCDR2, and HCDR3, wherein:
LCDR1 comprises the amino acid sequence of SEQ ID NO. 1,
LCDR2 comprises the amino acid sequence of SEQ ID NO. 2,
LCDR3 comprises the amino acid sequence of SEQ ID NO. 3,
HCDR1 comprises the amino acid sequence of SEQ ID NO. 4,
HCDR2 comprises the amino acid sequence of SEQ ID NO. 5, and
HCDR3 comprises the amino acid sequence of SEQ ID NO. 6,
wherein, said step of administering provides a therapeutically favorable level of injection-associated pain.
19. The method of claim 18, wherein the therapeutically favorable level of injection-associated pain comprises a VAS score of less than 30 mm or less than 20 mm.
20. The method of claim 18, wherein the pharmaceutical formulation of claim 1 is substantially free of an ionic tonicity excipient and is substantially free of L-amino acid excipients and the surfactant of the pharmaceutical formulation is polysorbate 80, and wherein the LCVR comprises the amino acid sequence of SEQ ID NO. 7 and the HCVR comprises the amino acid sequence of SEQ ID NO. 8.
21. The method of claim 10, wherein the surfactant is polysorbate 20 or polysorbate 80.
22. The method of claim 21, wherein the surfactant is polysorbate 80.
23. The method of claim 10, wherein the pharmaceutical formulation is substantially free of ionic tonicity excipient.
24. The method of claim 10, wherein the pharmaceutical formulation is substantially free of L-amino acid excipients.
25. The method of claim 10, wherein the LCVR comprises the amino acid sequence of SEQ ID NO. 7 and the HCVR comprises the amino acid sequence of SEQ ID NO. 8.
26. The method of claim 10, wherein anti-IL17A antibody comprises a light chain (LC) and a heavy chain (HC), wherein the LC comprises the amino acid sequence of SEQ ID NO. 9 and the HC comprises the amino acid sequence of SEQ ID NO. 10.
27. The method of claim 26, wherein the anti-IL17A antibody is ixekizumab.
28. The method of claim 10, wherein the surfactant is polysorbate 80, the pharmaceutical formulation is substantially free of ionic tonicity excipient and is substantially free of L-amino acid excipients, and wherein the LCVR comprises the amino acid sequence of SEQ ID NO. 7 and the HCVR comprises the amino acid sequence of SEQ ID NO. 8.
US18/176,844 2019-02-18 2023-03-01 Therapeutic antibody formulation Pending US20230183334A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/176,844 US20230183334A1 (en) 2019-02-18 2023-03-01 Therapeutic antibody formulation

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201962807006P 2019-02-18 2019-02-18
US201962880846P 2019-07-31 2019-07-31
US201962947198P 2019-12-12 2019-12-12
US16/787,254 US11634485B2 (en) 2019-02-18 2020-02-11 Therapeutic antibody formulation
US18/176,844 US20230183334A1 (en) 2019-02-18 2023-03-01 Therapeutic antibody formulation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/787,254 Division US11634485B2 (en) 2019-02-18 2020-02-11 Therapeutic antibody formulation

Publications (1)

Publication Number Publication Date
US20230183334A1 true US20230183334A1 (en) 2023-06-15

Family

ID=72040829

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/787,254 Active 2040-12-14 US11634485B2 (en) 2019-02-18 2020-02-11 Therapeutic antibody formulation
US18/176,844 Pending US20230183334A1 (en) 2019-02-18 2023-03-01 Therapeutic antibody formulation

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/787,254 Active 2040-12-14 US11634485B2 (en) 2019-02-18 2020-02-11 Therapeutic antibody formulation

Country Status (21)

Country Link
US (2) US11634485B2 (en)
EP (1) EP3927729A4 (en)
JP (2) JP7266108B2 (en)
KR (1) KR20210114989A (en)
CN (1) CN113474360A (en)
AU (2) AU2020225202B2 (en)
BR (1) BR112021015034A2 (en)
CA (1) CA3129901A1 (en)
CL (1) CL2021002182A1 (en)
CO (1) CO2021010697A2 (en)
CR (1) CR20210435A (en)
DO (1) DOP2021000170A (en)
EC (1) ECSP21060917A (en)
IL (1) IL285134A (en)
JO (1) JOP20210229A1 (en)
MA (1) MA55033A (en)
MX (1) MX2021009851A (en)
PE (1) PE20212185A1 (en)
SG (1) SG11202108627SA (en)
UA (1) UA128098C2 (en)
WO (1) WO2020172002A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2020225202B2 (en) 2019-02-18 2023-10-26 Eli Lilly And Company Therapeutic antibody formulation
CN117083295A (en) * 2021-01-08 2023-11-17 拉尼尔生物治疗公司 Neutralizing antibodies to IL-17A, fusion proteins thereof and uses thereof
CN116874596B (en) * 2023-09-06 2023-11-24 南京佰抗生物科技有限公司 Monoclonal antibody of anti S100 beta protein, preparation method and application thereof

Family Cites Families (359)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6884879B1 (en) 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
US6541606B2 (en) 1997-12-31 2003-04-01 Altus Biologics Inc. Stabilized protein crystals formulations containing them and methods of making them
GB0113179D0 (en) 2001-05-31 2001-07-25 Novartis Ag Organic compounds
JP4317010B2 (en) 2001-07-25 2009-08-19 ピーディーエル バイオファーマ,インコーポレイティド Stable lyophilized pharmaceutical formulation of IgG antibody
CN101721362B (en) 2002-02-14 2018-07-03 中外制药株式会社 Pharmaceutical solutions comprising antibody
WO2003087327A2 (en) 2002-04-11 2003-10-23 Medimmune Vaccines, Inc. Preservation of bioactive materials by freeze dried foam
AU2003230908A1 (en) 2002-04-11 2003-10-27 Medimmune Vaccines, Inc. Spray freeze dry of compositions for intranasal administration
EP1572915A4 (en) 2002-04-11 2011-01-05 Medimmune Vaccines Inc Preservation of bioactive materials by spray drying
AU2011226771B2 (en) 2002-06-14 2012-10-04 Medimmune, Llc Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US20060246060A1 (en) 2002-07-02 2006-11-02 Nesta Douglas P Novel stable formulation
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
US7055598B2 (en) 2002-08-26 2006-06-06 Halliburton Energy Services, Inc. Fluid flow control device and method for use of same
AU2003276844A1 (en) 2002-08-28 2004-03-19 Pharmacia Corporation Formulations of modified antibodies and methods of making the same
WO2004039337A2 (en) 2002-10-31 2004-05-13 Protein Design Labs, Inc. Stable liquid pharmaceutical formulation of antibodies that are prone to isomerization
AU2003293543A1 (en) 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
EP1578394A4 (en) 2002-12-31 2011-02-23 Nektar Therapeutics Antibody-containing particles and compositions
US20040208869A1 (en) 2003-01-30 2004-10-21 Medimmune, Inc. Uses of anti-integrin alphanubeta3 antibody formulations
KR101208291B1 (en) 2003-04-04 2012-12-05 노파르티스 아게 High concentration antibody and protein formulations
MX369959B (en) 2003-05-14 2019-11-27 Immunogen Inc Drug conjugate composition.
BRPI0411276A (en) 2003-06-05 2006-08-01 Genentech Inc B-cell depletion methods, B-cell neoplasm or malignancy treatment method, B-cell-regulated autoimmune dysfunction relief method, composition and industrialized article
EP1656170B1 (en) 2003-08-12 2019-03-13 Eli Lilly And Company Medication dispensing apparatus with triple screw threads for mechanical advantage
EP1698640B2 (en) 2003-10-01 2019-06-19 Kyowa Hakko Kirin Co., Ltd. Method of stabilizing antibody and stabilized solution-type antibody preparation
PE20050928A1 (en) 2003-11-21 2005-11-08 Schering Corp THERAPEUTIC COMBINATIONS OF ANTI-IGFR1 ANTIBODY
AR046774A1 (en) 2003-12-24 2005-12-21 Boehringer Ingelheim Pharma LIOFILIZED FORMULATION OF ANTIBODY CONJUGATES
EP1712240B1 (en) 2003-12-25 2015-09-09 Kyowa Hakko Kirin Co., Ltd. Stable water-based medicinal preparation containing antibody
TW200539855A (en) 2004-03-15 2005-12-16 Wyeth Corp Calicheamicin conjugates
EA009497B1 (en) 2004-03-30 2008-02-28 Эли Лилли Энд Компани Medication dispensing apparatus with spring-driven locking feature enabled by administration of final dose
CA2562771C (en) 2004-04-12 2013-04-09 Medimmune, Inc. Anti-il-9 antibody formulations and uses thereof
US8658203B2 (en) 2004-05-03 2014-02-25 Merrimack Pharmaceuticals, Inc. Liposomes useful for drug delivery to the brain
US20070196364A1 (en) 2004-07-27 2007-08-23 Human Genome Sciences, Inc. Pharmaceutical Formulation and Process
GB0417487D0 (en) 2004-08-05 2004-09-08 Novartis Ag Organic compound
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
ATE476994T1 (en) 2004-11-30 2010-08-15 Curagen Corp ANTIBODIES TO GPNMB AND THEIR USES
GT200600031A (en) 2005-01-28 2006-08-29 ANTI-BETA ANTIBODY FORMULATION
DK2567976T3 (en) 2005-03-23 2017-10-23 Genmab As ANTIBODIES AGAINST CD38 FOR TREATMENT OF MULTIPLE MYLOMA
ES2407468T3 (en) 2005-04-18 2013-06-12 Yeda Research And Development Company Limited Stabilized anti-hepatitis B (HBV) antibody formulations
CN105153306B (en) 2005-06-07 2020-12-11 艾斯巴技术-诺华有限责任公司 Stable and soluble antibodies inhibiting TNF alpha
CA2610839C (en) 2005-06-14 2019-06-25 Amgen Inc. Self-buffering protein formulations
NZ564098A (en) 2005-06-15 2010-04-30 Schering Corp Anti-IGF1R antibody formulations
WO2007002543A2 (en) 2005-06-23 2007-01-04 Medimmune, Inc. Antibody formulations having optimized aggregation and fragmentation profiles
NZ599176A (en) 2005-08-03 2014-04-30 Immunogen Inc Immunoconjugate formulations
CA2915270C (en) 2005-08-05 2017-07-11 Amgen Inc. Stable aqueous protein or antibody pharmaceutical formulations and their preparation
KR20140077946A (en) 2005-10-13 2014-06-24 휴먼 게놈 사이언시즈, 인코포레이티드 Methods and compositions for use in treatment of patients with autoantibody positive diseases
DOP2005000210A (en) 2005-10-19 2006-04-30 Genentech Inc ANTIBONY FORMULATIONS
US20070172520A1 (en) 2005-11-18 2007-07-26 University Of South Florida Immunotargeting of Nonionic Surfactant Vesicles
EP1951305A1 (en) 2005-11-22 2008-08-06 Wyeth a Corporation of the State of Delaware Immunoglobulin fusion protein formulations
EP3366702B1 (en) * 2005-12-13 2023-08-09 Eli Lilly And Company Anti-il-17 antibodies
US7390786B2 (en) 2005-12-21 2008-06-24 Wyeth Protein formulations with reduced viscosity and uses thereof
WO2007074880A1 (en) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
CN102887954B (en) 2005-12-29 2015-02-04 詹森生物科技公司 Human anti-il-23 antibodies, compositions, methods and uses
JP2009525986A (en) 2006-02-03 2009-07-16 メディミューン,エルエルシー Protein preparation
EA200870264A1 (en) 2006-02-15 2009-02-27 Имклоун Системз Инкорпорейтед ANTIBODY COMPOSITION
US9283260B2 (en) 2006-04-21 2016-03-15 Amgen Inc. Lyophilized therapeutic peptibody formulations
MX2008013535A (en) 2006-04-21 2008-10-29 Amgen Inc Buffering agents for biopharmaceutical formulations.
KR20090021298A (en) 2006-06-14 2009-03-02 임클론 시스템즈 인코포레이티드 Lyophilized formulations of anti-egfr antibodies
JPWO2008029908A1 (en) 2006-09-07 2010-01-21 協和発酵キリン株式会社 Stable lyophilized pharmaceutical formulation containing antibody
CA2663892A1 (en) 2006-09-25 2008-04-03 Medimmune, Llc Stabilized antibody formulations and uses thereof
ES2827180T3 (en) 2006-10-06 2021-05-20 Amgen Inc Stable Antibody Formulations
MX2009003982A (en) 2006-10-20 2009-04-27 Amgen Inc Stable polypeptide formulations.
DE102006053375A1 (en) 2006-11-10 2008-05-15 Boehringer Ingelheim Pharma Gmbh & Co. Kg Process for mixing powders
CN101553504A (en) 2006-12-11 2009-10-07 豪夫迈·罗氏有限公司 Abeta antibody parenteral formulation
CA2790018C (en) 2006-12-21 2015-02-03 Amgen Inc. Formulations
JP5419709B2 (en) 2007-01-09 2014-02-19 ワイス・エルエルシー Anti-IL-13 antibody preparation and use thereof
EP2109460B1 (en) 2007-01-11 2016-05-18 Novo Nordisk A/S Anti-kir antibodies, formulations, and uses thereof
KR20090113340A (en) 2007-03-22 2009-10-29 임클론 엘엘씨 Stable antibody formulations
KR101540823B1 (en) 2007-03-30 2015-07-30 메디뮨 엘엘씨 Antibody formulation
TW200909005A (en) * 2007-06-14 2009-03-01 Elan Pharm Inc Lyophilized immunoglobulin formulations and methods of preparation
US20090208492A1 (en) 2007-06-14 2009-08-20 Elan Pharmaceuticals, Inc. Lyophilized Immunoglobulin Formulations and Methods of Preparation
US20110014676A1 (en) 2007-06-29 2011-01-20 Battelle Memorial Institute Protein stabilization
WO2009015345A1 (en) 2007-07-25 2009-01-29 Amgen Inc. Pharmaceutical compositions comprising fc fusion proteins
EP2219602A1 (en) 2007-11-15 2010-08-25 Amgen, Inc Aqueous formulation of erythropoiesis stimulating protein stablised by antioxidants for parenteral administration
US9308257B2 (en) 2007-11-28 2016-04-12 Medimmune, Llc Protein formulation
KR20150080038A (en) 2007-11-30 2015-07-08 애브비 바이오테크놀로지 리미티드 Protein formulations and methods of making same
WO2009120684A1 (en) 2008-03-25 2009-10-01 Medimmune, Llc Antibody formulation
WO2009141239A1 (en) 2008-05-20 2009-11-26 F. Hoffmann-La Roche Ag A pharmaceutical formulation comprising an antibody against ox40l, uses thereof
EP2324315A2 (en) 2008-06-26 2011-05-25 Wyeth LLC Lyophilization cycle robustness strategy
EP3572073B1 (en) 2008-08-05 2023-12-20 Wyeth LLC Lyophilization above collapse
MX2011002159A (en) 2008-08-27 2011-03-29 Schering Corp Lyophilized formulatons of engineered anti-il-23p19 antibodies.
CN102159194A (en) 2008-09-19 2011-08-17 霍夫曼-拉罗奇有限公司 Novel antibody formulation
HUE036126T2 (en) 2008-09-19 2018-06-28 Pfizer Stable liquid antibody formulation
WO2010042705A1 (en) 2008-10-09 2010-04-15 Medimmune, Llc Antibody formulation
JP5933975B2 (en) 2008-11-12 2016-06-15 メディミューン,エルエルシー Antibody preparation
JP2012509269A (en) 2008-11-17 2012-04-19 ジェネンテック, インコーポレイテッド Methods and formulations for reducing polymer aggregation under physiological conditions
AU2009313756B2 (en) 2008-11-17 2015-02-26 F. Hoffmann-La Roche Ag Method and formulation for reducing aggregation of a macromolecule under physiological conditions
CN102281901A (en) 2008-11-20 2011-12-14 弗·哈夫曼-拉罗切有限公司 Therapeutic protein formulations
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
NZ606283A (en) 2008-11-28 2014-08-29 Abbvie Inc Stable antibody compositions and methods for stabilizing same
CN102245206A (en) * 2008-12-09 2011-11-16 弗·哈夫曼-拉罗切有限公司 Method for obtaining an excipient-free antibody solution
EP2196476A1 (en) 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
WO2010069858A1 (en) 2008-12-19 2010-06-24 F. Hoffmann-La Roche Ag Pharmaceutical composition
FR2944448B1 (en) 2008-12-23 2012-01-13 Adocia STABLE PHARMACEUTICAL COMPOSITION COMPRISING AT LEAST ONE MONODONAL ANTIBODY AND AT LEAST ONE AMPHIPHILIC POLYSACHARIDE COMPRISING SUBSTITUENTS DERIVED FROM HYDROFOB ALCOHOLS OR HYDROPHOBIC AMINES.
WO2010102251A2 (en) 2009-03-05 2010-09-10 Abbott Laboratories Il-17 binding proteins
WO2010100200A2 (en) 2009-03-05 2010-09-10 Novartis Ag Lyophilised antibody formulation
EP2403873A1 (en) 2009-03-05 2012-01-11 Ablynx N.V. Novel antigen binding dimer-complexes, methods of making/avoiding and uses thereof
WO2010102276A2 (en) 2009-03-06 2010-09-10 Medimmune, Llc Humanized anti-cd19 antibody formulations
JP5426661B2 (en) 2009-03-31 2014-02-26 デンカ生研株式会社 Immunoassay method and reagent therefor
JP2010241718A (en) 2009-04-03 2010-10-28 Kyowa Hakko Kirin Co Ltd Stable aqueous solution preparation of antibody
US20100322943A1 (en) 2009-06-17 2010-12-23 Thomas Cantor Therapeutic and diagnostic affinity purified specific polyclonal antibodies
WO2010148321A1 (en) 2009-06-18 2010-12-23 Wyeth Llc Slow dissolution method for reconstitution of lyophilized material
AU2010263058A1 (en) 2009-06-18 2012-01-12 Wyeth Llc Lyophilized formulations for small modular immunopharmaceuticals
CA2765220A1 (en) 2009-07-14 2011-01-20 Biogen Idec Ma Inc. Methods for inhibiting yellow color and peroxide formation in a composition
EP2458990B1 (en) 2009-07-28 2016-03-30 Merck Sharp & Dohme Corp. Methods for producing high concentration lyophilized pharmaceutical formulations
US9345661B2 (en) 2009-07-31 2016-05-24 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
US20110059079A1 (en) 2009-09-04 2011-03-10 Xoma Technology Ltd. Antibody Coformulations
AR078161A1 (en) 2009-09-11 2011-10-19 Hoffmann La Roche VERY CONCENTRATED PHARMACEUTICAL FORMULATIONS OF AN ANTIBODY ANTI CD20. USE OF THE FORMULATION. TREATMENT METHOD
CA2781467C (en) 2009-11-20 2015-10-13 Biocon Limited Formulations of antibody
KR20120110175A (en) 2009-12-29 2012-10-09 에프. 호프만-라 로슈 아게 Antibody formulation
JO3417B1 (en) 2010-01-08 2019-10-20 Regeneron Pharma Stabilized formulations containing anti-interleukin-6 receptor (il-6r) antibodies
ES2652637T3 (en) 2010-01-15 2018-02-05 Kirin-Amgen, Inc. Antibody formulation and therapeutic regimens
CA2794929C (en) 2010-03-01 2018-06-05 Progenics Pharmaceuticals, Inc. Concentrated protein formulations and uses thereof
EP2708252B1 (en) 2010-03-01 2015-07-29 Eli Lilly and Company Automatic injection device with delay mechanism including dual functioning biasing member
US20110223208A1 (en) 2010-03-09 2011-09-15 Beth Hill Non-Aqueous High Concentration Reduced Viscosity Suspension Formulations
AR080685A1 (en) 2010-03-17 2012-05-02 Abbott Res Bv COMPOSITIONS OF ANTI-FACTOR ANTI-FACTOR OF NERVOUS GROWTH (NGF)
RU2600847C2 (en) 2010-05-10 2016-10-27 Интас Биофармасьютикалс Лимитед Liquid composition of polypeptides containing fc domain of immunoglobulin
ME02819B (en) 2010-05-14 2018-01-20 Amgen Inc High concentration antibody formulations
EP2399604A1 (en) 2010-06-25 2011-12-28 F. Hoffmann-La Roche AG Novel antibody formulation
SG186421A1 (en) 2010-07-02 2013-01-30 Medimmune Llc Antibody formulations
JOP20190250A1 (en) 2010-07-14 2017-06-16 Regeneron Pharma Stabilized formulations containing anti-ngf antibodies
FR2962650B1 (en) 2010-07-19 2013-04-05 Lab Francais Du Fractionnement COMPOSITION OF HUMAN CONCENTRATED IMMUNOGLOBULINS
WO2012028683A1 (en) 2010-09-02 2012-03-08 Novartis Ag Antibody gel system for sustained drug delivery
EP2616814A1 (en) 2010-09-17 2013-07-24 AbbVie Inc. Raman spectroscopy for bioprocess operations
NZ609557A (en) 2010-10-06 2014-12-24 Regeneron Pharma Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies
DE20187001T1 (en) 2010-11-05 2021-04-01 Novartis Ag METHOD OF TREATMENT OF PSORIASUS ARTHRITIS WITH IL-17 ANTAGONISTS
PT2637690T (en) 2010-11-11 2016-12-27 Abbvie Biotechnology Ltd High concentration anti-tnfalpha antibody liquid formulations
CA2818612C (en) 2010-11-19 2020-12-29 Forsight Vision4, Inc. Therapeutic agent formulations for implanted devices
US9458240B2 (en) 2010-12-10 2016-10-04 Novartis Pharma Ag Anti-BAFFR antibody formulations
EP2471554A1 (en) 2010-12-28 2012-07-04 Hexal AG Pharmaceutical formulation comprising a biopharmaceutical drug
EP2500035A1 (en) 2011-03-15 2012-09-19 Icon Genetics GmbH Pharmaceutical formulation containing immunglobulin
US20130344074A1 (en) 2011-03-16 2013-12-26 Sanofi Uses of a dual v region antibody-like protein
EP2691112B1 (en) 2011-03-31 2018-05-23 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
UA116189C2 (en) 2011-05-02 2018-02-26 Мілленніум Фармасьютікалз, Інк. FORMULATION FOR ANTI-α4β7 ANTIBODY
MX354101B (en) 2011-05-02 2018-02-13 Millennium Pharm Inc Formulation for anti-î±4î²7 antibody.
JOP20200043A1 (en) 2011-05-10 2017-06-16 Amgen Inc Methods of treating or preventing cholesterol related disorders
WO2013003680A1 (en) 2011-06-30 2013-01-03 Genentech, Inc. Anti-c-met antibody formulations
AR087305A1 (en) 2011-07-28 2014-03-12 Regeneron Pharma STABILIZED FORMULATIONS CONTAINING ANTI-PCSK9 ANTIBODIES, PREPARATION METHOD AND KIT
SG11201401360XA (en) 2011-10-25 2014-05-29 Onclave Therapeutics Ltd Antibody formulations and methods
RS63948B1 (en) 2011-10-31 2023-02-28 Hoffmann La Roche Anti-il13 antibody formulations
WO2013083497A1 (en) 2011-12-06 2013-06-13 F. Hoffmann-La Roche Ag Antibody formulation
WO2013096901A1 (en) 2011-12-23 2013-06-27 Mersana Therapeutics, Inc. Pharmaceutical formulations for fumagillin derivative-phf conjugates
EP2807190B1 (en) 2012-01-23 2018-12-26 Regeneron Pharmaceuticals, Inc. Stabilized formulations containing anti-ang2 antibodies
BR112014021325A2 (en) 2012-03-07 2017-08-22 Cadila Healthcare Ltd LIQUID PHARMACEUTICAL FORMULATION AND LYOPHILIZED FORMULATION
EP3156073A1 (en) * 2012-03-07 2017-04-19 Eli Lilly and Company Il-17 antibody formulation
JP6254956B2 (en) 2012-03-07 2017-12-27 デカ・プロダクツ・リミテッド・パートナーシップ Infusion pump assembly
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
US20140004131A1 (en) 2012-05-04 2014-01-02 Novartis Ag Antibody formulation
SG10201709555SA (en) 2012-05-18 2017-12-28 Genentech Inc High-concentration monoclonal antibody formulations
AR092325A1 (en) 2012-05-31 2015-04-15 Regeneron Pharma STABILIZED FORMULATIONS CONTAINING ANTI-DLL4 ANTIBODIES AND KIT
EP2863951A1 (en) 2012-06-12 2015-04-29 Boehringer Ingelheim International GmbH Pharmaceutical formulation for a therapeutic antibody
US9216219B2 (en) 2012-06-12 2015-12-22 Novartis Ag Anti-BAFFR antibody formulation
EP2866833B1 (en) 2012-06-27 2019-05-15 Merck Sharp & Dohme Corp. Crystalline anti-human il-23 antibodies
US20140227250A1 (en) 2012-08-23 2014-08-14 Merck Sharp & Dohme Corp. Stable formulations of antibodies to tslp
US8883979B2 (en) 2012-08-31 2014-11-11 Bayer Healthcare Llc Anti-prolactin receptor antibody formulations
US8613919B1 (en) 2012-08-31 2013-12-24 Bayer Healthcare, Llc High concentration antibody and protein formulations
UA115789C2 (en) 2012-09-05 2017-12-26 Трейкон Фармасутікалз, Інк. Antibody formulations and uses thereof
ES2784861T3 (en) * 2012-09-07 2020-10-01 Coherus Biosciences Inc Stable aqueous formulations of adalimumab
KR20150070384A (en) 2012-10-25 2015-06-24 메디뮨 엘엘씨 Stable, low viscosity antibody formulation
JP6339578B2 (en) 2012-10-31 2018-06-06 タケダ・ゲー・エム・ベー・ハーTakeda GmbH Lyophilized preparation containing GM-CSF neutralizing compound
EP2727602A1 (en) 2012-10-31 2014-05-07 Takeda GmbH Method for preparation of a high concentration liquid formulation of an antibody
AU2013337644A1 (en) 2012-11-01 2015-05-07 Abbvie Inc. Stable Dual Variable Domain Immunoglobulin protein formulations
EP2931311A4 (en) 2012-12-13 2016-08-17 Merck Sharp & Dohme Lyophilized spherical pellets of anti-il-23 antibodies
UA117466C2 (en) 2012-12-13 2018-08-10 Мерк Шарп Енд Доме Корп. SOLUTION FORMULATIONS OF ENGINEERED ANTI-IL-23p19 ANTIBODIES
US9844594B2 (en) 2012-12-18 2017-12-19 Merck Sharp & Dohme Corp. Liquid formulations for an anti-TNF α antibody
BR112015014853A2 (en) 2012-12-21 2017-08-22 Glenmark Pharmaceuticals Sa PHARMACEUTICAL FORMULATION, LYOPHILIZED PHARMACEUTICAL FORMULATION, USE OF A PHARMACEUTICAL FORMULATION AND ARTICLE OF MANUFACTURE
AU2014214843A1 (en) 2013-02-07 2015-05-21 Immunomedics, Inc. Pro-drug form (P2PDox) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
EP2968460B1 (en) 2013-03-11 2021-01-06 Amgen Inc. Protein formulations
CA2901575C (en) 2013-03-13 2022-08-30 Seattle Genetics, Inc. Cyclodextrin and antibody-drug conjugate formulations
NZ711567A (en) 2013-03-13 2020-04-24 Genentech Inc Antibody formulations
WO2014141152A2 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Low concentration antibody formulations
CA2906101A1 (en) 2013-03-15 2014-09-18 Bayer Healthcare Llc Anti-prolactin receptor antibody formulations
WO2014140361A1 (en) 2013-03-15 2014-09-18 Takeda Gmbh Formulation of an antibody and use thereof
SG10201707633VA (en) 2013-03-15 2017-11-29 Abbvie Deutschland Anti-egfr antibody drug conjugate formulations
US9700485B2 (en) * 2013-04-24 2017-07-11 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
TWI679019B (en) 2013-04-29 2019-12-11 法商賽諾菲公司 Anti-il-4/anti-il-13 bispecific antibody formulations
KR101814895B1 (en) 2013-06-04 2018-01-04 바이옴 바이오사이언스 피브이티. 엘티디. Coated particles and compositions comprising same
CN105517577A (en) 2013-06-21 2016-04-20 先天制药公司 Enzymatic conjugation of polypeptides
US10513555B2 (en) 2013-07-04 2019-12-24 Prothena Biosciences Limited Antibody formulations and methods
CN110559435B (en) 2013-09-11 2023-10-20 伊戈尔生物药品股份有限公司 Liquid protein formulations comprising ionic liquids
JP6942465B2 (en) 2013-09-27 2021-09-29 ジェネンテック, インコーポレイテッド Anti-PDL1 antibody preparation
WO2015061584A1 (en) 2013-10-24 2015-04-30 Medimmune, Llc Stable, aqueous antibody formulations
CN106163567B (en) 2013-11-21 2021-06-11 根马布股份公司 Antibody-drug conjugate lyophilized formulation
WO2015110930A1 (en) 2014-01-24 2015-07-30 Pfizer Inc. Modified interleukin 21 receptor proteins
WO2015121318A1 (en) 2014-02-12 2015-08-20 Sanofi Anti-il-4/anti-il-13 bispecific antibody/polyglutamate formulations
US20150274819A1 (en) 2014-03-03 2015-10-01 La Jolla Biologics, Inc. Stable aqueous recombinant protein formulations
US20150291689A1 (en) 2014-03-09 2015-10-15 Abbvie, Inc. Compositions and Methods for Treating Rheumatoid Arthritis
CA2941232C (en) 2014-03-11 2020-08-25 Green Cross Holdings Corporation Method for purifying immunoglobulin
EP3118209B1 (en) 2014-03-11 2020-02-19 Green Cross Holdings Corporation Method for purifying immunoglobulin
TW201623331A (en) 2014-03-12 2016-07-01 普羅帝納生物科學公司 Anti-MCAM antibodies and associated methods of use
AU2015241038A1 (en) 2014-03-31 2016-10-13 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and OX40 binding agonists
KR102530900B1 (en) 2014-03-31 2023-05-12 암젠 케이-에이, 인크. Methods of treating nail and scalp psoriasis
US10688187B2 (en) 2014-04-02 2020-06-23 Intas Pharmaceuticals Ltd. Liquid pharmaceutical composition of adalimumab
SG11201607306PA (en) 2014-04-07 2016-09-29 Seattle Genetics Inc Stable formulations for anti-cd19 antibodies and antibody-drug conjugates
JP2015209384A (en) 2014-04-24 2015-11-24 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Pharmaceutical formulation
EP4285930A3 (en) 2014-06-26 2024-02-28 Amgen Inc. Protein formulations
WO2016025911A1 (en) 2014-08-14 2016-02-18 Brown University Compositions for stabilizing and delivering proteins
WO2016036678A1 (en) 2014-09-02 2016-03-10 Medimmune, Llc Formulations of bispecific antibodies
US20170281769A1 (en) 2014-09-03 2017-10-05 Medimmune Limited STABLE ANTI-IL-4Ra FORMULATION
US20160137727A1 (en) 2014-09-15 2016-05-19 Genentech, Inc. Antibody formulations
GB201416960D0 (en) 2014-09-25 2014-11-12 Antikor Biopharma Ltd Biological materials and uses thereof
WO2016061551A1 (en) 2014-10-17 2016-04-21 Amgen Inc. Antibodies directed to angiopoietin-1 and angiopoietin-2 for ocular therapies
BR112017007393A2 (en) 2014-10-18 2017-12-19 Pfizer anti-il-7r antibody compositions
TWI806150B (en) 2014-11-07 2023-06-21 瑞士商諾華公司 Stable protein solution formulation containing high concentration of an anti-vegf antibody
US20160144025A1 (en) 2014-11-25 2016-05-26 Regeneron Pharmaceuticals, Inc. Methods and formulations for treating vascular eye diseases
AR103173A1 (en) 2014-12-22 2017-04-19 Novarits Ag PHARMACEUTICAL PRODUCTS AND STABLE LIQUID COMPOSITIONS OF ANTIBODIES IL-17
EP3237000A1 (en) 2014-12-23 2017-11-01 Pfizer Inc Stable aqueous antibody formulation for anti tnf alpha antibodies
WO2016109822A1 (en) 2014-12-31 2016-07-07 Novelmed Therapeutics, Inc. Formulation of aglycosylated therapeutic antibodies
AU2016210918A1 (en) 2015-01-28 2017-07-13 Pfizer Inc., Stable aqueous anti- vascular endothelial growth factor (VEGF) antibody formulation
US10039826B2 (en) 2015-02-09 2018-08-07 Ucb Biopharma Sprl Pharmaceutical formulation comprising antibodies which bind colony stimulating factor-1 receptor (CSF1R)
RU2017131618A (en) 2015-02-13 2019-03-13 Санофи STABLE LIQUID COMPOSITION FOR MONOCLONAL ANTIBODIES
PE20180394A1 (en) 2015-04-17 2018-02-28 Bristol Myers Squibb Co COMPOSITIONS INCLUDING A COMBINATION OF A PROGRAMMED ANTI DEATH ANTIBODY 1 (PD-1) AND ANOTHER ANTIBODY
KR101808234B1 (en) 2015-06-23 2017-12-12 (주)알테오젠 A stable liquid formulation of fusion protein with IgG Fc domain
EA201792527A1 (en) 2015-07-16 2018-06-29 Эли Лилли Энд Компани TREATMENT OF ZUD
ES2818229T3 (en) 2015-08-19 2021-04-09 Astrazeneca Ab Stable anti-IFNAR1 formulation
CN106474470B (en) 2015-08-28 2020-05-22 江苏恒瑞医药股份有限公司 Composition of anti-IL-17A antibody
MY186352A (en) 2015-09-09 2021-07-15 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
US9862760B2 (en) 2015-09-16 2018-01-09 Novartis Ag Polyomavirus neutralizing antibodies
US20230134160A1 (en) 2015-09-22 2023-05-04 Pfizer Inc. Method Of Preparing A Therapeutic Protein Formulation And Antibody Formulation Produced By Such A Method
US10786567B2 (en) 2015-09-28 2020-09-29 Suzhou Suncediabiopharmaeuticals Co., Ltd. Stable anti-PD-1 antibody pharmaceutical preparation and application thereof in medicine
WO2017055966A1 (en) 2015-10-01 2017-04-06 Pfizer Inc. Low viscosity antibody compositions
BR112018008769A2 (en) 2015-10-30 2018-12-04 Genentech Inc pharmaceutical formulations, reconstituted and lyophilized aqueous liquid formulations, syringe, method of manufacturing a pharmaceutical formulation, method for treating a disease and use of a reconstructed formulation
CN106620690A (en) 2015-10-30 2017-05-10 上海抗体药物国家工程研究中心有限公司 Stable antibody preparation
MX2018006139A (en) 2015-11-18 2019-03-14 Sio2 Medical Products Inc Pharmaceutical package for ophthalmic formulations.
EP3383435A4 (en) 2015-11-30 2019-07-10 Medimmune, LLC Optimized ratios of amino acids and sugars as amorphous stabilizing compounds in pharmaceutical compositions containing high concentrations of protein-based therapeutic agents
CN108367072B (en) 2015-12-07 2022-12-27 默克专利股份有限公司 Aqueous pharmaceutical formulation comprising anti-PD-L1 antibody AVELUMAB
CN109152831A (en) 2015-12-29 2019-01-04 安口生物公司 The buffer preparation of Avastin
RU2736830C2 (en) 2016-01-12 2020-11-20 Др. Редди'С Лабораторис Лимитед Stable pharmaceutical composition
EP3402465B1 (en) 2016-01-13 2024-03-13 Genmab A/S Formulation for antibody and drug conjugate thereof
JP6953433B2 (en) 2016-01-26 2021-10-27 フォーマイコン アーゲーFormycon Ag A liquid pharmaceutical composition containing a VEGF antagonist and a prefilled syringe containing the pharmaceutical composition.
WO2017132457A1 (en) * 2016-01-28 2017-08-03 Janssen Biotech, Inc. BISPECIFIC ANTI-TNF-α/IL-17A ANTIBODIES AND ANTI-TNF-α ANTIBODIES AND METHODS OF THEIR USE
AR103622A1 (en) 2016-02-05 2017-05-24 Ucb Biopharma Sprl PHARMACEUTICAL FORMULATION
WO2017147248A1 (en) 2016-02-24 2017-08-31 Visterra, Inc. Formulations of antibody molecules to influenza virus
WO2017149513A1 (en) 2016-03-03 2017-09-08 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
GB201604124D0 (en) 2016-03-10 2016-04-27 Ucb Biopharma Sprl Pharmaceutical formulation
WO2017165736A1 (en) 2016-03-25 2017-09-28 Visterra, Inc. Formulation of antibody molecules to dengue virus
JP6992262B2 (en) 2016-03-31 2022-02-15 東ソー株式会社 Manufacturing method of denaturing antibody measurement reagent
EP3442503B1 (en) 2016-04-12 2023-03-15 Capsular Technologies Pty Ltd. Injectable composition for delivery of a biologically active agent
WO2017180594A1 (en) 2016-04-13 2017-10-19 Medimmune, Llc Use of amino acids as stabilizing compounds in pharmaceutical compositions containing high concentrations of protein-based therapeutic agents
GB201608323D0 (en) 2016-05-12 2016-06-29 Ucb Biopharma Sprl Pharmaceutical compositions
TWI826351B (en) 2016-05-31 2023-12-21 大陸商鴻運華寧(杭州)生物醫藥有限公司 R antibodies, their pharmaceutical compositions and uses
WO2017208210A1 (en) 2016-06-03 2017-12-07 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
CN109475508A (en) 2016-06-07 2019-03-15 阿瑞迪思医药品股份有限责任公司 The preparation method of the instant film containing bioactive materials of thermal stability with enhancing
PT3475303T (en) 2016-06-27 2021-06-25 Morphosys Ag Anti-cd19 antibody formulations
CR20180599A (en) 2016-06-30 2019-04-09 Celltrion Inc STABLE LIQUID PHARMACEUTICAL PREPARATION
KR20220137159A (en) 2016-07-05 2022-10-11 사노피 Antibody formulations
EP3487881A1 (en) 2016-07-19 2019-05-29 Novartis AG Methods of treating new-onset plaque type psoriasis using il-17 antagonists
JOP20170170B1 (en) 2016-08-31 2022-09-15 Omeros Corp Highly concentrated low viscosity masp-2 inhibitory antibody formulations, kits, and methods
FR3056912B1 (en) 2016-09-30 2019-12-27 Laboratoire Francais Du Fractionnement Et Des Biotechnologies METHOD OF VIRAL INACTIVATION OF A PREPARATION OF MONOCLONAL ANTIBODIES
US11459401B2 (en) 2016-10-06 2022-10-04 Amgen Inc. Reduced viscosity protein pharmaceutical formulations
US11351256B2 (en) 2016-10-07 2022-06-07 Regeneron Pharmaceuticals, Inc. Room temperature stable lyophilized protein
CA3047530A1 (en) 2016-12-23 2018-06-28 Serum Institute Of India Private Limited Improved methods for enhancing antibody productivity in mammalian cell culture and minimizing aggregation during downstream, formulation processes and stable antibody formulationsobtained thereof
KR102471458B1 (en) 2016-12-28 2022-11-25 제이씨알 파마 가부시키가이샤 Freeze-dried formulation
WO2018122053A1 (en) 2016-12-29 2018-07-05 F. Hoffmann-La Roche Ag Anti-angiopoietin-2 antibody formulation
CN108261544B (en) 2016-12-30 2023-05-05 江苏太平洋美诺克生物药业股份有限公司 Stable pharmaceutical formulation comprising CD147 monoclonal antibody
CN108261391B (en) 2016-12-30 2022-03-01 江苏太平洋美诺克生物药业有限公司 Stable pharmaceutical formulation comprising CD147 monoclonal antibody
EP3569224B1 (en) 2017-01-11 2022-12-14 Celltrion Inc. Stable liquid formula
KR20200113292A (en) 2017-01-17 2020-10-06 제넨테크, 인크. Subcutaneous her2 antibody formulations
BR112019014785A2 (en) 2017-01-19 2020-05-12 Bayer Pharma Aktiengesellschaft NEW STABLE FORMULATION FOR FXIA ANTIBODIES
GB201703063D0 (en) 2017-02-24 2017-04-12 Arecor Ltd Stabilized antibody protein solutions
GB201703062D0 (en) 2017-02-24 2017-04-12 Arecor Ltd Stabilized antibody protein solutions
CA3235178A1 (en) 2017-03-01 2018-09-07 Medimmmune Limited Formulations of monoclonal antibodies
TW202228779A (en) 2017-03-01 2022-08-01 英商梅迪繆思有限公司 Anti-rsv monoclonal antibody formulation
JP7379159B2 (en) 2017-03-06 2023-11-14 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Aqueous anti-PD-L1 antibody preparation
EP3372242A1 (en) 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
UA123847C2 (en) 2017-03-16 2021-06-09 Лг Кем, Лтд. Liquid formulation of anti-tnf alpha antibody
TW201836637A (en) 2017-03-29 2018-10-16 持田製藥股份有限公司 Liquid pharmaceutical agent comprising antibody characterized by realizing the stability of dimer formation inhibition and deamidation inhibition during long-term storage and being suitable for a wide range of antibody concentration
WO2018179138A1 (en) 2017-03-29 2018-10-04 持田製薬株式会社 Antibody-containing liquid preparation
EA201991912A1 (en) 2017-03-29 2020-03-10 Селджин Корпорейшн COMPOSITIONS CONTAINING PD-1-BINDING PROTEINS AND METHODS FOR PRODUCING THEREOF
WO2018181876A1 (en) 2017-03-31 2018-10-04 Meiji Seikaファルマ株式会社 Aqueous formulation, aqueous formulation in injector, antibody protein disaggregating agent, and antibody protein disaggregation method
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
JOP20190255A1 (en) 2017-04-28 2019-10-27 Amgen Inc Formulations of human anti-rankl antibodies, and methods of using the same
JOP20190260A1 (en) 2017-05-02 2019-10-31 Merck Sharp & Dohme Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
BR112019022873A8 (en) 2017-05-02 2023-04-11 Merck Sharp & Dohme FORMULATION, AND, INJECTION VESSEL OR DEVICE.
SG11202003754YA (en) 2017-05-16 2020-05-28 Bhamis Research Laboratory Pvt Ltd High concentration protein formulations with reduced viscosity
JP2020527552A (en) 2017-07-10 2020-09-10 バイエル・ファルマ・アクティエンゲゼルシャフト Prolactin receptor antibody for androgenetic alopecia and female pattern baldness
CN107400164A (en) 2017-07-18 2017-11-28 中山和芯生物技术有限公司 A kind of biological products stabilizer containing sucrose and its preparation method and application
WO2019018640A1 (en) 2017-07-21 2019-01-24 Novartis Ag Dosage regimens for anti-gitr antibodies and uses thereof
US11077059B2 (en) 2017-07-25 2021-08-03 Elektrofi, Inc. Electrospraying formation of particles including agents
US11498961B2 (en) 2017-07-27 2022-11-15 Jiangsu Hengrui Medicine Co., Ltd. SOST antibody pharmaceutical composition and uses thereof
CA3069073A1 (en) 2017-07-28 2019-01-31 F. Hoffmann-La Roche Ag Bispecific antibody formulation
WO2019036382A1 (en) 2017-08-15 2019-02-21 Progenity Inc. Treatment of inflammatory disease using ingestible device to release immune modulator
TW201919710A (en) 2017-08-23 2019-06-01 日商第一三共股份有限公司 Formulation of antibody-drug conjugate and lyophilization method thereof
RU2020112302A (en) 2017-09-05 2021-10-06 Мерк Шарп И Доум Корп. COMPOUNDS FOR REDUCING THE VISCOSITY OF BIOLOGICAL COMPOSITIONS
EP3459527B1 (en) 2017-09-20 2022-11-23 Tillotts Pharma Ag Method for preparing a solid dosage form comprising antibodies by wet granulation, extrusion and spheronization
MA50670A (en) 2017-09-29 2020-08-05 Janssen Biotech Inc NEW FORMULATIONS TO STABILIZE LOW-DOSE ANTIBODY COMPOSITIONS
JP7170983B2 (en) 2017-10-13 2022-11-15 国立大学法人大阪大学 Protein-containing liquid formulation with improved storage stability and method for producing the same
CN109745559A (en) 2017-11-01 2019-05-14 三生国健药业(上海)股份有限公司 The liquid preparation of the monoclonal antibody of anti-human IL-17A
GB201718888D0 (en) 2017-11-15 2017-12-27 Ucb Biopharma Sprl Method
GB201719447D0 (en) 2017-11-23 2018-01-10 Ucb Biopharma Sprl Pharmaceutical composition
KR20200090164A (en) 2017-11-29 2020-07-28 프로테나 바이오사이언시즈 리미티드 Lyophilized formulations of monoclonal antibodies to transthyretin
EP3718531A4 (en) 2017-11-30 2023-08-16 Bio-Thera Solutions, Ltd. Liquid preparation of humanized antibody for treating il-6-related disease
TW201927337A (en) 2017-12-22 2019-07-16 大陸商江蘇恆瑞醫藥股份有限公司 LAG-3 antibody pharmaceutical composition and use thereof
MA51747A (en) 2018-02-08 2020-12-16 Amgen Inc LOW PH PHARMACEUTICAL ANTIBODY FORMULATION
MX2020009275A (en) 2018-03-07 2021-01-08 Pfizer Anti-pd-1 antibody compositions.
US11427639B2 (en) 2018-04-02 2022-08-30 Richter Gedeon Nyrt. Antibody-containing aqueous formulation and use thereof
US20210087250A1 (en) 2018-04-06 2021-03-25 Cyprumed Gmbh Pharmaceutical compositions for the transmucosal delivery of therapeutic peptides and proteins
EP3552631A1 (en) 2018-04-10 2019-10-16 Inatherys Antibody-drug conjugates and their uses for the treatment of cancer
CA3095986A1 (en) 2018-04-10 2019-10-17 Genmab A/S Axl-specific antibodies for cancer treatment
AU2019256289A1 (en) 2018-04-17 2020-11-12 Outlook Therapeutics, Inc. Buffered formulations of bevacizumab for use of treating diseases
KR20210005096A (en) 2018-04-25 2021-01-13 메디뮨 리미티드 Formulation of human anti-PD-L1 antibody
KR20210021299A (en) 2018-05-10 2021-02-25 리제너론 파아마슈티컬스, 인크. Formulation containing high concentration VEGF receptor fusion protein
CN112638941A (en) 2018-05-14 2021-04-09 免疫医疗有限公司 Antibodies to LIF and dosage forms thereof
CN110538321B (en) 2018-05-29 2023-03-10 江苏恒瑞医药股份有限公司 CD47 antibody pharmaceutical composition and application thereof
CN112512583B (en) 2018-06-01 2023-11-14 乐天医药生技股份有限公司 Phthalocyanine dye conjugate composition
US20210223262A1 (en) 2018-06-07 2021-07-22 Merck Sharp & Dohme Corp. Lyosphere critical reagent kit
EP3810268A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an il-12/il-23 inhibitor
EP3810085A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an integrin inhibitor
US20230041197A1 (en) 2018-06-20 2023-02-09 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an immunomodulator
WO2019246317A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease or condition in a tissue originating from the endoderm
EP3810095A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a tnf inhibitor
WO2019245373A1 (en) 2018-06-22 2019-12-26 Bioralix B.V. Formulations of biological polymers for oral administration
US11738068B2 (en) 2018-06-25 2023-08-29 Jcr Pharmaceuticals Co., Ltd. Protein-containing aqueous liquid formulation
TW202011995A (en) 2018-07-03 2020-04-01 比利時商葛萊伯格有限公司 High concentration liquid antibody formulations
NZ772400A (en) 2018-07-05 2024-02-23 Hangzhou Dac Biotech Co Ltd Cross-linked pyrrolobenzodiazepine dimer (pbd) derivative and its conjugates
US11730698B2 (en) 2018-07-19 2023-08-22 Celltrion Inc. Stable liquid pharmaceutical preparation
WO2020016417A1 (en) 2018-07-19 2020-01-23 Ichnos Sciences S.A. Liquid antibody formulation
US11396542B2 (en) 2018-08-21 2022-07-26 Synkrino Biotherapeutics, Inc. Astrotactin1-based compositions and pharmaceutical formulations
EP3849513A1 (en) 2018-09-11 2021-07-21 Ichnos Sciences SA Compositions comprising a bispecific antibody, bufffer and one or more stabilizing agents
CA3111858A1 (en) 2018-09-13 2020-03-19 F. Hoffmann-La Roche Ag Csf-1r antibody formulation
CA3112215A1 (en) 2018-10-05 2020-04-09 Five Prime Therapeutics, Inc. Anti-fgfr2 antibody formulations
US20210355233A1 (en) 2018-10-10 2021-11-18 Astellas Pharma Inc. Pharmaceutical composition containing tagged site-antihuman antibody fab fragment complex
JP2022504745A (en) 2018-10-12 2022-01-13 ハンジョウ ディーエーシー バイオテック シーオー.,エルティディ. Conjugated conjugate containing 2,3-diaminosuccinyl group
US20210324052A1 (en) 2018-10-18 2021-10-21 Merck Sharp & Dohme Corp. Formulations of anti-rsv antibodies and methods of use thereof
US20210353713A1 (en) 2018-10-26 2021-11-18 Amgen Inc. Formulations comprising a tris buffer and a protein
EP3873519A1 (en) 2018-10-29 2021-09-08 F. Hoffmann-La Roche AG Antibody formulation
AU2019372568A1 (en) 2018-10-31 2021-05-20 Richter Gedeon Nyrt Aqueous pharmaceutical formulations
EP3877409A1 (en) 2018-11-06 2021-09-15 Genmab A/S Antibody formulation
US20210380694A1 (en) 2018-11-07 2021-12-09 Merck Sharp & Dohme Corp. Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
MX2021005910A (en) 2018-11-21 2021-08-16 Regeneron Pharma High concentration protein formulation.
EP3890778A4 (en) 2018-12-03 2022-08-03 Agensys, Inc. Pharmaceutical compositions comprising anti-191p4d12 antibody drug conjugates and methods of use thereof
US20220110872A1 (en) 2018-12-14 2022-04-14 Morphosys Ag Antibody formulations
PE20211707A1 (en) 2019-01-08 2021-09-01 H Lundbeck As ACUTE TREATMENT AND RAPID TREATMENT OF HEADACHE USING ANTICGRP ANTIBODIES
JP2022523488A (en) 2019-01-25 2022-04-25 エスアイオーツー・メディカル・プロダクツ・インコーポレイテッド Common contact surfaces used in the manufacture, packaging, delivery and evaluation of biopharmaceutical products
MX2021009147A (en) 2019-01-31 2021-09-10 Hangzhou Dac Biotech Co Ltd A conjugate of an amanita toxin with branched linkers.
AU2020214626A1 (en) 2019-01-31 2021-09-16 Elektrofi, Inc. Particle formation and morphology
AU2020225202B2 (en) 2019-02-18 2023-10-26 Eli Lilly And Company Therapeutic antibody formulation
TW202102260A (en) 2019-03-21 2021-01-16 美商再生元醫藥公司 Stabilized formulations containing anti-il-33 antibodies
CN113365665A (en) 2019-03-26 2021-09-07 荣昌生物制药(烟台)股份有限公司 anti-Her 2 antibody drug conjugate pharmaceutical formulations
BR112021019612A2 (en) 2019-04-01 2021-11-30 Genentech Inc Formulations, container, article of manufacture, method for producing the formulation and method for inhibiting aggregation of a protein present in an aqueous solution
MA55559A (en) 2019-04-04 2022-02-09 Janssen Biotech Inc METHOD FOR ADMINISTRATION OF AN ANTI-IFN-ALPHA/-OMEGA ANTIBODY
SG11202110942SA (en) 2019-04-18 2021-11-29 Janssen Biotech Inc Sialylated glycoproteins
EP3962942A1 (en) 2019-05-01 2022-03-09 Novo Nordisk A/S Anti-il-6 antibody formulation
CN110179746A (en) 2019-05-17 2019-08-30 通化东宝生物科技有限公司 A kind of stable Su Jin monoclonal antibody injection and preparation method thereof
CN111939267A (en) 2019-05-17 2020-11-17 百奥泰生物制药股份有限公司 Antibody-drug conjugate preparation, preparation method and application
US11827671B2 (en) 2019-05-24 2023-11-28 Sanofi Methods for treating systemic sclerosis
CA3141988A1 (en) 2019-05-28 2020-12-03 The General Hospital Corporation Apoe antibodies, fusion proteins and uses thereof
WO2020247572A1 (en) 2019-06-05 2020-12-10 Seattle Genetics, Inc. Masked antibody formulations
CN110124030A (en) 2019-06-10 2019-08-16 通化东宝生物科技有限公司 A kind of Su Jin monoclonal antibody injection and preparation method thereof
US11655302B2 (en) 2019-06-10 2023-05-23 Sanofi Anti-CD38 antibodies and formulations
WO2020250252A1 (en) 2019-06-11 2020-12-17 Sifi S.P.A. Microemulsion compositions
CA3144784A1 (en) 2019-06-24 2020-12-30 Hangzhou Dac Biotech Co., Ltd A conjugate of a cytotoxic agent to a cell binding molecule with branched linkers
US20220273796A1 (en) 2019-07-19 2022-09-01 Ichnos Sciences SA Lyophilized antibody formulation
EP3766481A1 (en) 2019-07-19 2021-01-20 Ichnos Sciences SA Liquid antibody formulation
CN112805566A (en) 2019-07-29 2021-05-14 上海谷森医药有限公司 Antibody pharmaceutical preparation for inhalation treatment of lung cancer
EP4027978A1 (en) 2019-09-13 2022-07-20 Elektrofi, Inc. Compositions and methods for the delivery of therapeutic biologics for treatment of disease
EP4031570A1 (en) 2019-09-20 2022-07-27 Novartis AG Methods of treating autoimmune diseases using interleukin-17 (il-17) antagonists
CN110585430B (en) 2019-09-29 2023-09-08 华博生物医药技术(上海)有限公司 Pharmaceutical composition of humanized anti-human IL-17A monoclonal antibody
AU2020358101A1 (en) 2019-10-02 2022-04-28 Alamab Therapeutics, Inc. Anto-connexin antibody formulations
WO2021067820A1 (en) 2019-10-04 2021-04-08 Seagen Inc. Formulation of antibody-drug conjugate
CN112891531B (en) 2020-06-19 2021-10-08 北京东方百泰生物科技股份有限公司 Injection preparation of anti-IL-17 RA monoclonal antibody

Also Published As

Publication number Publication date
JOP20210229A1 (en) 2023-01-30
CR20210435A (en) 2021-09-20
MA55033A (en) 2021-12-29
SG11202108627SA (en) 2021-09-29
EP3927729A1 (en) 2021-12-29
US20200262911A1 (en) 2020-08-20
DOP2021000170A (en) 2021-09-30
CO2021010697A2 (en) 2021-10-29
AU2020225202A1 (en) 2021-08-12
JP7266108B2 (en) 2023-04-27
JP2022520857A (en) 2022-04-01
KR20210114989A (en) 2021-09-24
PE20212185A1 (en) 2021-11-11
CA3129901A1 (en) 2020-08-27
BR112021015034A2 (en) 2021-10-05
AU2020225202B2 (en) 2023-10-26
ECSP21060917A (en) 2021-09-30
MX2021009851A (en) 2021-09-10
CL2021002182A1 (en) 2022-03-18
UA128098C2 (en) 2024-04-03
WO2020172002A1 (en) 2020-08-27
CN113474360A (en) 2021-10-01
AU2023251529A1 (en) 2024-01-18
IL285134A (en) 2021-09-30
JP2023089190A (en) 2023-06-27
US11634485B2 (en) 2023-04-25
EP3927729A4 (en) 2023-10-11

Similar Documents

Publication Publication Date Title
US11926670B2 (en) Stabilized formulations containing anti-interleukin-4 receptor (IL-4R) antibodies
US20240150479A1 (en) Liquid Pharmaceutical Composition Comprising an Anti-IL-6 Receptor Antibody
US20230183334A1 (en) Therapeutic antibody formulation
TWI690329B (en) Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies
US20210252146A1 (en) Stable antibody formulation
EA045866B1 (en) COMPOSITION OF THERAPEUTIC ANTIBODY
US20210393779A1 (en) Activin a antibody formulations and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: ELI LILLY AND COMPANY, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CORVARI, VINCENT JOHN;PISUPATI, KARTHIK;SIGNING DATES FROM 20200102 TO 20200103;REEL/FRAME:062867/0607

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION