WO2016036678A1 - Formulations of bispecific antibodies - Google Patents

Formulations of bispecific antibodies Download PDF

Info

Publication number
WO2016036678A1
WO2016036678A1 PCT/US2015/047843 US2015047843W WO2016036678A1 WO 2016036678 A1 WO2016036678 A1 WO 2016036678A1 US 2015047843 W US2015047843 W US 2015047843W WO 2016036678 A1 WO2016036678 A1 WO 2016036678A1
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
bispecific antibody
pharmaceutical formulation
buffer
binding domain
Prior art date
Application number
PCT/US2015/047843
Other languages
French (fr)
Inventor
Sureshkumar CHOUDHARY
Arun PARUPUDI
Original Assignee
Medimmune, Llc
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 Medimmune, Llc filed Critical Medimmune, Llc
Publication of WO2016036678A1 publication Critical patent/WO2016036678A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • 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
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific

Definitions

  • the present invention provides pharmaceutical formulations of bispecific antibodies or fragments thereof that minimize aggregation.
  • the pharmaceutical formulations disclosed herein include bispecific antibodies comprising a first binding domain that binds the CD3 T cell surface antigen.
  • the pharmaceutical formulations disclosed herein comprise BiTE® molecules, including BiTE® molecules having at least a first binding site specific for CD3 and a second binding site specific for carcinoembryonic antigen (CEA) antigens (e.g. MEDI-565) or CD 19 antigens (e.g. MT103).
  • CEA carcinoembryonic antigen
  • BiTE® molecules formulated according to the methods disclosed herein are designed for parenteral administration including subcutaneous administration or injection.
  • the present invention also provides methods of determining suitable buffers and excipients for use in protein formulations, particularly for antibody formulations including bispecific antibodies.
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Bispecific antibodies may bind to two different epitopes of a single cell surface marker or they may bind a first cell surface marker and further bind a second cell surface marker on the same cell. A binding arm specific for a surface antigen of a target cell may also be combined with an arm which binds to a triggering molecule on a leukocyte such as a T cell receptor molecule (e.g., CD2 or CD3), or Fc receptors for IgG (FcyR), so as to focus cellular defense mechanisms to the target cell. Bispecific antibodies can be prepared as full-length antibodies or antibody fragments (e.g., F(ab ')).
  • MEDI-565 also known as CEA-BiTE; AMG 211 ; MT111 (SEQ ID NO:26) is a
  • CEA/CD3 bispecific single-chain antibody or BiTE® molecule reported to prevent subcutaneous tumor growth and formation of metastases in preclinical models. See, e.g., WO 07/071426, Lutterbuese et al., 2009, Journal of Immunother 32: 341-352, and Osada et al. 2010, British Journal of Cancer, 102: 124-33.
  • MEDI-565 is in phase I clinical trials (clinicaltrials.gov identifier: NCT01284231) for the treatment of gastrointestinal adenocarcinomas.
  • MT103 also known as Blinatumomab; (SEQ ID NO: 25) is a recombinant, bispecific single-chain antibody or BiTE® molecule, which binds to CD 19 on target cells and the CD3 complex on T effector cells. See Hoffmann Pet. al., Int J Cancer. 2005 May 20;115(1):98-104. It is a single polypeptide chain of 55 kDa with the variable binding domains of two murine monoclonal antibodies. By simultaneously binding to target and effector cells, MT103 triggers the T effector cell to form an immunological synapse, and to release perforin and granzymes which induce cell death of target cells.
  • Target cells comprise human B cells of all development stages (with the exception of plasma cells) and malignant cells of most B cell-derived lymphoma and leukemia.
  • MT103 is currently in development for treatment of B-cell cancers (clinicaltrials.gov identifier: NCT02013167).
  • Exemplary pharmaceutical formulations for single chain bispecific antibodies including MEDI-565 and MT103 are disclosed in U.S. Patent Publication No. 2010/0303827, herein incorporated by reference in its entirety. These pharmaceutical formulations comprise a bispecific antibody stabilized with lysine.
  • stability One major concern during the development of protein formulations is stability. Insufficient protein stability can result in aggregation, which is undesirable, as it can impact protein activity and immunogenicity. This is a particular problem when formulating single chain antibodies at ultra-low concentrations (e.g. between approximately 25 ⁇ g/ml and 500 ⁇ g/ml) since at these protein concentrations even a low level of aggregation or brief contact with vial surfaces (i.e.
  • the antibody formulations disclosed in U.S. Patent Publication No. 2010/0303827 prevent absorption loss to contact surfaces and prevent aggregation when formulated at about 25 ⁇ g/ml to about 500 ⁇ g/ml antibody concentration.
  • these formulations are generally not suitable at increased protein concentrations (about 1.0 mg/ml to about 5.0 mg/ml) as aggregation of single chain bispecific antibodies (e.g. MEDI-565 and MT103) increases significantly at these higher antibody concentrations.
  • single chain bispecific antibodies e.g. MEDI-565 and MT103
  • small increases in protein aggregation often result in super-potent antibodies unsuitable for clinical use.
  • the formulations disclosed herein provide stability by reducing antibody aggregation of single chain bispecific antibodies (e.g. MEDI-565 and MT103) at increased antibody concentrations.
  • other benefits of the formulations disclosed herein include reduced volume dosing, alternative dosing routes (e.g. IV or subcutaneous delivery) and suitably exclude lysine which can increase the osmolarity of single chain antibody formulations potentially leading to increase pain upon parenteral administration (e.g. subcutaneous or injection).
  • a formulation comprising about 1.0 mg/mL to about 5.0 mg/mL of a single chain bispecific antibody that 1) excludes lysine, 2) minimizes aggregate formation during storage (e.g. total aggregate ⁇ 2.0%), 3) maximizes stability of the drug substance and drug product, and 4) is suitable for parenteral administration including subcutaneous administration or injection.
  • the disclosed pharmaceutical formulations allow for use of high amounts of bispecific antibodies (e.g. about 1.0 mg/mL to about 5.0 mg/mL) while exhibiting increased stability as to provide liquid pharmaceutical formulations and lyophilized formulations for extended storage and shipping, prior to administration to a patient.
  • the lyophilized pharmaceutical formulations can be reconstituted in a reduced volume, i.e., about 1/2 to about 1/3 of the starting volume, so as to increase the final amount of bispecific antibody, yet still maintain the desired stability (i.e., low aggregation and/or fragmentation) and also still maintain a time of reconstitution on the order of minutes (e.g., 5-15 minutes), so as to allow for simple use in clinical settings and to increase patient and physician compliance.
  • compositions comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 1.0 mg/mL to about 5.0 mg/mL of the bispecific antibody, about 15 mM to about 60 mM citrate, about 25 mM to about 0.21 M of a lyoprotectant, and about 0.002% to about 0.1% of a surfactant, wherein the formulation has a pH of about 5.0 to about 7.0.
  • the formulations comprise about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody, or about 1.7 mg/mL or about 2.5 mg/mL of the bispecific antibody.
  • the formulations can comprise about 20 mM to about 30 mM citrate, or about 20 mM citrate or about 30 mM citrate. In embodiments, the formulations comprise about 60 mM to about 80 mM of the lyoprotectant, or about 70 mM of the lyoprotectant. Suitably, the formulations comprise about 0.008% to about 0.012% of the surfactant, suitably about 0.01% of the surfactant.
  • Exemplary lyoprotectants include, but are not limited to, trehalose, lactose, mannitol, raffinose and sucrose.
  • Exemplary surfactants include, but are not limited to, polysorbate-20, polysorbate-40, polysorbate-60 and polysorbate-80.
  • compositions comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody, about 20 mM to about 30 mM citrate, about 60 mM to about 80 mM trehalose, and about 0.002% to about 0.1% polysorbate 80, wherein the formulation has a pH of about 5.0 to about 7.0.
  • compositions comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 1.7 mg/mL of the bispecific antibody, about 20 mM citrate, about 70 mM trehalose, and about 0.01% polysorbate 80, wherein the formulation has a pH of about 6.0.
  • Additional pharmaceutical formulations comprise a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 5.0 mg/mL of the bispecific antibody, about 60 mM citrate, about 0.21 M trehalose, and about 0.03% polysorbate 80, wherein the formulation has a pH of about 6.0.
  • the pharmaceutical formulations have a volume of about 0.5 mL to about
  • the pharmaceutical formulations are administered to a subject parenterally, for example subcutaneously or by injection.
  • the formulations are a liquid formulation, a frozen formulation, a lyophilized formulation or a reconstituted formulation.
  • lyophilized pharmaceutical formulations comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 2.5 mg to about 61 mg of the bispecific antibody, about 8.5 mg to about 204 mg of citrate, about 40 mg to about 954mg trehalose and about 0.15 ⁇ g to about 3 ⁇ g of polysorbate 80, more suitably about 7 mg of the bispecific antibody, about 25 mg or about 24.95 mg citrate, about 117 mg or about 116.5 mg trehalose, and about 0.4 ⁇ g polysorbate 80.
  • Also provided are methods of preparing a pharmaceutical formulation comprising preparing a liquid pharmaceutical formulation as described herein, lyophilizing the liquid pharmaceutical formulation of to produce a lyophilized formulation, and reconstituting the lyophilized formulation in a volume suitable for parenteral administration.
  • the methods further comprise freezing the liquid pharmaceutical formulation (e.g., to at least about -40° C or at least about -80° C) prior to the lyophilizing.
  • the lyophilized formulations are reconstituted in about 1/2 to about 1/3 volume of the liquid pharmaceutical formulation, suitably in a volume of about 0.5 mL to about 12.0 mL, e.g., in a volume of about 1.4 mL, or a volume suitable for subcutaneous administration or injection.
  • the pharmaceutical formulations disclosed herein contain a bispecific antibody wherein the first binding domain comprises SEQ ID NO: 1, SEQ ID NO: 30 or SEQ ID NO: 31.
  • the second binding domain specifically binds an antigen selected from the group consisting of: CD19, CD20, CD22, EphA2, EphA4, INFR, ICOS, Ep-CAM, CEA, and IL-5 receptor.
  • the second binding domain comprises the amino acid sequence of any one of SEQ ID NOs: 2-24 and 28-29
  • the bispecific antibody comprises SEQ ID NO: 26
  • the second binding domain specifically binds CD19
  • the second binding domain comprises SEQ ID NO: 25
  • the bispecific antibody comprises SEQ ID NO: 27.
  • Also provided are methods for selecting a buffer or an excipient for use in a protein formulation or for selecting a buffer or excipient suitable to reduce or prevent protein aggregation in a protein formulation the method comprising providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient, wherein the concentration of the protein is the same in each buffer solution, and wherein the concentration of the buffer or the excipient in each buffer solution is the same, but the buffer or the excipient in each buffer solution is different, measuring a light scattering intensity of each of the plurality of buffer solutions, constructing a Debye plot, determining a second virial coefficient of each of the plurality of buffer solutions, and comparing the second virial coefficient of each of the plurality of buffer solutions, wherein a more positive second virial coefficient indicates a buffer or excipient for use in the protein formulation.
  • methods for selecting a concentration of a buffer or an excipient for use in a protein formulation, or for selecting a buffer or excipient concentration suitable to reduce or prevent protein aggregation in a protein formulation, the method comprising providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient, wherein the concentration of the protein is the same in each buffer solution, and wherein the concentration of the buffer or the excipient in each buffer solution is different, but the buffer or the excipient in each buffer solution is the same, measuring a light scattering intensity of each of the plurality of buffer solutions, constructing a Debye plot, determining a second virial coefficient of each of the plurality of buffer solutions, comparing the second virial coefficient of each of the plurality of buffer solutions, wherein a more positive second virial coefficient indicates the concentration of the buffer or the excipient for use in the protein formulation.
  • the protein is an antibody or antigen-binding fragment thereof, a peptide or a fusion protein, including a bispecific antibody comprising a first and a second binding domain, and wherein the first binding domain specifically binds the CD3 T cell surface antigen, including the sequences recited herein.
  • the buffer is selected from the group consisting of citrate, succinate, phosphate, histidine, acetate, TRIS and combinations thereof, the concentration of the buffer or the excipient is from about 5 mM to about 200 mM, suitably less than about 100 mM. In additional embodiments, the excipient is a sugar.
  • FIG 1A shows a schematic representation of a bispecific antibody comprising a
  • CD3 binding domain and a carcinoembryonic antigen binding domain.
  • FIG. IB shows a schematic representation of a bispecific antibody comprising a
  • CD3 binding domain and a carcinoembryonic antigen binding domain targeting and killing tumor cells targeting and killing tumor cells.
  • FIG. 1C shows the aggregation propensity of a bispecific antibody (MEDI-565; circles) compared to a monoclonal, whole IgGl antibody (squares) incubated at 25 °C as measured by High Pressure Size Exclusion Chromatography (HPSEC).
  • the bispecific antibody (MEDI-565) aggregates at very high rate compared to the monoclonal antibody.
  • FIG. 2 shows the effect of different buffers on the stability ( Aggregate) of a bispecific antibody (MEDI-565) at ⁇ 5mg/mL incubated at 30°C for 15 days as measured by High Pressure Size Exclusion Chromatography (HPSEC).
  • HPSEC High Pressure Size Exclusion Chromatography
  • FIG. 3 shows the effect of different buffers on the stability (Aggregation rate/day at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) compared to the second virial coefficient (B 22 ) for each of the compositions.
  • the left Y axis represents aggregation rate per day at 30°C and the right Y axis represents second virial coefficient (B 22 ) in lOe "4 mol.ml per g 2 .
  • the bars from left to right represent citrate, phosphate, succinate and histidine, respectively.
  • Citrate buffer had the lowest aggregation rate and a less negative B 22 compared to other buffers.
  • mol Moles
  • ml milliliter
  • g gram.
  • FIG. 4A shows the impact of citrate concentration on the stability ( Aggregate at
  • FIG. 4B shows the impact of citrate concentration on the stability (Aggregation rate at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) at ⁇ 5 mg/mL compared to the second virial coefficient for the compositions.
  • the left Y axis represents aggregation rate per day at 30°C and the right Y axis represents second virial coefficient (B 22 ) in lOe "4 mol.ml per g 2 .
  • the bars from left to right represent increasing citrate concentration (lOmM, 20mM, 30mM, 50mM and lOOmM, respectively).
  • the lowest aggregation rate (lOOmM citrate) corresponded to the formulation having the most positive B 22 .
  • mol Moles
  • ml milliliter
  • g gram.
  • FIG. 5 shows the impact of protein concentration on the stability ( Aggregate at
  • FIG. 6 shows the stability ( Aggregate at 25 °C as measured by High Pressure
  • FIG. 7 A shows the stability (% Aggregate at 5°C as measured by High Pressure
  • FIG. 7B shows the stability (% Aggregate at 40°C as measured by High Pressure
  • FIG. 8A shows the impact of succinate concentration on the stability (%
  • FIG. 8B shows the impact of succinate concentration on the stability (Aggregation rate/day at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) at ⁇ 5 mg/mL compared to the second virial coefficient for the compositions.
  • the left Y axis represents aggregation rate per day at 30°C and the right Y axis represents second virial coefficient (B 22 ) in lOe "4 mol.ml per g 2 .
  • the bars from left to right bar represent increasing succinate concentration (lOmM, 30mM, 30mM and lOOmM, respectively).
  • the lowest aggregation rate (100 mM) corresponded to the formulation having the most positive B 22 .
  • FIG. 9A shows stability (% Aggregate as measured by High Pressure Size
  • FIG. 9B shows stability (% Aggregate as measured by High Pressure Size
  • FIG. 10 shows the stability (% Aggregate at -80°C as measured by High Pressure
  • FIG. 11 shows stability (% Aggregate at 25°C as measured by High Pressure Size
  • FIG. 12 shows % relative activity relative to a reference standard of a bispecific antibody formulation (1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS- 80, pH 6.0 (drug substance) or 5mg/mL MEDI-565, 59 mM citrate, 0.21 M trehalose, 0.03% polysorbate 80, pH 6.0 (post-reconstituted drug product)) after storage for up to 27 months. No significant changes to the in vitro activity of frozen drug substance (circle), lyophilized drug product stored at 2-8°C (square) and lyophilized drug product stored at 40°C (triangle) relative to a reference standard was observed.
  • a bispecific antibody formulation 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS- 80, pH 6.0 (drug substance) or 5mg/mL MEDI-565, 59 mM citrate, 0.21 M trehalose,
  • the term “and/or” as used in a phrase such as “A and/or B” is intended to include “A and B,” “A or B,” “A” (alone), and “B” (alone).
  • the term “and/or” as used in a phrase such as "A, B, and/or C” is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
  • the terms "stability” and “stable” as used herein in the context of a formulation comprising BiTE® molecules refer to the resistance of the antibody (including antibody fragment thereof) in the formulation to aggregation, degradation or fragmentation under given manufacture, preparation, transportation and storage conditions.
  • the “stable” formulations of the invention retain biological activity under given manufacture, preparation, transportation and storage conditions.
  • the stability of said antibody can be assessed by degrees of aggregation, degradation or fragmentation, as measured by HPSEC, static light scattering (SLS), Fourier Transform Infrared Spectroscopy (FTIR), circular dichroism (CD), urea unfolding techniques, intrinsic tryptophan fluorescence, differential scanning calorimetry, and/or ANS binding techniques, compared to a reference formulation.
  • the overall stability of a formulation comprising BiTE® molecules can be assessed by various immunological assays including, for example, ELISA and radioimmunoassay using isolated antigen molecules.
  • isolated when used to describe the various polypeptides and antibodies disclosed herein, means a polypeptide or antibody that has been identified, separated and/or recovered from a component of its production environment.
  • the isolated polypeptide is free of association with all other components from its production environment.
  • Contaminant components of its production environment such as that resulting from recombinant transfected cells, are materials that would typically interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the polypeptide will be purified (1) to a degree sufficient to obtain at least 15 residues of N- terminal or internal amino acid sequence by use of a spinning cup sequenator, or (2) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or silver stain.
  • an isolated antibody will be prepared by at least one purification step.
  • excipient refers to an inert substance which is commonly used as a diluent, vehicle, preservative, binder or stabilizing agent for drugs which imparts a beneficial physical property to a formulation, such as increased protein stability, increased protein solubility, and decreased viscosity.
  • excipients include, but are not limited to, proteins (for example, but not limited to, serum albumin), surfactants (for example, but not limited to, SDS, Tween 20, Tween 80, polysorbate, polysorbate 80 and nonionic surfactants), saccharides (for example, but not limited to, glucose, sucrose, maltose and trehalose), polyols (for example, but not limited to, mannitol and sorbitol), fatty acids and phospholipids (for example, but not limited to, alkyl sulfonates and caprylate).
  • proteins for example, but not limited to, serum albumin
  • surfactants for example, but not limited to, SDS, Tween 20, Tween 80, polysorbate, polysorbate 80 and nonionic surfactants
  • saccharides for example, but not limited to, glucose, sucrose, maltose and trehalose
  • polyols for example, but not limited to, mannitol and sorbito
  • trehalose refers to anhydrous trehalose or trehalose dihydrate.
  • lyophilized or “freeze-dried” includes a state of a substance that has been subjected to a drying procedure such as lyophilization, where at least 50% of moisture has been removed.
  • the phrase "bulking agent” includes a compound that is pharmaceutically acceptable and that adds bulk to a lyo cake.
  • Bulking agents known to the art include, for example, carbohydrates, including simple sugars such as dextrose, ribose, fructose and the like, alcohol sugars such as mannitol, inositol and sorbitol, disaccharides including trehalose, sucrose and lactose, naturally occurring polymers such as starch, dextrans, chitosan, hyaluronate, proteins (e.g., gelatin and serum albumin), glycogen, and synthetic monomers and polymers.
  • carbohydrates including simple sugars such as dextrose, ribose, fructose and the like, alcohol sugars such as mannitol, inositol and sorbitol, disaccharides including trehalose, sucrose and lactose, naturally occurring polymers such as starch, dextrans, chitosan,
  • compositions comprising a bispecific antibody.
  • antibody and “antibodies” encompass monoclonal antibodies (including full-length monoclonal antibodies), polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, human antibodies, humanized antibodies, camelised antibodies, chimeric antibodies, single-chain Fvs (scFv), single-chain antibodies, single domain antibodies, domain antibodies, Fab fragments, F(ab')2 fragments, antibody fragments that exhibit the desired biological activity, disulfide-linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), intrabodies, and epitope- binding fragments of any of the above.
  • Antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen-binding site.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass.
  • bispecific antibody As used herein, "bispecific antibody,” “bispecific single chain antibody” or
  • BiTE ® refer to an antibody comprising a first binding domain and a second binding domain that comprises a variable heavy (VH) domain and a variable light (VL) domain of an antibody.
  • bispecific antibodies for use in the formulations and methods described herein comprise a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the bispecific antibodies described herein comprise a second binding domain that comprises a VH domain and a VL domain of an antibody that immunospecifically binds to an antigen of interest (e.g., CD 19 or EpbA2 or CEA).
  • the first and second binding domains of a bispecific antibody may be derived from synthetic antibodies, monoclonal antibodies, recombinantly produced antibodies, multispecific antibodies (including bi- specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, intrabodies, single-chain Fvs (scFvs) (e.g., including monospecific and bi-specific, etc.), Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • scFvs single-chain Fvs
  • sdFv single-chain Fvs
  • sdFv disulfide-linked Fvs
  • anti-Id anti-idiotypic antibodies
  • the formulations provided herein comprise a bispecific single chain antibody comprising a first binding domain that comprises a variable heavy (VH) domain and a variable light (VL) domain of an antibody that immunospecifically binds to the T-cell antigen CD3 and a second binding domain that comprises a VH domain and a VL domain of an antibody that immunospecifically binds to carcinoembryonic antigen (CEA).
  • Carcinoembryonic antigen (CEA) is a glycosylated human oncofetal antigen that belongs to the CEA-related cell adhesion molecule (CEACAM) family of the immunoglobulin gene superfamily.
  • CEA has been suggested to mediate cell-cell adhesion, facilitate bacterial colonization of the intestine, and protect the colon from microbial infection by binding and trapping infectious microorganisms.
  • Carcinoembryonic antigen (CEA) is a well-characterized tumor-associated antigen that is frequently over-expressed in human carcinomas and melanomas.
  • FIG. 1A shows a schematic representation of a bispecific antibody described herein comprising a first binding domain that binds to CD3 and a second binding domain that binds to CEA.
  • FIG. IB shows a schematic representation of a bispecific antibody comprising a CD3 binding domain and a carcinoembryonic antigen binding domain targeting and killing tumor cells.
  • FIG. 1C shows the aggregation propensity of a bispecific antibody (MEDI-565; circles) compared to a monoclonal, whole IgGl antibody (squares) incubated at 25 °C as measured by HPSEC.
  • bispecific antibody aggregates at a very high rate compared to a monoclonal IgGl antibody, underscoring the difficulties in formulating stable single chain, bi-specific antibodies (e.g. MEDI-565, MT103) that are addressed herein.
  • the antibody that immunospecifically binds to the T-cell antigen CD3 and the antibody that immunospecifically binds CEA may be derived from synthetic antibodies, monoclonal antibodies, recombinantly produced antibodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, intrabodies, single- chain Fvs (scFvs) (e.g., including monospecific and bi- specific, etc.), Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • antibodies of the formulations described herein include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen-binding domain that immunospecifically binds to an antigen of interest.
  • binding domain refers to a domain comprising a three- dimensional structure capable of immunospecifically binding to an epitope.
  • said domain can comprise the VH and/or VL domain of an antibody chain.
  • said domain comprises at least the VH domain.
  • said binding domains contained in the bispecific antibodies of a formulation of the invention may comprise at least one complementarity determining region (CDR) of an antibody chain recognizing the CD 19 and CEA antigens, respectively.
  • CDR complementarity determining region
  • single-chain Fv refers to antibody fragments comprising the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding.
  • Methods for producing scFvs are well known in the art. For a review of methods for producing scFvs see Pluckthun, in "The Pharmacology of Monoclonal Antibodies," vol. 113, Rosenburg and Moore eds. Springer- Verlag, New York, pp. 269-315 (1994).
  • compositions of antibodies and/or antibody fragments that specifically bind to an antigen of interest are herein collectively referred to as "pharmaceutical formulations of the invention,” “formulations of the invention”, “liquid formulations of the invention”, “lyophilized formulations of the invention”, “reconstituted formulations of the invention”, “antibody liquid formulations of the invention”, “antibody lyophilized formulations of the invention”, “antibody reconstituted formulations of the invention” or “antibody formulations of the invention”.
  • the phrases "pharmaceutical formulation” and “formulation” are used interchangeably and refer to a composition comprising a bispecific antibody and one or more appropriate buffers and/or excipients.
  • the pharmaceutical formulations described herein are “pharmaceutically acceptable,” and thus would meet the necessary approval requirements required by a regulatory agency of the Federal or a state government, or listed in the U.S. Pharmacopeia, European Pharmacopeia, or other generally recognized pharmacopeia, so as to be used in animals, and more particularly in humans.
  • the pharmaceutical formulations described herein comprise about 1.0 mg/mL to about 10.0 mg/mL of a bispecific antibody, more suitably about 1.0 mg/mL to about 5.0 mg/mL of a bispecific antibody.
  • the formulations described herein comprise about 1.0 mg/mL to about 7 mg/mL, about 1.0 mg/mL to about 6 mg/mL, about 1.0 mg/mL to about 5 mg/mL, about 1.0 mg/mL to about 4 mg/mL, about 1.0 mg/mL to about 3.5 mg/mL, about 1.0 mg/mL to about 3 mg/mL, about 1.5 mg/mL to about 3 mg/mL, about 1.5 mg/mL to about 2.5 mg/mL, about 1.7 mg/mL to about 3 mg/mL, about 1.7 mg/mL to about 2.5 mg/mL, or about 1.0 mg/mL, about 1.1 mg/mL, about 1.2 mg/mL, about 1.3 mg/mL, about 1.4 mg/mL, about 1.5 mg/mL, about 1.6 mg/mL, about 1.7 mg/mL, about 1.8 mg/mL, about 1.9 mg/mL, about 2.0 mg/mL, about 2.1 mg/mL, about 1.0
  • the pharmaceutical formulations described herein suitably further comprise one or more buffers.
  • buffer refers to an excipient for maintaining the pH of a formulation.
  • Exemplary buffers for use in the pharmaceutical formulations provided herein include, but are not limited to histidine, citrate, phosphate, succinate, glycine, and acetate.
  • the buffer for use in the pharmaceutical formulations described herein is citrate.
  • the concentration of a buffer, suitably citrate, in the pharmaceutical formulations described herein is generally in the range of about 10 mM to about 100 mM, more suitably about 15 mM to about 80 mM, about 15 mM to about 60 mM, about 20 mM to about 60 mM, about 20 mM to about 50 mM, about 20 mM to about 40 mM, about 20 mM to about 30 mM, or about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM or about 60 mM, including any ranges or values within these ranges.
  • the pharmaceutical formulations provided herein also suitably further comprise a lyoprotectant.
  • a "lyoprotectant” refers to a molecule which, when amorphous and combined with a protein (e.g., an antibody such as a bispecific antibody), provides retention of a protein's physical and chemical stability and integrity upon lyophilization and storage, and when crystallized, a lyoprotectant acts as a bulking agent.
  • Lyoprotectants include, but are not limited to, sugars and their corresponding sugar alcohols; amino acids such as monosodium glutamate or histidine; a methylamine such as betaine; a lyotropic salt such as magnesium sulfate; a polyol such as trihydric or higher molecular weight sugar alcohols, e.g. glycerin, dextran, erythritol, glycerol, arabitol, xylitol, sorbitol, and mannitol; propylene glycol; polyethylene glycol; Pluronics ® ; and combinations thereof.
  • amino acids such as monosodium glutamate or histidine
  • a methylamine such as betaine
  • a lyotropic salt such as magnesium sulfate
  • a polyol such as trihydric or higher molecular weight sugar alcohols, e.g. glycerin, dextran, erythritol, gly
  • lyoprotectants include, but are not limited to, glycerin and gelatin, and the sugars mellibiose, melezitose, raffinose, mannotriose and stachyose.
  • reducing sugars include, but are not limited to, glucose, maltose, lactose, maltulose, iso-maltulose and lactulose.
  • non-reducing sugars include, but are not limited to, trehalose, non-reducing glycosides of polyhydroxy compounds selected from sugar alcohols and other straight chain polyalcohols.
  • sugar alcohols include, but are not limited to, monoglycosides, compounds obtained by reduction of disaccharides such as lactose, maltose, lactulose and maltulose.
  • the glycosidic side group can be either glucosidic or galactosidic.
  • Additional examples of sugar alcohols include, but are not limited to, glucitol, maltitol, lactitol and iso-maltulose.
  • trehalose is used as a lyoprotectant in the formulations described herein. [0071]
  • the lyoprotectant is added to the pharmaceutical formulations in a
  • lyoprotecting amount which means that, following lyophilization of the protein (bispecific antibody) in the presence of the lyoprotecting amount of the lyoprotectant, the protein essentially retains its physical and chemical stability and integrity upon lyophilization and storage.
  • the lyoprotectant is provided in the pharmaceutical formulations at a concentration of about 20 mM to about 300 mM, more suitably, about 25 mM to about 0.21 M, for example, about 30 mM to about 0.21 M, about 40 mM to about 0.21 M, about 60 mM to about 0.21 M, about 60 mM to about 150 mM, about 60 mM to about 100 mM, about 60 mM to about 80 mM, or about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, about 60 mM, about 65 mM, about 70 mM, about 75 mM, about 80 mM, about 85 mM, about 90 mM, about 95 mM, about 100 mM, about 125 mM, about 150 mM, about 200 mM, or about 0.21 M, including any ranges
  • the pharmaceutical formulations also further comprise a surfactant.
  • surfactant refers to organic substances having amphipathic structures; namely, they are composed of groups of opposing solubility tendencies, typically an oil- soluble hydrocarbon chain and a water-soluble ionic group. Surfactants can be classified, depending on the charge of the surface-active moiety, into anionic, cationic, and nonionic surfactants. Surfactants are often used as wetting, emulsifying, solubilizing, and dispersing agents for various pharmaceutical formulations and preparations of biological materials. Pharmaceutically acceptable surfactants like polysorbates (e.g. polysorbates 20, 40, 60 or 80); polyoxamers (e.g.
  • poloxamer 188 Triton; sodium octyl glycoside; lauryl-, myristyl-, linoleyl- , or stearyl-sulfobetaine; lauryl-, myristyl-, linoleyl- or stearyl-sarcosine; linoleyl-, myristyl-, or cetyl-betaine; lauroamidopropyl-, cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-betaine (e.g.
  • the pharmaceutical formulations described herein can be used in the pharmaceutical formulations described herein.
  • the surfactant is polysorbate 80.
  • the pharmaceutical formulations described herein comprise a surfactant
  • a surfactant for example about 0.005% to about 0.2%, about 0.002% to about 0.05%, about 0.006% to about 0.1%, about 0.007% to about 0.08%, about 0.008% to about 0.07%, about 0.009% to about 0.05%, about 0.01% to about 0.05%, about 0.01% to about 0.04%, about 0.01% to about 0.03%, or about 0.002%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.010%, about 0.011%, about 0.012%, about 0.013%, about 0.014%, about 0.015%, about 0.016%, about 0.017%, about 0.018%, about 0.019%, about 0.020%, about 0.021%, about 0.022%, about 0.023%, about 0.024%, about 0.025%, about 0.026%, about 0.027%, about 0.028%, about 0.019%, about 0.020%, about 0.021%, about 0.022%, about 0.023%, about 0.024%, about 0.02
  • the pharmaceutical formulations described herein can also include one or more additional excipients, including for example, one or more sugars, salts, amino acids, polyols, chelating agents, emulsifiers and/or preservatives.
  • additional excipients including for example, one or more sugars, salts, amino acids, polyols, chelating agents, emulsifiers and/or preservatives.
  • the formulations provided herein are formulated at a pharmaceutically acceptable pH, for example, in the range of about pH 5.0 to about 7.0, more suitably about pH 6.0 to about pH 7.0, or about pH 5.0, pH 5.5, pH 6.0, pH 6.5 or pH 7.0.
  • liquid, lyophilized and/or frozen pharmaceutical formulations allow for use of high amounts of bispecific antibodies while exhibiting stability for extended storage and shipping, prior to administration to a patient.
  • the pharmaceutical formulations described herein comprise about 1.7 mg/mL to about 2.5 mg/mL of a bispecific antibody, including for example about 1.7 mg/mL or about 2.5 mg/mL of the bispecific antibody.
  • the pharmaceutical formulations comprise about 20 mM to about 30 mM citrate, including for example about 20 mM citrate or about 30 mM citrate.
  • the pharmaceutical formulations also suitably further comprise about 60 mM to about 80 mM of the lyoprotectant, including for example 70 mM of the lyoprotectant.
  • the pharmaceutical formulations also suitably further comprise about 0.008% to about 0.012% of the surfactant, including for example about 0.01% of the surfactant.
  • suitable lyoprotectants for use in the pharmaceutical formulations include, but are not limited to, trehalose, lactose, mannitol, raffinose and sucrose.
  • Suitable surfactants include, but are not limited to, polysorbate-20, polysorbate- 40, polysorbate-60 and polysorbate-80.
  • compositions which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the formulations comprise about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody, about 20 mM to about 30 mM citrate, about 60 mM to about 80 mM trehalose, and about 0.002% to about 0.1% polysorbate 80.
  • the formulations have a pH of about 5.0 to about 7.0.
  • compositions which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the formulations comprise about 1.7 mg/mL of the bispecific antibody, about 20 mM citrate, about 70 mM trehalose, and about 0.01% polysorbate 80.
  • the formulations have a pH of about 6.0.
  • compositions which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the formulations comprise about 2.5mg/ml of the bispecific antibody, about 30mM Citrate, about 80mM Trehalose and about 0.01% polysorbate 80.
  • the formulations have a pH of about 6.0.
  • compositions which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the formulations comprise about 5.0 mg/mL of the bispecific antibody, about 60 mM citrate, about 0.21 M trehalose, and about 0.03% polysorbate 80.
  • the formulation has a pH of about 6.0.
  • compositions which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the formulations comprise the components described in Table 2 or Figures 5 and 6 (as described in Example 5), in particular, 2.5mg/ml, 30mM Citrate, 80mM Trehalose, 0.01% polysorbate 80, pH 6; 1.7mg/ml, 20mM Citrate, 70mM Trehalose, 0.01% polysorbate 80, pH 6; or 1.7mg/ml, 30mM Citrate, 60mM Trehalose, 0.01% polysorbate 80, pH 6.
  • the pharmaceutical formulations described herein can have any desired volume for shipping, storage and/or administration to a patient.
  • the volume of the pharmaceutical formulations are provided such that they are easily produced in mass quantities, placed in suitable vials or containers for storage and then shipped to a distribution center and/or directly to a hospital, clinic, doctor's office, or other setting where the pharmaceutical formulations can be administered to a patient.
  • the formulations are prepared in sterile water or water for injection at the desired volume.
  • the pharmaceutical formulations have a volume of about 0.1 mL to about 20.0 mL, more suitably about 0.5 mL to about 15.0 mL, about 0.5 mL to about 12.0 mL, about 1.0 mL to about 10.0 mL, about 1.0 mL to about 5.0 mL, about 1.0 mL to about 2.0 mL or about 0.5 mL, about 0.6 mL, about 0.7 mL, about 0.8 mL, about 0.9 mL, about 1.0 mL, about 1.1 mL, about 1.2 mL, about 1.3 mL, about 1.4 mL, about 1.5 mL, about 1.6 mL, about 1.7 mL, about 1.8 mL, about 1.9 mL, about 2.0 mL, about 2.1 mL, about 2.2 mL, about 2.3 mL, about 2.4 mL, about 2.5 mL, about 2.6 mL
  • the pharmaceutical formulations described herein are liquid formulations, i.e., pharmaceutical formulations prepared in sterile water or water for injection (WFI), the pharmaceutical formulations can also be frozen formulations or lyophilized formulations.
  • WFI water for injection
  • frozen formulations by providing a liquid pharmaceutical formulation as described herein, and freezing the formulation under appropriate conditions.
  • the frozen formulations can be provided by freezing the liquid formulations to less than 0° C, more suitably to about -20° C, about -40° C, about -60° C, or suitably to about -80° C.
  • Suitable protocols and methods for preparing lyophilized pharmaceutical formulations from liquid and/or frozen formulations are known in the art and described herein.
  • lyophilized pharmaceutical formulations are provided.
  • such formulations have been prepared from the liquid and/or frozen formulations provided throughout by removing water from the formulations to yield a freeze-dried formulation.
  • the lyophilized pharmaceutical formulations provided herein are described as containing recited weight amounts of components, indicating dried formulations, rather than a liquid formulation.
  • lyophilized pharmaceutical formulations which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen.
  • the lyophilized pharmaceutical formulations comprise about 2 mg to about 10 mg of the bispecific antibody, about 15 mg to about 40 mg citrate, about 100 mg to about 200 mg trehalose and about 0.01 ⁇ g to about 0.08 ⁇ g polysorbate 80.
  • the lyophilized pharmaceutical formulations comprise about 3 mg to about 10 mg of the bispecific antibody, or about 3 mg to about 8 mg, about 5 mg to about 8 mg, or about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg or about 10 mg of the bispecific antibody, including any ranges or values within these ranges.
  • the lyophilized formulations comprise about 20 mg to about 30 mg citrate, or about 20 mg, about 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg or about 30 mg citrate, including any ranges or values within these ranges.
  • the lyophilized pharmaceutical formulations comprise about 100 mg to about 180 mg trehalose, or about 100 mg to about 150 mg, or about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg or about 150 mg trehalose, including any ranges or values within these ranges.
  • the lyophilized pharmaceutical formulations comprise about 0.02 ⁇ g to about 0.08 ⁇ g polysorbate 80, or about 0.03 ⁇ g to about 0.06 ⁇ g, or about 0.02 ⁇ g, about 0.03 ⁇ g, about 0.04 ⁇ g, about 0.05 ⁇ g, about 0.06 ⁇ g, about 0.07 ⁇ g, or about 0.08 ⁇ g polysorbate 80, including any ranges or values within these ranges.
  • the lyophilized pharmaceutical formulations comprise about about 2.5 mg to about 61 mg of the bispecific antibody, about 8.5 mg to about 204 mg of citrate, about 40 mg to about 954mg trehalose and about 0.15 ⁇ g to about 3.6 ⁇ g of polysorbate 80, more suitably about 7 mg of the bispecific antibody, about 25 mg or about 24.95 mg citrate, about 117 mg or about 116.5 mg trehalose, and about 0.4 ⁇ g polysorbate 80.
  • the lyophilized pharmaceutical formulations are designed to provide enhanced stability and limited aggregation upon reconstitution.
  • a "reconstituted" formulation is one which has been prepared by dissolving a lyophilized pharmaceutical formulation in a diluent such that the bispecific antibody is dispersed in the reconstituted formulation (i.e., the act of reconstitution).
  • the reconstituted formulation is suitable for administration (e.g. parenteral administration) to a patient to be treated with the bispecific antibody of interest and, in certain embodiments of the invention, may be one which is suitable for parenteral administration, including for example subcutaneous administration or injection.
  • diluent of interest herein is one which is pharmaceutically acceptable (safe and nontoxic for administration to a human) and is useful for the preparation of a liquid pharmaceutical formulation, such as a pharmaceutical formulation reconstituted after lyophilization.
  • diluents include, but are not limited to, sterile water, bacteriostatic water for injection (BWFI), water for injection (WFI), a pH buffered solution (e.g. phosphate -buffered saline), sterile saline solution, Ringer's solution or dextrose solution, though suitably water (sterile, WFI or BWFI) is used.
  • An "isotonic" pharmaceutical formulation is one which has essentially the same osmotic pressure as human blood. Isotonic pharmaceutical formulations generally have an osmotic pressure from about 250 to 350 mOsm.
  • the term “hypotonic” describes a pharmaceutical formulation with an osmotic pressure below that of human blood.
  • the term “hypertonic” is used to describe a pharmaceutical formulation with an osmotic pressure above that of human blood. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer.
  • the lyophilized pharmaceutical formulations described herein are stable for extended periods of storage at room temperature.
  • room temperature is generally in the range of about 22-25° C.
  • the lyophilized pharmaceutical formulations are also suitably stable after storage at 25° C for 6 months or more.
  • the lyophilized pharmaceutical formulations are stable after storage at 2-8° C (e.g. 5° C) for 24 months or more.
  • the term “stable” for a period of storage is used to indicate that the bispecific antibodies in the lyophilized pharmaceutical formulations resist aggregation, degradation and/or fragmentation following reconstitution; or the bispecific antibodies in the liquid pharmaceutical formulations resist aggregation, degradation and/or fragmentation.
  • the stability of the bispecific antibodies can be assessed by degrees of aggregation, degradation or fragmentation, as measured by high performance size exclusion chromatography (HPSEC), static light scattering (SLS), Fourier Transform Infrared Spectroscopy (FTIR), circular dichroism (CD), urea unfolding techniques, intrinsic tryptophan fluorescence, differential scanning calorimetry, and/or ANS binding techniques, compared to a reference.
  • the overall stability of a pharmaceutical formulation comprising bispecific antibodies can be assessed by various immunological assays including, for example, ELISA and radioimmunoassay using isolated antigen molecules.
  • low to undetectable levels of aggregation refers to pharmaceutical formulations containing no more than about 5%, no more than about 4%, no more than about 3%, no more than about 2%, no more than about 1%, or no more than about 0.5% aggregation by weight of protein as measured by high performance size exclusion chromatography (HPSEC) or static light scattering (SLS) techniques.
  • HPSEC high performance size exclusion chromatography
  • SLS static light scattering
  • the lyophilized pharmaceutical formulations exhibit ⁇ 5.0% aggregation when reconstituted, more suitably ⁇ 4.0% aggregation when reconstituted, ⁇ 3.0% aggregation when reconstituted, ⁇ 2.0% aggregation when reconstituted, ⁇ 1.0% aggregation when reconstituted, or 0.5% aggregation when reconstituted.
  • the liquid pharmaceutical formulations exhibit ⁇ 5.0% aggregation, more suitably ⁇ 4.0% aggregation, ⁇ 3.0% aggregation, ⁇ 2.0% aggregation, ⁇ 1.0% aggregation, or 0.5% aggregation.
  • low to undetectable levels of fragmentation refers to pharmaceutical formulations containing equal to or more than about 80%, about 85%, about 90%, about 95%, about 98% or about 99% of the total bispecific antibody, for example, in a single peak as determined by HPSEC, or reduced Capillary Gel Electrophoresis (rCGE), representing the non-degraded bispecific antibody or a non- degraded fragment thereof, and containing no other single peaks having more than about 5%, more than about 4%, more than about 3%, more than about 2%, more than about 1%, or more than about 0.5% of the total bispecific antibody in each.
  • rCGE reduced Capillary Gel Electrophoresis
  • the pharmaceutical formulations described herein consist of the recited components, such that no additional components beyond those recited are allowed.
  • the pharmaceutical formulations described herein consist essentially of the recited components.
  • the addition of components that alter the stability of the bispecific antibody are considered a material alteration to such pharmaceutical formulations and are thus excluded from such methods that consist essentially of the recited components.
  • bispecific antibodies including, e.g., MEDI-565 and MT103
  • This allows the preparation of pharmaceutical formulations containing amounts of bispecific antibodies in the 1.7 mg/L to 5 mg/mL range, and for example, bispecific antibodies (including, e.g., MEDI-565 and MT103) at 5 mg/mL, so as to allow for parenteral administration, including subcutaneous administration or injection if a large amount of bispecific antibody.
  • methods are provided for preparing pharmaceutical formulations, as well as pharmaceutical formulations prepared by such methods.
  • Such methods suitably comprise preparing a liquid pharmaceutical formulation in accordance with the various embodiments described herein, suitably comprising a bispecific antibody (e.g., at about 1.0 mg/mL to about 5.0 mg/mL of the bispecific antibody), citrate (e.g., about 15 mM to about 60 mM citrate), a lyoprotectant (e.g., about 25 mM to about 0.21 M of a lyoprotectant) and a surfactant (e.g., about 0.002% to about 0.1% of a surfactant).
  • the liquid pharmaceutical formulation is then lyophilized to produce a lyophilized formulation.
  • the lyophilized formulation is then reconstituted in a volume suitable for parenteral administration.
  • the methods of preparation can also further comprise freezing the liquid pharmaceutical formulation (e.g., to a temperature of less than 0° C, more suitably to about -20° C, about -40° C, about -60° C, or suitably to about -80° C), prior to lyophilizing the formulation.
  • freezing the liquid pharmaceutical formulation e.g., to a temperature of less than 0° C, more suitably to about -20° C, about -40° C, about -60° C, or suitably to about -80° C
  • the pharmaceutical formulations can be frozen (e.g., to about -80 C) at a first facility, shipped in the frozen state (if desired), thawed and then lyophilized at a later time in the first facility or in a second facility (i.e., days to weeks to months or even a year).
  • the methods described herein suitably utilize a reconstituting volume, i.e., the volume of desired diluent such as sterile water or WFI, that is on the order of about 1/2 to about 1/3 of the volume of the initial, starting liquid pharmaceutical formulation that was prepared and then lyophilized.
  • a reconstituting volume i.e., the volume of desired diluent such as sterile water or WFI, that is on the order of about 1/2 to about 1/3 of the volume of the initial, starting liquid pharmaceutical formulation that was prepared and then lyophilized.
  • the lyophilized pharmaceutical formulations can be reconstituted in a reduced volume, i.e., about 1/2 to about 1/3 of the starting volume, so as to increase the final amount of bispecific antibody, yet still maintain the desired stability (i.e., low aggregation and/or fragmentation) and also still maintain a time of reconstitution on the order of minutes (e.g., 5-15 minutes), so as to allow for simple use in clinical settings and to increase patient and physician compliance.
  • the total amount of volume for the reconstitution of the lyophilized pharmaceutical formulations is on the order of about 0.5 mL to about 12.0 mL, for example, about 0.5 mL to about 10.0 mL, about 0.5 mL to about 8.0 mL, about 0.5 mL to about 6.0 mL, about 0.5 mL to about 5.0 mL, about 0.5 mL to about 4.0 mL, about 0.5 mL to about 3.0 mL, about 0.5 mL to about 2.0 mL, or about 0.5 mL, about 0.6 mL, about 0.7 mL, about 0.8 mL, about 0.9 mL, about 1.0 mL, about 1.1 mL, about 1.2 mL, about 1.3 mL, about 1.4 mL, about 1.5 mL, about 1.6 mL, about 1.7 mL, about 1.8 mL, about 1.9 mL, or about 2.0 mL
  • the bispecific antibodies for use in the pharmaceutical formulations comprise a first binding domain that specifically binds the CD3 T cell surface antigen.
  • the first binding domain comprises amino acids 283-525 of SEQ ID NO: 5 of WO 2009/070642, published June 4, 2009 corresponding to US 2010/0303827 (published December 2, 2010), (SEQ ID NO: 1 in the present disclosure), the entire disclosure of which is incorporated by reference herein for all purposes, including for the disclosure of the bispecific antibody sequences recited therein and in the Sequence Listing.
  • the second binding domain of the bispecific antibodies used in the pharmaceutical formulations described herein specifically bind an antigen selected from the group consisting of CD19, CD20, CD22, EphA2, EphA4, INFR, ICOS, Ep-CAM, CEA, and IL-5 receptor.
  • the second binding domain specifically binds CEA, and comprises the amino acid sequence of any one of SEQ ID NOs: 28-44 and 46-51 of US 2013/0035249, published February 7, 2013 (SEQ ID NOs: 2-24 of the present disclosure), the entire disclosure of which is incorporated by reference herein for all purposes, including for the disclosure of the bispecific antibody sequences recited therein and in the Sequence Listing.
  • the second binding domain of the bispecific antibody comprises the amino acid sequence of residues 28-277 of SEQ ID NO:5 from WO2009/070642, corresponding to US 2010/0303827, (SEQ ID NO: 25 herein).
  • the bispecific antibody comprises the amino acid sequence of SEQ ID NO:26, also known as "MEDI-565," a bispecific single chain antibody of the BiTE class that includes an anti-CEA binding portion and an anti-CD3 binding portion.
  • the anti-CEA binding portion is a humanized scFv derived from mouse monoclonal antibody A5B7.
  • MEDI-565 is described and disclosed in WO07/071426, Lutterbuese et al., 2009, Journal of Immunother 32: 341-352, and Osada et al. 2010, British Journal of Cancer, 102: 124-33; each of which is incorporated by reference herein for all purposes.
  • the bispecific antibody comprises the amino acid sequence of residues 28-525 of SEQ ID NO 5 from WO2009/070642, corresponding to US 2010/0303827, (SEQ ID NO: 27 herein), the entire disclosure of which is incorporated by reference herein for all purposes, including for the disclosure of the bispecific antibody sequences recited therein and in the Sequence Listing.
  • the formulations of the invention comprise a BiTE® molecule.
  • a formulation of the invention comprises a BiTE® wherein the BiTE® molecule comprises an anti-CEA VL domain having the amino acid sequence of SEQ ID NO:21, an anti-CEA VH domain having the amino acid sequence of SEQ ID NO:22.
  • a formulation of the invention comprises a BiTE® molecule wherein the BiTE® molecule comprises an anti-CD19 VL domain having the amino acid sequence of SEQ ID NO:28, an anti-CD19 VH domain having the amino acid sequence of SEQ ID NO:29.
  • a formulation of the invention comprises a BiTE® molecule wherein the BiTE® molecule comprises an anti- CD3 VH domain having the amino acid sequence of SEQ ID NO:30 and an anti-CD3 VL domain having the amino acid sequence of SEQ ID NO:31.
  • the invention encompasses stable formulations comprising a single BiTE® molecule, for example, a BiTE® molecule comprising at least a first and a second binding site specific for the CD3 and CEA antigens, respectively; or at least a first and a second binding site specific for the CD3 and CD 19 antigens, respectively.
  • the invention also encompasses formulations comprising two or more bispecific antibodies, for example, bispecific antibodies comprising at least a first and a second binding site specific for the CD3 and CEA antigens, respectively; or at least a first and a second binding site specific for the CD3 and CD19 antigens, respectively.
  • a formulation of the invention comprises MEDI-565 or MT103.
  • a formulation of the invention comprises two or more bispecific antibodies, wherein one of the bispecific antibodies is MEDI-565. In another embodiment, a formulation of the invention comprises two or more bispecific antibodies, wherein one of the bispecific antibodies is MT103.
  • kits for treating a subject by administering a therapeutically effective amount of the pharmaceutical formulations to the subject.
  • the pharmaceutical formulations are administered to a subject parenterally, i.e. no involving the gastrointestinal tract, and are suitably administered subcutaneously (i.e, directly below the dermis and epidermis of the skin) or by injection.
  • Subcutaneous administration can include various pumps or other methods of administration below the skin surface.
  • the term "subject” includes any human or nonhuman animal.
  • nonhuman animal includes all vertebrates, for example, but not limited to, mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
  • treat By the terms “treat,” “treating” or “treatment of (or grammatically equivalent terms) it is meant that the severity of the subject's condition is reduced or at least partially improved or ameliorated and/or that some alleviation, mitigation or decrease in at least one clinical symptom is achieved and/or there is an inhibition or delay in the progression of the condition and/or prevention or delay of the onset of a disease or illness.
  • treat By the terms “treat,” “treating” or “treatment of refer to both prophylactic and therapeutic treatment regimes.
  • a “therapeutically effective” amount as used herein is an amount that provides some improvement or benefit to the subject.
  • a "therapeutically effective" amount is an amount that provides some alleviation, mitigation, and/or decrease in at least one clinical symptom.
  • Clinical symptoms associated with the disorders that can be treated by the methods of the invention are well- known to those skilled in the art. Further, those skilled in the art will appreciate that the therapeutic effects need not be complete or curative, as long as some benefit is provided to the subject. Appropriate dosages and timing of such dosages for administration of the pharmaceutical formulations can be readily determined by those of ordinary skill in the art, and are described for example in U.S. 2013/0035249 ⁇ see e.g., paragraphs [0236- 0245]), which is incorporated by reference herein in its entirety.
  • methods for selecting a buffer or an excipient for use in a protein formulation are provided. Such methods can also be utilized for selecting a buffer or excipient suitable to reduce or prevent protein aggregation in a protein formulation.
  • polypeptide refers to a polymer of amino acid residues.
  • the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.
  • a protein formulation refers to a composition comprising one or more proteins (may be the same or different) and one or more appropriate buffers and/or excipients.
  • polypeptide refers including antibodies or antigen-binding fragments thereof.
  • the methods comprise providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient.
  • concentration of the protein is the same in each buffer solution, and the concentration of the buffer or the excipient in each buffer solution is the same, but the buffer or the excipient in each buffer solution is different, so as to provide a method for investigation of the effect of the type of buffer or excipient on the protein formulation.
  • the methods comprise measuring a light scattering intensity of each of the plurality of buffer solutions.
  • Light scattering intensity can be measured using various methods and instrumentations as described herein or otherwise known in the art.
  • 'K' is an optical constant
  • R90 is Rayleigh ratio
  • M (kDa) is the weight average molecular weight
  • C (mg/mL) is protein concentration
  • B 22 (mol » mL/g 2 ) is the second virial coefficient
  • the more positive the second virial coefficient in comparison to another buffer or excipient the more desirable the buffer or excipient is for use in a protein formulation.
  • selection of buffers and/or excipients with more positive second virial coefficients is an indication of the suitability of the buffer or excipient for use in protein formulations, by providing a more stable protein formulation with reduced aggregation and/or fragmentation of protein.
  • methods are provided for selecting a concentration of a buffer or an excipient for use in a protein formulation. Methods are also provided for selecting a buffer concentration or excipient concentration suitable to reduce or prevent protein aggregation in a protein formulation.
  • the methods suitably comprise providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient.
  • concentration of the protein is the same in each buffer solution, and the concentration of the buffer or the excipient in each buffer solution is different, but the buffer or the excipient in each buffer solution is the same, so as to provide a method for investigation of the effect of the buffer concentration or excipient concentration on the protein formulation.
  • the light scattering intensity of each of the plurality of buffer solutions is measured, and a Debye plot is constructed.
  • the second virial coefficients of each of the plurality of buffer solutions are compared.
  • a more positive second virial coefficient indicates the concentration of the buffer or the excipient for use in the protein formulation.
  • selection of a concentration of buffers and/or excipients with more positive second virial coefficients is an indication of the suitability of the concentration of buffer or excipeint for use in protein formulations, by providing a more stable protein formulation with reduced aggregation and/or fragmentation of protein.
  • the methods provided herein are suitably used with any protein formulation, including formulations where the protein is an antibody or antigen-binding fragment thereof, a peptide or a fusion protein.
  • the antibody investigated in the methods described herein is a bispecific antibody comprising a first and a second binding domain, suitably where the first binding domain specifically binds the CD3 T cell surface antigen.
  • the buffer includes, but is not limited to, citrate, succinate, phosphate, histidine, acetate, TRIS and combinations thereof.
  • the concentration of the buffer or the excipient is from about 5 mM to about 200 mM, suitably less than about 100 mM.
  • excipients include, but are not limited to, proteins (for example, but not limited to, serum albumin), amino acids (for example, but not limited to, aspartic acid, glutamic acid, lysine, arginine, glycine), surfactants (for example, but not limited to, SDS, Tween 20, Tween 80, polysorbate, polysorbate 80 and nonionic surfactants), sugars and saccharides (for example, but not limited to, glucose, sucrose, maltose and trehalose), polyols (for example, but not limited to, mannitol and sorbitol), fatty acids and phospholipids (for example, but not limited to, alkyl sulfonates and caprylate).
  • proteins for example, but not limited to, serum albumin
  • amino acids for example, but not limited to, aspartic acid, glutamic acid, lysine, arginine, glycine
  • surfactants for example, but not limited to, SDS,
  • the methods for selection of a buffer or excipient or concentration of a buffer or excipient are carried out with bispecific antibody formulations, including formulations which include the various bispecific antibodies and binding domains described throughout.
  • MEDI-565/BiTE® comprising SEQ ID NO: 26 was prepared according to standard methods. Other bispecific antibodies are readily prepared by those in the field using well-known methods.
  • Buffer components including sodium citrate, citric acid, sodium phosphate, sodium succinate, L-histidine and L-histidine hydrochloride were purchased from
  • Lyoprotectant, ⁇ , ⁇ -trehalose dihydrate and surfactant, polysorbate 80 were purchased from Ferro Pfanstiehl (Waukegan, IL) and J.T.Baker (Center Valley, PA) respectively.
  • Second virial coefficient (B 22 ) measurements were performed using a DAWN ® HELEOS ® Multi-Angle Light Scattering (MALS) detector coupled to OPTILAB ® REXTM refractive index detector (Wyatt Technologies, Santa Barbara, CA) to obtain virial coefficients (B 22 ) and apparent molecular weight.
  • MALS Multi-Angle Light Scattering
  • OPTILAB REXTM refractive index detector
  • Protein and buffer solutions were filtered through 0.22 ⁇ filters and delivered into the light scattering cell and refractive index detector using a CALYPSO ® dispensing system, (Wyatt Technologies, Santa Barbara, CA). Relative scattering intensities in excess of the background (solvent) are converted into absolute scattering intensities and plotted as a function of concentration to obtain B 22 and weight average molecular weight from the Debye working equation:
  • 'K' is an optical constant
  • R90 is Rayleigh ratio
  • M (kDa) is the weight average molecular weight
  • C (mg/mL) is protein concentration
  • B 22 (mol- mL/g 2 ) is the second virial coefficient.
  • Bispecific antibody liquid formulations were subjected to 5°C, 25°C/60 relative humidity (RH) and 30°C.
  • the aggregation of bispecific antibody increased at elevated temperature, thus 25°C/60 RH and 30°C were selected as accelerated temperature conditions to evaluate the impact of excipients and excipient concentrations on antibody aggregation.
  • samples were incubated at -80°C and/or -40°C.
  • compositions were investigated comprising different buffers at varying concentrations (generally 10 mM to 100 mM), with different lyoprotectants at a concentration range of about 50 mM to 150 mM, and polysorbate 80 (0.002 to 0.05 )
  • Lyophilized bispecific antibody formulations were subjected to 5°C, 25°C/60 RH and 40°C/75 RH to evaluate stability.
  • Freeze drying was performed using a laboratory scale VirTis freeze dryer.
  • HP-SEC samples were analyzed using Agilent (Santa Clara, CA) 1100 HP-SEC, equipped with a YMC ® DIOL (5 ⁇ , 8x300mm; 200A) size exclusion column with an FLD detector (FLD method) or Agilent 1200 series HPLC TSK-Gel G3000SWXL (5 ⁇ , 7.8x300mm; 250A) size exclusion column with a UV-vis detector (DAD method).
  • Samples analyzed using DAD method were run undiluted to achieve a target column load of 170-250 ⁇ g whereas samples analyzed using the FLD method were diluted (with phosphate buffered saline) to achieve a target load of 0.5 ⁇ g.
  • samples were incubated in the auto-sampler (5°C) for approximately 1 hour prior to analysis.
  • the chilled samples were injected onto respective columns and eluted isocratically with 0.1 M disodium phosphate containing 0.1 M sodium sulfate and 0.05% sodium azide, pH 6.8, at a flow rate of 1.0 ml/minute (DAD method) or 0.2 M disodium phosphate containing 0.2 M sodium sulfate, pH 7, at a flow rate of 0.75 ml/minute (FLD method).
  • the eluted protein was detected using UV absorbance at 280 nm (DAD method) and fluorescence detection with excitation at 280nm and emission at 345nm (FLD method). The results are reported as the area percent of the product monomer peak. Peaks eluting earlier than the monomer are recorded as percent aggregate and peaks eluting after the monomer are recorded as percent fragment/other.
  • a T-Cell mediated apoptosis bioassay was used to measure the ability of a test sample to induce apoptotic cell death of a CEA-expressing human colon tumor cell line (LS174T).
  • LS174T CEA-expressing human colon tumor cell line
  • MCI 5 effector T-cells
  • the transient ligation of the effector cell and the target cell via the bispecific antibody activates the effector cells to release cytotoxic granules (Granzyme B), which enter the target cells through Perforin- mediated tunnels in the cell membrane.
  • This process induces the LS174T cells to undergo apoptosis via Caspase 3/7 activation that can be measured by the Caspase-GloTM 3/7 Assay System.
  • the amount of luminescence proportional to the amount of apoptosis was quantified in a luminometer after reaction with a Caspase 3/7 luciferase substrate.
  • LS174T and MC15 cells were plated in the cell growth media (10% fetal bovine serum (FBS) in RPMI) in a 96-well plate. A series of sample dilutions were added to the plates. After incubation for 23 hours, the Caspase 3/7 reagent was added to the plate and the luminescence was measured on a plate reader. The data was analyzed in Softmax Pro using a four-parameter logistical curve fit to determine the EC 50 values of the samples. The potency of the test sample was determined by dividing the EC 50 of Reference Standard by the EC 50 of the test sample.
  • FIG. 2 shows the effect of buffer on stability of the bispecific antibody MEDI-565, examining aggregation percent for the antibody at 5 mg/mL, at 30° C for 15 days as measured by HPSEC, using the methods described in Example 1.
  • the study examined four buffers: histidine (circle), succinate (square), citrate (triangle) and phosphate (cross), each at 30 mM. Citrate buffer demonstrated the lowest amount of aggregation over the time-course of the study.
  • PPI protein-protein interactions
  • B 22 The second virial coefficient (B 22 ) is a thermodynamic parameter known to reflect PPI in dilute solution conditions and can be utilized to determine the colloidal stability.
  • citrate, succinate, phosphate and histidine was determined for samples comprising 30 mM buffer, 5 mg/mL of MEDI-565 (SEQ ID NO: 26) at pH 6.0.
  • FIG. 3 further demonstrates that the single chain bispecific antibody in citrate showed a surprisingly low amount of aggregation as compared to the other buffers. Also shown on the second Y axis is the second virial coefficient for the formulations. As described, the most positive, i.e., least negative, value for B22 corresponded to citrate, which also demonstrated the lowest aggregation rate, evidence of the weakly attractive protein-protein interaction in citrate compared to other buffers.
  • EXAMPLE 4 IMPACT OF CITRATE CONCENTRATION ON STABILITY
  • the lowest aggregation rate also corresponded to the formulation having the most positive second virial coefficient. See FIG. 4B.
  • Increasing the citrate ion concentration lowered the aggregation rate demonstrating improved colloidal stability, with higher citrate concentrations showing a net protein-protein interactions change from attractive to repulsive, likely explaining the increased colloidal stability also observed at high ion concentrations.
  • No significant changes to conformation stability by DSF & DSC were observed as a function of citrate concentration.
  • concentrations above approximately 60-100 mM citrate subject discomfort associated at the injection site of a pharmaceutical formulation is often considered a limiting factor for using higher citrate concentrations.
  • these experiments support selecting a target citrate concentration of about 60 mM for a single chain bispecific antibody formulation comprising about 5.0 mg/mL of the bispecific antibody to maximize antibody stability while minimizing potential injection site discomfort.
  • Formulations 1.7 mg/mL MEDI-565, 30 mM citrate, 70 mM trehalose, 0.01 % polysorbate-80, pH 6.0; 2.5 mg/mL MEDI-565, 30 mM citrate, 80 mM trehalose, 0.01 % polysorbate-80, pH 6.0; and
  • Example 6 it was determined that in order to provide a formulation capable of being delivered to a patient at 5 mg/mL which is stable when held at 25 °C in preparation for and during administration, a lower concentration of protein was necessary during preparation of the liquid pharmaceutical formulations prior to lyophilization, and subsequent reconstitution in a volume that provides the final 5 mg/mL of protein for subject administration.
  • DS osmolality refers to the osmolality of the drug substance (DS), or liquid formulation, prior to lyophilization.
  • DP osmolality refers to the osmolality of the lyophilized drug product (DP) following reconstitution in a volume of 1.4 mL (WFI).
  • Liquid formulations 1-3 were examined for stability over the course of 12 days at 25° C by measuring % aggregation by HSPEC.
  • formulation 2 was selected as the lead formulation.
  • FIG. 7 A shows the stability (% Aggregate at 5° C as measured by HPSEC) of 3 bispecific antibody (MEDI-565) lyophilized formulations at 5° C.
  • the study examined the three formulations described in Table 2. As demonstrated, all 3 formulations were stable when stored at 5° C.
  • FIG. 7B shows the stability (% Aggregate at 40° C as measured by HPSEC) of 3 bispecific antibody (MEDI-565) lyophilized formulations at an accelerated temperature.
  • the study examined the three formulations described in Table 2. As demonstrated, no significant increase in aggregation was observed for all 3 formulations at 40° C.
  • All three formulations (1, 2 and 3 described in Table 2) were lyophilization suitable with the trehalose content optimized to improve cake appearance (about 60- 80mM) as described in Example 1. All three formulations had the following characteristics: (1) Reconstitution time ⁇ 5min; (2) Moisture content ⁇ 1.3%, and (3) an acceptable cake quality and acceptable shrinkage observed following a visual inspection.
  • all three formulations were stable at 5°C, 25 °C and 40°C with no significant increase in aggregation observed in accelerated stability studies performed at 40°C.
  • EXAMPLE 7 EFFECT OF SUCCINATE BUFFER ON STABILITY &
  • FIG. 10 demonstrates percent aggregation of liquid MEDI-565 pharmaceutical formulations comprising the amounts of bispecific antibody, citrate and trehalose, following storage at -80° C for 3 months for formulations 1 and 3 and 27 months for formulation 2 (and then thawing and measurement). In all cases, aggregation % did not change significantly and was maintained below 0.5%, and virtually no sub-particles larger than 2 ⁇ were observed. Stability was demonstrated for periods of up to 2 years after storage at -80° C. In addition, virtually no particles larger than 2 ⁇ for the pharmaceutical formulations after three cycles of freeze-thaw (-80° C and room temperature) were observed.
  • FIG. 11 shows the results of an investigation of pH on aggregation for a MEDI- 565 formulation (1.7 mg/mL, 20 mM citrate, 70 mM trehalose, 0.01% polysorbate-80). No significant effects on aggregation were noted, which suggests that MEDI-565 can be formulation at a pH range of 6 + 1 until while maintaining stability.
  • the Examples demonstrate that a 1.7mg/ml single chain bispecific antibody, 20mM Sodium Citrate/Citric Acid, 70mM Trehalose Dihydrate, 0.01% PS80, pH 6.0 pharmaceutical formulation and a 5mg/ml single chain bispecific antibody, 59mM Sodium Citrate/Citric Acid, 0.21M Trehalose Dihydrate, 0.03% PS80, pH 6.0 (having an osmolality of 436mOsm/kg) pharmaceutical formulation are each suitable for parenteral administration of the single chain bispecific antibody, including subcutaneous administration or injection. These formulations provided good stability at 5° C, 25° C and -80° C with combined aggregation ⁇ 2.0%.
  • EXAMPLE 10 INVITRO ACTIVITY (BIOASSAY) OF DRUG SUBSTANCE AND
  • FIG. 12 shows the results of in vitro activity assay (bioassay) of frozen drug substance (circle), lyophilized drug product stored at 2-8°C (square) and lyophilized drug product stored at 40°C (triangle) relative to a reference standard.
  • the drug substance shown in FIG. 12 was composed of 1.7 mg/mL MEDI-565, 20 mM citrate, 70 mM trehalose, 0.01% polysorbate-80, pH 6 and the corresponding post reconstitution drug product contained 5mg/mL MEDI-565, 59 mM citrate, 0.21 M trehalose, 0.03% polysorbate 80, pH 6.0.

Abstract

The present invention provides pharmaceutical formulations of bispecific antibodies, including bispecific antibodies comprising a first binding domain that suitably binds the CD3 T cell surface antigen and a second binding domain, which suitably binds carcinoembryonic antigen (CEA). The formulations are suitably designed for subcutaneous administration or injection. The present invention also provides methods of determining suitable buffers and excipients for use in protein formulations, particularly for antibody formulations.

Description

FORMULATIONS OF BISPECIFIC ANTIBODIES
FIELD OF THE INVENTION
[0001] The present invention provides pharmaceutical formulations of bispecific antibodies or fragments thereof that minimize aggregation. In some aspects, the pharmaceutical formulations disclosed herein include bispecific antibodies comprising a first binding domain that binds the CD3 T cell surface antigen. In some aspects, the pharmaceutical formulations disclosed herein comprise BiTE® molecules, including BiTE® molecules having at least a first binding site specific for CD3 and a second binding site specific for carcinoembryonic antigen (CEA) antigens (e.g. MEDI-565) or CD 19 antigens (e.g. MT103). In further embodiments, BiTE® molecules formulated according to the methods disclosed herein are designed for parenteral administration including subcutaneous administration or injection. The present invention also provides methods of determining suitable buffers and excipients for use in protein formulations, particularly for antibody formulations including bispecific antibodies.
BACKGROUND
[0002] Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Bispecific antibodies may bind to two different epitopes of a single cell surface marker or they may bind a first cell surface marker and further bind a second cell surface marker on the same cell. A binding arm specific for a surface antigen of a target cell may also be combined with an arm which binds to a triggering molecule on a leukocyte such as a T cell receptor molecule (e.g., CD2 or CD3), or Fc receptors for IgG (FcyR), so as to focus cellular defense mechanisms to the target cell. Bispecific antibodies can be prepared as full-length antibodies or antibody fragments (e.g., F(ab ')).
[0003] Methods for making bispecific antibodies are known in the art. (See, for example,
Millstein et al., Nature, 305:537-539 (1983); Traunecker et al., EMBO J., 10:3655- 3659 (1991); Suresh et al., Methods in Enzymology, 121:210 (1986); Kostelny et al., J Immunol., 148(5):1547-1553 (1992); Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993); Gruber et al., J Immunol., 152:5368 (1994); U.S. Patent Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; 5,601,81; 95,731,168; 4,676,980; and 4,676,980, WO94/04690; WO 91/00360; WO 92/200373; W093/17715; WO92/08802; and EP 03089. Each herein incorporated by reference in its entirety).
[0004] MEDI-565 (also known as CEA-BiTE; AMG 211 ; MT111 (SEQ ID NO:26)) is a
CEA/CD3 bispecific single-chain antibody or BiTE® molecule reported to prevent subcutaneous tumor growth and formation of metastases in preclinical models. See, e.g., WO 07/071426, Lutterbuese et al., 2009, Journal of Immunother 32: 341-352, and Osada et al. 2010, British Journal of Cancer, 102: 124-33. MEDI-565 is in phase I clinical trials (clinicaltrials.gov identifier: NCT01284231) for the treatment of gastrointestinal adenocarcinomas. MT103 (also known as Blinatumomab; (SEQ ID NO: 25) is a recombinant, bispecific single-chain antibody or BiTE® molecule, which binds to CD 19 on target cells and the CD3 complex on T effector cells. See Hoffmann Pet. al., Int J Cancer. 2005 May 20;115(1):98-104. It is a single polypeptide chain of 55 kDa with the variable binding domains of two murine monoclonal antibodies. By simultaneously binding to target and effector cells, MT103 triggers the T effector cell to form an immunological synapse, and to release perforin and granzymes which induce cell death of target cells. Target cells comprise human B cells of all development stages (with the exception of plasma cells) and malignant cells of most B cell-derived lymphoma and leukemia. MT103 is currently in development for treatment of B-cell cancers (clinicaltrials.gov identifier: NCT02013167).
BRIEF SUMMARY OF THE INVENTION
[0005] Exemplary pharmaceutical formulations for single chain bispecific antibodies including MEDI-565 and MT103 are disclosed in U.S. Patent Publication No. 2010/0303827, herein incorporated by reference in its entirety. These pharmaceutical formulations comprise a bispecific antibody stabilized with lysine. One major concern during the development of protein formulations is stability. Insufficient protein stability can result in aggregation, which is undesirable, as it can impact protein activity and immunogenicity. This is a particular problem when formulating single chain antibodies at ultra-low concentrations (e.g. between approximately 25 μg/ml and 500 μg/ml) since at these protein concentrations even a low level of aggregation or brief contact with vial surfaces (i.e. absorption loss) can lead to significant changes in antibody activity. The antibody formulations disclosed in U.S. Patent Publication No. 2010/0303827 prevent absorption loss to contact surfaces and prevent aggregation when formulated at about 25 μg/ml to about 500 μg/ml antibody concentration. However, these formulations are generally not suitable at increased protein concentrations (about 1.0 mg/ml to about 5.0 mg/ml) as aggregation of single chain bispecific antibodies (e.g. MEDI-565 and MT103) increases significantly at these higher antibody concentrations. Given the nature of single chain bispecific antibodies (e.g. MEDI-565 and MT103), small increases in protein aggregation often result in super-potent antibodies unsuitable for clinical use. Accordingly, the formulations disclosed herein provide stability by reducing antibody aggregation of single chain bispecific antibodies (e.g. MEDI-565 and MT103) at increased antibody concentrations. In addition, other benefits of the formulations disclosed herein include reduced volume dosing, alternative dosing routes (e.g. IV or subcutaneous delivery) and suitably exclude lysine which can increase the osmolarity of single chain antibody formulations potentially leading to increase pain upon parenteral administration (e.g. subcutaneous or injection).
[0006] Therefore, further development was initiated to generate a formulation comprising about 1.0 mg/mL to about 5.0 mg/mL of a single chain bispecific antibody that 1) excludes lysine, 2) minimizes aggregate formation during storage (e.g. total aggregate < 2.0%), 3) maximizes stability of the drug substance and drug product, and 4) is suitable for parenteral administration including subcutaneous administration or injection. As described herein and in the Examples, it was surprisingly found that the disclosed pharmaceutical formulations allow for use of high amounts of bispecific antibodies (e.g. about 1.0 mg/mL to about 5.0 mg/mL) while exhibiting increased stability as to provide liquid pharmaceutical formulations and lyophilized formulations for extended storage and shipping, prior to administration to a patient. In addition, the lyophilized pharmaceutical formulations can be reconstituted in a reduced volume, i.e., about 1/2 to about 1/3 of the starting volume, so as to increase the final amount of bispecific antibody, yet still maintain the desired stability (i.e., low aggregation and/or fragmentation) and also still maintain a time of reconstitution on the order of minutes (e.g., 5-15 minutes), so as to allow for simple use in clinical settings and to increase patient and physician compliance.
[0007] Described throughout are embodiments that meet the needs described above.
[0008] In one embodiment pharmaceutical formulations are provided comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 1.0 mg/mL to about 5.0 mg/mL of the bispecific antibody, about 15 mM to about 60 mM citrate, about 25 mM to about 0.21 M of a lyoprotectant, and about 0.002% to about 0.1% of a surfactant, wherein the formulation has a pH of about 5.0 to about 7.0.
[0009] Suitably, the formulations comprise about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody, or about 1.7 mg/mL or about 2.5 mg/mL of the bispecific antibody.
[0010] The formulations can comprise about 20 mM to about 30 mM citrate, or about 20 mM citrate or about 30 mM citrate. In embodiments, the formulations comprise about 60 mM to about 80 mM of the lyoprotectant, or about 70 mM of the lyoprotectant. Suitably, the formulations comprise about 0.008% to about 0.012% of the surfactant, suitably about 0.01% of the surfactant.
[0011] Exemplary lyoprotectants include, but are not limited to, trehalose, lactose, mannitol, raffinose and sucrose. Exemplary surfactants include, but are not limited to, polysorbate-20, polysorbate-40, polysorbate-60 and polysorbate-80.
[0012] Also provided are pharmaceutical formulations comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody, about 20 mM to about 30 mM citrate, about 60 mM to about 80 mM trehalose, and about 0.002% to about 0.1% polysorbate 80, wherein the formulation has a pH of about 5.0 to about 7.0.
[0013] In further embodiments, pharmaceutical formulations are provided comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 1.7 mg/mL of the bispecific antibody, about 20 mM citrate, about 70 mM trehalose, and about 0.01% polysorbate 80, wherein the formulation has a pH of about 6.0.
[0014] Additional pharmaceutical formulations comprise a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 5.0 mg/mL of the bispecific antibody, about 60 mM citrate, about 0.21 M trehalose, and about 0.03% polysorbate 80, wherein the formulation has a pH of about 6.0.
[0015] Suitably, the pharmaceutical formulations have a volume of about 0.5 mL to about
12.0 mL, for example about 1.4 mL.
[0016] Suitably, the pharmaceutical formulations are administered to a subject parenterally, for example subcutaneously or by injection.
[0017] Suitably, the formulations are a liquid formulation, a frozen formulation, a lyophilized formulation or a reconstituted formulation.
[0018] Also provided are lyophilized pharmaceutical formulations comprising a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises about 2.5 mg to about 61 mg of the bispecific antibody, about 8.5 mg to about 204 mg of citrate, about 40 mg to about 954mg trehalose and about 0.15 μg to about 3^g of polysorbate 80, more suitably about 7 mg of the bispecific antibody, about 25 mg or about 24.95 mg citrate, about 117 mg or about 116.5 mg trehalose, and about 0.4 μg polysorbate 80.
[0019] Suitably, the lyophilized formulations following storage at 25° C for 6 months, or storage at 2-8° C for 24 months, exhibit < 2.0% aggregation when reconstituted.
[0020] Also provided are methods of preparing a pharmaceutical formulation comprising preparing a liquid pharmaceutical formulation as described herein, lyophilizing the liquid pharmaceutical formulation of to produce a lyophilized formulation, and reconstituting the lyophilized formulation in a volume suitable for parenteral administration. In embodiments, the methods further comprise freezing the liquid pharmaceutical formulation (e.g., to at least about -40° C or at least about -80° C) prior to the lyophilizing.
[0021] Suitably, the lyophilized formulations are reconstituted in about 1/2 to about 1/3 volume of the liquid pharmaceutical formulation, suitably in a volume of about 0.5 mL to about 12.0 mL, e.g., in a volume of about 1.4 mL, or a volume suitable for subcutaneous administration or injection.
[0022] In some embodiments, the pharmaceutical formulations disclosed herein contain a bispecific antibody wherein the first binding domain comprises SEQ ID NO: 1, SEQ ID NO: 30 or SEQ ID NO: 31. In additional embodiments, the second binding domain specifically binds an antigen selected from the group consisting of: CD19, CD20, CD22, EphA2, EphA4, INFR, ICOS, Ep-CAM, CEA, and IL-5 receptor. Suitably, the second binding domain comprises the amino acid sequence of any one of SEQ ID NOs: 2-24 and 28-29, the bispecific antibody comprises SEQ ID NO: 26, the second binding domain specifically binds CD19, the second binding domain comprises SEQ ID NO: 25 and/or the bispecific antibody comprises SEQ ID NO: 27.
[0023] Also provided are methods for selecting a buffer or an excipient for use in a protein formulation or for selecting a buffer or excipient suitable to reduce or prevent protein aggregation in a protein formulation, the method comprising providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient, wherein the concentration of the protein is the same in each buffer solution, and wherein the concentration of the buffer or the excipient in each buffer solution is the same, but the buffer or the excipient in each buffer solution is different, measuring a light scattering intensity of each of the plurality of buffer solutions, constructing a Debye plot, determining a second virial coefficient of each of the plurality of buffer solutions, and comparing the second virial coefficient of each of the plurality of buffer solutions, wherein a more positive second virial coefficient indicates a buffer or excipient for use in the protein formulation.
[0024] In additional embodiments, methods are provided for selecting a concentration of a buffer or an excipient for use in a protein formulation, or for selecting a buffer or excipient concentration suitable to reduce or prevent protein aggregation in a protein formulation, the method comprising providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient, wherein the concentration of the protein is the same in each buffer solution, and wherein the concentration of the buffer or the excipient in each buffer solution is different, but the buffer or the excipient in each buffer solution is the same, measuring a light scattering intensity of each of the plurality of buffer solutions, constructing a Debye plot, determining a second virial coefficient of each of the plurality of buffer solutions, comparing the second virial coefficient of each of the plurality of buffer solutions, wherein a more positive second virial coefficient indicates the concentration of the buffer or the excipient for use in the protein formulation. [0025] Suitably, the protein is an antibody or antigen-binding fragment thereof, a peptide or a fusion protein, including a bispecific antibody comprising a first and a second binding domain, and wherein the first binding domain specifically binds the CD3 T cell surface antigen, including the sequences recited herein.
[0026] In embodiments of the methods, the buffer is selected from the group consisting of citrate, succinate, phosphate, histidine, acetate, TRIS and combinations thereof, the concentration of the buffer or the excipient is from about 5 mM to about 200 mM, suitably less than about 100 mM. In additional embodiments, the excipient is a sugar.
[0027] Further embodiments, features, and advantages of the embodiments, as well as the structure and operation of the various embodiments, are described in detail below with reference to accompanying drawings.
BRIEF DESCRIPTIONS OF THE DRAWINGS
[0028] FIG 1A. shows a schematic representation of a bispecific antibody comprising a
CD3 binding domain and a carcinoembryonic antigen binding domain.
[0029] FIG. IB shows a schematic representation of a bispecific antibody comprising a
CD3 binding domain and a carcinoembryonic antigen binding domain targeting and killing tumor cells.
[0030] FIG. 1C shows the aggregation propensity of a bispecific antibody (MEDI-565; circles) compared to a monoclonal, whole IgGl antibody (squares) incubated at 25 °C as measured by High Pressure Size Exclusion Chromatography (HPSEC). As demonstrated, the bispecific antibody (MEDI-565) aggregates at very high rate compared to the monoclonal antibody.
[0031] FIG. 2 shows the effect of different buffers on the stability ( Aggregate) of a bispecific antibody (MEDI-565) at ~5mg/mL incubated at 30°C for 15 days as measured by High Pressure Size Exclusion Chromatography (HPSEC). The study examined four buffers: histidine (circles), succinate (squares), citrate (triangles) and phosphate (cross), each at 30 mM. Citrate buffer demonstrated the lowest amount of aggregation.
[0032] FIG. 3 shows the effect of different buffers on the stability (Aggregation rate/day at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) compared to the second virial coefficient (B22) for each of the compositions. The left Y axis represents aggregation rate per day at 30°C and the right Y axis represents second virial coefficient (B22) in lOe"4 mol.ml per g2. The bars from left to right represent citrate, phosphate, succinate and histidine, respectively. Citrate buffer had the lowest aggregation rate and a less negative B22 compared to other buffers. mol=Moles; ml=milliliter; g=gram.
[0033] FIG. 4A shows the impact of citrate concentration on the stability ( Aggregate at
30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC) of a bispecific antibody (MEDI-565) at ~5 mg/mL. The study examined five citrate concentrations: lOmM (circles), 20mM (squares), 30mM (triangle), 50mM (inverted triangles) and lOOmM (diamonds). As demonstrated, an increase in citrate concentration resulted in decrease in aggregation of the bispecific antibody.
[0034] FIG. 4B shows the impact of citrate concentration on the stability (Aggregation rate at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) at ~5 mg/mL compared to the second virial coefficient for the compositions. The left Y axis represents aggregation rate per day at 30°C and the right Y axis represents second virial coefficient (B22) in lOe"4 mol.ml per g2. The bars from left to right represent increasing citrate concentration (lOmM, 20mM, 30mM, 50mM and lOOmM, respectively). As demonstrated, the lowest aggregation rate (lOOmM citrate) corresponded to the formulation having the most positive B22. mol=Moles; ml=milliliter; g=gram.
[0035] FIG. 5 shows the impact of protein concentration on the stability ( Aggregate at
25 °C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) over 12 days. The study examined three formulations: 1.7mg/mL MEDI-565, 30 mM citrate, 70 mM trehalose, 0.01 % polysorbate-80, pH 6.0 (1.7mg/ml; circles), 2.5 mg/mL MEDI-565, 30 mM citrate, 80 mM trehalose, 0.01 % polysorbate-80, pH 6.0 (2.5mg/mL; squares) and 5.0 mg/mL MEDI-565, 60 mM citrate, 150 mM trehalose, 0.02 % polysorbate-80, pH 6.0 (5mg/mL; triangles). As demonstrated, increased protein concentration resulted in increased antibody aggregation and lower stability, with the 1.7mg/mL and 2.5 mg/mL formulations showing significant stability.
[0036] FIG. 6 shows the stability ( Aggregate at 25 °C as measured by High Pressure
Size Exclusion Chromatography (HPSEC)) of 3 bispecific antibody (MEDI-565) formulations. The study examined three formulations: formulation 1: 2.5mg/ml MEDI- 565, 30mM Citrate, 80mM Trehalose, 0.01% PS-80, pH 6.0 (circles), formulation 2: 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80, pH 6.0 (squares) and formulation 3: 1.7mg/ml MEDI-565, 30mM Citrate, 60mM Trehalose 0.01% PS-80, pH 6.0 (triangles). As demonstrated, formulation 2 and formulation 3 provided improved stability over formulation 1 over 12 days.
[0037] FIG. 7 A shows the stability (% Aggregate at 5°C as measured by High Pressure
Size Exclusion Chromatography (HPSEC)) of 3 bispecific antibody (MEDI-565) lyophilized formulations. The study examined three formulations: formulation 1 : 2.5mg/ml MEDI-565, 30mM Citrate, 80mM Trehalose, 0.01% PS-80, pH 6.0 (circles), formulation 2: 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80, pH 6.0 (squares) and formulation 3: 1.7mg/ml MEDI-565, 30mM Citrate, 60mM Trehalose 0.01% PS-80, pH 6.0 (triangles). As demonstrated, all three formulations are stable when stored at 5° C.
[0038] FIG. 7B shows the stability (% Aggregate at 40°C as measured by High Pressure
Size Exclusion Chromatography (HPSEC)) of 3 bispecific antibody (MEDI-565) lyophilized formulations. The study examined three formulations: formulation 1 : 2.5mg/ml MEDI-565, 30mM Citrate, 80mM Trehalose, 0.01% PS-80, pH 6.0 (circles), formulation 2: 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80, pH 6.0 (squares) and formulation 3: 1.7mg/ml MEDI-565, 30mM Citrate, 60mM Trehalose 0.01% PS-80, pH 6.0 (triangles). As demonstrated, no significant increase in aggregation was observed for all three formulations at 40° C.
[0039] FIG. 8A shows the impact of succinate concentration on the stability (%
Aggregate at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) at ~5 mg/mL. The study examined four succinate concentrations: lOmM (circles), 30mM (squares), 50mM (triangles) and lOOmM (closed triangles). As demonstrated, increased succinate concentration resulted in a decrease in aggregation of the bispecific antibody.
[0040] FIG. 8B shows the impact of succinate concentration on the stability (Aggregation rate/day at 30°C as measured by High Pressure Size Exclusion Chromatography (HPSEC)) of a bispecific antibody (MEDI-565) at ~5 mg/mL compared to the second virial coefficient for the compositions. The left Y axis represents aggregation rate per day at 30°C and the right Y axis represents second virial coefficient (B22) in lOe"4 mol.ml per g2. The bars from left to right bar represent increasing succinate concentration (lOmM, 30mM, 30mM and lOOmM, respectively). As demonstrated, the lowest aggregation rate (100 mM) corresponded to the formulation having the most positive B22.
[0041] FIG. 9A shows stability (% Aggregate as measured by High Pressure Size
Exclusion Chromatography (HPSEC)) of 3 bispecific antibody (MEDI-565) formulations after reconstitution and storage at room temperature for 0 or 4 hours. The study examined three formulations: from left to right formulation 1: 2.5mg/ml MEDI-565, 30mM Citrate, 80mM Trehalose, 0.01% PS-80, pH 6.0; formulation 2: 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80, pH 6.0; and formulation 3: 1.7mg/ml MEDI-565, 30mM Citrate, 60mM Trehalose 0.01% PS-80, pH 6.0. As demonstrated, all 3 formulations showed comparable stability right after reconstitution (T-0) and 4 hours after reconstitution evidencing that each of the three formulations provides 4 hours of post reconstitution in use stability at room temperature.
[0042] FIG. 9B shows stability (% Aggregate as measured by High Pressure Size
Exclusion Chromatography (HPSEC)) of 3 bispecific antibody (MEDI-565) formulations after reconstitution and storage for 0 or 24 hours at 5°C. The study examined three formulations: from left to right formulation 1: 2.5mg/ml MEDI-565, 30mM Citrate, 80mM Trehalose, 0.01% PS-80, pH 6.0; formulation 2: 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80, pH 6.0; and formulation 3: 1.7mg/ml MEDI- 565, 30mM Citrate, 60mM Trehalose 0.01% PS-80, pH 6.0. As demonstrated, all 3 formulations showed comparable stability right after reconstitution (T-0) and 24 hours after reconstitution evidencing that each of the three formulations provides 24 hours of post reconstitution in use stability at 5°C.
[0043] FIG. 10 shows the stability (% Aggregate at -80°C as measured by High Pressure
Size Exclusion Chromatography (HPSEC)) of 3 bispecific antibody (MEDI-565) formulations at -80° C. The study examined three formulations: formulation 1: 2.5mg/ml MEDI-565, 30mM Citrate, 80mM Trehalose, 0.01% PS-80, pH 6.0 (circles), formulation 2: 1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80, pH 6.0 (squares) and formulation 3: 1.7mg/ml MEDI-565, 30mM Citrate, 60mM Trehalose 0.01% PS-80, pH 6.0 (triangles). As demonstrated, all 3 formulations are stable when stored frozen.
[0044] FIG. 11 shows stability (% Aggregate at 25°C as measured by High Pressure Size
Exclusion Chromatography (HPSEC)) of a bispecific antibody formulation (1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS-80) over the pH range 5-7. The study examined five pH conditions: pH 5 (circles), pH 5.5 (squares), pH 6 (triangles), pH 6.5 (inverted triangles) and pH 7 (diamonds). As demonstrated, pH has no significant impact on aggregation.
[0045] FIG. 12 shows % relative activity relative to a reference standard of a bispecific antibody formulation (1.7mg/ml MEDI-565, 20mM Citrate, 70mM Trehalose, 0.01% PS- 80, pH 6.0 (drug substance) or 5mg/mL MEDI-565, 59 mM citrate, 0.21 M trehalose, 0.03% polysorbate 80, pH 6.0 (post-reconstituted drug product)) after storage for up to 27 months. No significant changes to the in vitro activity of frozen drug substance (circle), lyophilized drug product stored at 2-8°C (square) and lyophilized drug product stored at 40°C (triangle) relative to a reference standard was observed.
DETAILED DESCRIPTION OF THE INVENTION
[0046] It should be appreciated that the particular implementations shown and described herein are examples and are not intended to otherwise limit the scope of the application in any way.
[0047] The published patents, patent applications, websites, company names, and scientific literature referred to herein are hereby incorporated by reference in their entirety to the same extent as if each was specifically and individually indicated to be incorporated by reference. Any conflict between any reference cited herein and the specific teachings of this specification shall be resolved in favor of the latter. Likewise, any conflict between an art-understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this specification shall be resolved in favor of the latter.
[0048] As used in this specification, the singular forms "a," "an" and "the" specifically also encompass the plural forms of the terms to which they refer, unless the content clearly dictates otherwise. The term "about" is used herein to mean approximately, in the region of, roughly, or around. When the term "about" is used in conjunction with a numerical value or range, it modifies that value or range by extending the boundaries above and below the numerical values or ranges set forth. In general, the term "about" is used herein to modify a numerical value or range above and below the stated value or range by a variance of 20%. [0049] Furthermore, "and/or" is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, the term "and/or" as used in a phrase such as "A and/or B" is intended to include "A and B," "A or B," "A" (alone), and "B" (alone). Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
[0050] Units, prefixes, and symbols are denoted in their Systeme International de Unites
(SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Unless otherwise indicated, amino acid sequences are written left to right in amino to carboxy orientation. The headings provided herein are not limitations of the various aspects of the disclosure, which can be had by reference to the specification as a whole. Accordingly, the terms defined immediately below are more fully defined by reference to the specification in its entirety. Technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which the present application pertains, unless otherwise defined. For example, Goodman and Oilman's "The Pharmaceutical Basis of Therapeutics," 12th Edition, 2011, McGraw-Hill Companies, Inc. The Concise Dictionary of Biomedicine and Molecular Biology, Juo, Pei-Show, 2nd ed., 2002, CRC Press; The Dictionary of Cell and Molecular Biology, 3rd ed., 1999, Academic Press; and the Oxford Dictionary Of Biochemistry And Molecular Biology, Revised, 2000, Oxford University Press, provide one of skill with a general dictionary of many of the terms used in this disclosure.
[0051] The terms "stability" and "stable" as used herein in the context of a formulation comprising BiTE® molecules (e.g., MT103 or MEDI-565) refer to the resistance of the antibody (including antibody fragment thereof) in the formulation to aggregation, degradation or fragmentation under given manufacture, preparation, transportation and storage conditions. The "stable" formulations of the invention retain biological activity under given manufacture, preparation, transportation and storage conditions. The stability of said antibody (including antibody fragment thereof) can be assessed by degrees of aggregation, degradation or fragmentation, as measured by HPSEC, static light scattering (SLS), Fourier Transform Infrared Spectroscopy (FTIR), circular dichroism (CD), urea unfolding techniques, intrinsic tryptophan fluorescence, differential scanning calorimetry, and/or ANS binding techniques, compared to a reference formulation. The overall stability of a formulation comprising BiTE® molecules can be assessed by various immunological assays including, for example, ELISA and radioimmunoassay using isolated antigen molecules.
[0052] "Isolated" when used to describe the various polypeptides and antibodies disclosed herein, means a polypeptide or antibody that has been identified, separated and/or recovered from a component of its production environment. In some embodiments, the isolated polypeptide is free of association with all other components from its production environment. Contaminant components of its production environment, such as that resulting from recombinant transfected cells, are materials that would typically interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. In specific embodiments, the polypeptide will be purified (1) to a degree sufficient to obtain at least 15 residues of N- terminal or internal amino acid sequence by use of a spinning cup sequenator, or (2) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or silver stain. In some embodiments, an isolated antibody will be prepared by at least one purification step.
[0053] The term "excipient" as used herein refers to an inert substance which is commonly used as a diluent, vehicle, preservative, binder or stabilizing agent for drugs which imparts a beneficial physical property to a formulation, such as increased protein stability, increased protein solubility, and decreased viscosity. Examples of excipients include, but are not limited to, proteins (for example, but not limited to, serum albumin), surfactants (for example, but not limited to, SDS, Tween 20, Tween 80, polysorbate, polysorbate 80 and nonionic surfactants), saccharides (for example, but not limited to, glucose, sucrose, maltose and trehalose), polyols (for example, but not limited to, mannitol and sorbitol), fatty acids and phospholipids (for example, but not limited to, alkyl sulfonates and caprylate). For additional information regarding excipients, see Remington's Pharmaceutical Sciences (by Joseph P. Remington, 18th ed., Mack Publishing Co., Easton, Pa.), which is incorporated herein in its entirety. As used herein, the term "trehalose" refers to anhydrous trehalose or trehalose dihydrate. [0054] The term "lyophilized" or "freeze-dried" includes a state of a substance that has been subjected to a drying procedure such as lyophilization, where at least 50% of moisture has been removed.
[0055] The phrase "bulking agent" includes a compound that is pharmaceutically acceptable and that adds bulk to a lyo cake. Bulking agents known to the art include, for example, carbohydrates, including simple sugars such as dextrose, ribose, fructose and the like, alcohol sugars such as mannitol, inositol and sorbitol, disaccharides including trehalose, sucrose and lactose, naturally occurring polymers such as starch, dextrans, chitosan, hyaluronate, proteins (e.g., gelatin and serum albumin), glycogen, and synthetic monomers and polymers.
[0056] Reference is made herein to various methodologies and materials known to those of skill in the art.
Pharmaceutical Formulations
[0057] In embodiments, provided herein are pharmaceutical formulations comprising a bispecific antibody.
[0058] As used herein, the terms "antibody" and "antibodies" (immunoglobulins) encompass monoclonal antibodies (including full-length monoclonal antibodies), polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, human antibodies, humanized antibodies, camelised antibodies, chimeric antibodies, single-chain Fvs (scFv), single-chain antibodies, single domain antibodies, domain antibodies, Fab fragments, F(ab')2 fragments, antibody fragments that exhibit the desired biological activity, disulfide-linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), intrabodies, and epitope- binding fragments of any of the above. Antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen-binding site. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass.
[0059] As used herein, "bispecific antibody," "bispecific single chain antibody" or
"BiTE®" refer to an antibody comprising a first binding domain and a second binding domain that comprises a variable heavy (VH) domain and a variable light (VL) domain of an antibody. Suitably, bispecific antibodies for use in the formulations and methods described herein comprise a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably, the bispecific antibodies described herein comprise a second binding domain that comprises a VH domain and a VL domain of an antibody that immunospecifically binds to an antigen of interest (e.g., CD 19 or EpbA2 or CEA). The first and second binding domains of a bispecific antibody may be derived from synthetic antibodies, monoclonal antibodies, recombinantly produced antibodies, multispecific antibodies (including bi- specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, intrabodies, single-chain Fvs (scFvs) (e.g., including monospecific and bi-specific, etc.), Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
[0060] In suitable embodiments, the formulations provided herein comprise a bispecific single chain antibody comprising a first binding domain that comprises a variable heavy (VH) domain and a variable light (VL) domain of an antibody that immunospecifically binds to the T-cell antigen CD3 and a second binding domain that comprises a VH domain and a VL domain of an antibody that immunospecifically binds to carcinoembryonic antigen (CEA). Carcinoembryonic antigen (CEA) is a glycosylated human oncofetal antigen that belongs to the CEA-related cell adhesion molecule (CEACAM) family of the immunoglobulin gene superfamily. CEA has been suggested to mediate cell-cell adhesion, facilitate bacterial colonization of the intestine, and protect the colon from microbial infection by binding and trapping infectious microorganisms. Carcinoembryonic antigen (CEA) is a well-characterized tumor-associated antigen that is frequently over-expressed in human carcinomas and melanomas.
[0061] FIG. 1A shows a schematic representation of a bispecific antibody described herein comprising a first binding domain that binds to CD3 and a second binding domain that binds to CEA. FIG. IB shows a schematic representation of a bispecific antibody comprising a CD3 binding domain and a carcinoembryonic antigen binding domain targeting and killing tumor cells. FIG. 1C shows the aggregation propensity of a bispecific antibody (MEDI-565; circles) compared to a monoclonal, whole IgGl antibody (squares) incubated at 25 °C as measured by HPSEC. As demonstrated, the bispecific antibody (MEDI-565) aggregates at a very high rate compared to a monoclonal IgGl antibody, underscoring the difficulties in formulating stable single chain, bi-specific antibodies (e.g. MEDI-565, MT103) that are addressed herein.
[0062] The antibody that immunospecifically binds to the T-cell antigen CD3 and the antibody that immunospecifically binds CEA may be derived from synthetic antibodies, monoclonal antibodies, recombinantly produced antibodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, intrabodies, single- chain Fvs (scFvs) (e.g., including monospecific and bi- specific, etc.), Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above. Suitably, antibodies of the formulations described herein include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen-binding domain that immunospecifically binds to an antigen of interest.
[0063] As used herein, the term "binding domain" refers to a domain comprising a three- dimensional structure capable of immunospecifically binding to an epitope. Thus, said domain can comprise the VH and/or VL domain of an antibody chain. In some embodiments, said domain comprises at least the VH domain. On the other hand, said binding domains contained in the bispecific antibodies of a formulation of the invention may comprise at least one complementarity determining region (CDR) of an antibody chain recognizing the CD 19 and CEA antigens, respectively.
[0064] As used herein, the term "single-chain Fv" or "scFv" refers to antibody fragments comprising the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain. Generally, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding. Methods for producing scFvs are well known in the art. For a review of methods for producing scFvs see Pluckthun, in "The Pharmacology of Monoclonal Antibodies," vol. 113, Rosenburg and Moore eds. Springer- Verlag, New York, pp. 269-315 (1994).
[0065] All formulations of antibodies and/or antibody fragments that specifically bind to an antigen of interest are herein collectively referred to as "pharmaceutical formulations of the invention," "formulations of the invention", "liquid formulations of the invention", "lyophilized formulations of the invention", "reconstituted formulations of the invention", "antibody liquid formulations of the invention", "antibody lyophilized formulations of the invention", "antibody reconstituted formulations of the invention" or "antibody formulations of the invention".
[0066] As used herein the phrases "pharmaceutical formulation" and "formulation" are used interchangeably and refer to a composition comprising a bispecific antibody and one or more appropriate buffers and/or excipients. Suitably, the pharmaceutical formulations described herein are "pharmaceutically acceptable," and thus would meet the necessary approval requirements required by a regulatory agency of the Federal or a state government, or listed in the U.S. Pharmacopeia, European Pharmacopeia, or other generally recognized pharmacopeia, so as to be used in animals, and more particularly in humans.
[0067] In suitable embodiments, the pharmaceutical formulations described herein comprise about 1.0 mg/mL to about 10.0 mg/mL of a bispecific antibody, more suitably about 1.0 mg/mL to about 5.0 mg/mL of a bispecific antibody. For example, the formulations described herein comprise about 1.0 mg/mL to about 7 mg/mL, about 1.0 mg/mL to about 6 mg/mL, about 1.0 mg/mL to about 5 mg/mL, about 1.0 mg/mL to about 4 mg/mL, about 1.0 mg/mL to about 3.5 mg/mL, about 1.0 mg/mL to about 3 mg/mL, about 1.5 mg/mL to about 3 mg/mL, about 1.5 mg/mL to about 2.5 mg/mL, about 1.7 mg/mL to about 3 mg/mL, about 1.7 mg/mL to about 2.5 mg/mL, or about 1.0 mg/mL, about 1.1 mg/mL, about 1.2 mg/mL, about 1.3 mg/mL, about 1.4 mg/mL, about 1.5 mg/mL, about 1.6 mg/mL, about 1.7 mg/mL, about 1.8 mg/mL, about 1.9 mg/mL, about 2.0 mg/mL, about 2.1 mg/mL, about 2.2 mg/mL, about 2.3 mg/mL, about 2.4 mg/mL, about 2.5 mg/mL, about 2.6 mg/mL, about 2.7 mg/mL, about 2.8 mg/mL, about 2.9 mg/mL, about 3.0 mg/mL, about 3.1 mg/mL, about 3.2 mg/mL, about 3.3 mg/mL, about 3.4 mg/mL, about 3.5 mg/mL, about 3.6 mg/mL, about 3.7 mg/mL, about 3.8 mg/mL, about 3.9 mg/mL, about 4.0 mg/mL, about 4.1 mg/mL, about 4.2 mg/mL, about 4.3 mg/mL, about 4.4 mg/mL, about 4.5 mg/mL, about 4.6 mg/mL, about 4.7 mg/mL, about 4.8 mg/mL, about 4.9 mg/mL, or about 5.0 mg/mL of a bispecific antibody, including any ranges or values within these ranges.
[0068] The pharmaceutical formulations described herein suitably further comprise one or more buffers. As used herein, "buffer" refers to an excipient for maintaining the pH of a formulation. Exemplary buffers for use in the pharmaceutical formulations provided herein include, but are not limited to histidine, citrate, phosphate, succinate, glycine, and acetate. Suitably, the buffer for use in the pharmaceutical formulations described herein is citrate.
[0069] The concentration of a buffer, suitably citrate, in the pharmaceutical formulations described herein is generally in the range of about 10 mM to about 100 mM, more suitably about 15 mM to about 80 mM, about 15 mM to about 60 mM, about 20 mM to about 60 mM, about 20 mM to about 50 mM, about 20 mM to about 40 mM, about 20 mM to about 30 mM, or about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM or about 60 mM, including any ranges or values within these ranges.
[0070] The pharmaceutical formulations provided herein also suitably further comprise a lyoprotectant. As used herein, a "lyoprotectant" refers to a molecule which, when amorphous and combined with a protein (e.g., an antibody such as a bispecific antibody), provides retention of a protein's physical and chemical stability and integrity upon lyophilization and storage, and when crystallized, a lyoprotectant acts as a bulking agent. Lyoprotectants include, but are not limited to, sugars and their corresponding sugar alcohols; amino acids such as monosodium glutamate or histidine; a methylamine such as betaine; a lyotropic salt such as magnesium sulfate; a polyol such as trihydric or higher molecular weight sugar alcohols, e.g. glycerin, dextran, erythritol, glycerol, arabitol, xylitol, sorbitol, and mannitol; propylene glycol; polyethylene glycol; Pluronics®; and combinations thereof. Additional examples of lyoprotectants include, but are not limited to, glycerin and gelatin, and the sugars mellibiose, melezitose, raffinose, mannotriose and stachyose. Examples of reducing sugars include, but are not limited to, glucose, maltose, lactose, maltulose, iso-maltulose and lactulose. Examples of non-reducing sugars include, but are not limited to, trehalose, non-reducing glycosides of polyhydroxy compounds selected from sugar alcohols and other straight chain polyalcohols. Examples of sugar alcohols include, but are not limited to, monoglycosides, compounds obtained by reduction of disaccharides such as lactose, maltose, lactulose and maltulose. The glycosidic side group can be either glucosidic or galactosidic. Additional examples of sugar alcohols include, but are not limited to, glucitol, maltitol, lactitol and iso-maltulose. In specific embodiments, trehalose is used as a lyoprotectant in the formulations described herein. [0071] Suitably, the lyoprotectant is added to the pharmaceutical formulations in a
"lyoprotecting amount" which means that, following lyophilization of the protein (bispecific antibody) in the presence of the lyoprotecting amount of the lyoprotectant, the protein essentially retains its physical and chemical stability and integrity upon lyophilization and storage. In embodiments, the lyoprotectant, suitably trehalose, is provided in the pharmaceutical formulations at a concentration of about 20 mM to about 300 mM, more suitably, about 25 mM to about 0.21 M, for example, about 30 mM to about 0.21 M, about 40 mM to about 0.21 M, about 60 mM to about 0.21 M, about 60 mM to about 150 mM, about 60 mM to about 100 mM, about 60 mM to about 80 mM, or about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, about 60 mM, about 65 mM, about 70 mM, about 75 mM, about 80 mM, about 85 mM, about 90 mM, about 95 mM, about 100 mM, about 125 mM, about 150 mM, about 200 mM, or about 0.21 M, including any ranges or values within these ranges.
[0072] In suitable embodiments, the pharmaceutical formulations also further comprise a surfactant. The term "surfactant" as used herein refers to organic substances having amphipathic structures; namely, they are composed of groups of opposing solubility tendencies, typically an oil- soluble hydrocarbon chain and a water-soluble ionic group. Surfactants can be classified, depending on the charge of the surface-active moiety, into anionic, cationic, and nonionic surfactants. Surfactants are often used as wetting, emulsifying, solubilizing, and dispersing agents for various pharmaceutical formulations and preparations of biological materials. Pharmaceutically acceptable surfactants like polysorbates (e.g. polysorbates 20, 40, 60 or 80); polyoxamers (e.g. poloxamer 188); Triton; sodium octyl glycoside; lauryl-, myristyl-, linoleyl- , or stearyl-sulfobetaine; lauryl-, myristyl-, linoleyl- or stearyl-sarcosine; linoleyl-, myristyl-, or cetyl-betaine; lauroamidopropyl-, cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-betaine (e.g. lauroamidopropyl); myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-dimethylamine; sodium methyl cocoyl-, or disodium methyl oleyl-taurate; and the MONAQUA™ series (Mona Industries, Inc., Paterson, N. J.), polyethyl glycol, polypropyl glycol, and copolymers of ethylene and propylene glycol (e.g. Pluronics, PF68 etc), can be used in the pharmaceutical formulations described herein. Suitably the surfactant is polysorbate 80. [0073] Suitably, the pharmaceutical formulations described herein comprise a surfactant
(suitably polysorbate 80) at about 0.001% to about 0.5% (by volume), more suitably about 0.002% to about 0.1% of a surfactant, for example about 0.005% to about 0.2%, about 0.002% to about 0.05%, about 0.006% to about 0.1%, about 0.007% to about 0.08%, about 0.008% to about 0.07%, about 0.009% to about 0.05%, about 0.01% to about 0.05%, about 0.01% to about 0.04%, about 0.01% to about 0.03%, or about 0.002%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.010%, about 0.011%, about 0.012%, about 0.013%, about 0.014%, about 0.015%, about 0.016%, about 0.017%, about 0.018%, about 0.019%, about 0.020%, about 0.021%, about 0.022%, about 0.023%, about 0.024%, about 0.025%, about 0.026%, about 0.027%, about 0.028%, about 0.029%, about 0.03%, about 0.035%, about 0.04%, about 0.045%, or about 0.05% of a surfactant, including any ranges or values within these ranges.
[0074] The pharmaceutical formulations described herein can also include one or more additional excipients, including for example, one or more sugars, salts, amino acids, polyols, chelating agents, emulsifiers and/or preservatives.
[0075] Suitably, the formulations provided herein are formulated at a pharmaceutically acceptable pH, for example, in the range of about pH 5.0 to about 7.0, more suitably about pH 6.0 to about pH 7.0, or about pH 5.0, pH 5.5, pH 6.0, pH 6.5 or pH 7.0.
[0076] As described herein and in the Examples, it has been surprisingly found that the disclosed liquid, lyophilized and/or frozen pharmaceutical formulations allow for use of high amounts of bispecific antibodies while exhibiting stability for extended storage and shipping, prior to administration to a patient.
[0077] In exemplary embodiments, the pharmaceutical formulations described herein comprise about 1.7 mg/mL to about 2.5 mg/mL of a bispecific antibody, including for example about 1.7 mg/mL or about 2.5 mg/mL of the bispecific antibody. In embodiments, the pharmaceutical formulations comprise about 20 mM to about 30 mM citrate, including for example about 20 mM citrate or about 30 mM citrate. The pharmaceutical formulations also suitably further comprise about 60 mM to about 80 mM of the lyoprotectant, including for example 70 mM of the lyoprotectant. In addition, the pharmaceutical formulations also suitably further comprise about 0.008% to about 0.012% of the surfactant, including for example about 0.01% of the surfactant. [0078] As discussed herein, suitable lyoprotectants for use in the pharmaceutical formulations include, but are not limited to, trehalose, lactose, mannitol, raffinose and sucrose. Suitable surfactants include, but are not limited to, polysorbate-20, polysorbate- 40, polysorbate-60 and polysorbate-80.
[0079] In embodiments, pharmaceutical formulations are provided which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably, the formulations comprise about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody, about 20 mM to about 30 mM citrate, about 60 mM to about 80 mM trehalose, and about 0.002% to about 0.1% polysorbate 80. In suitable embodiments, the formulations have a pH of about 5.0 to about 7.0.
[0080] In further embodiments, pharmaceutical formulations are provided which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably, the formulations comprise about 1.7 mg/mL of the bispecific antibody, about 20 mM citrate, about 70 mM trehalose, and about 0.01% polysorbate 80. Suitably, the formulations have a pH of about 6.0.
[0081] In some embodiments, pharmaceutical formulations are provided which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably, the formulations comprise about 2.5mg/ml of the bispecific antibody, about 30mM Citrate, about 80mM Trehalose and about 0.01% polysorbate 80. Suitably, the formulations have a pH of about 6.0.
[0082] In additional embodiments, pharmaceutical formulations are provided which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably, the formulations comprise about 5.0 mg/mL of the bispecific antibody, about 60 mM citrate, about 0.21 M trehalose, and about 0.03% polysorbate 80. Suitably, the formulation has a pH of about 6.0.
[0083] In additional embodiments, pharmaceutical formulations are provided which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably, the formulations comprise the components described in Table 2 or Figures 5 and 6 (as described in Example 5), in particular, 2.5mg/ml, 30mM Citrate, 80mM Trehalose, 0.01% polysorbate 80, pH 6; 1.7mg/ml, 20mM Citrate, 70mM Trehalose, 0.01% polysorbate 80, pH 6; or 1.7mg/ml, 30mM Citrate, 60mM Trehalose, 0.01% polysorbate 80, pH 6.
[0084] The pharmaceutical formulations described herein can have any desired volume for shipping, storage and/or administration to a patient. Suitably, the volume of the pharmaceutical formulations are provided such that they are easily produced in mass quantities, placed in suitable vials or containers for storage and then shipped to a distribution center and/or directly to a hospital, clinic, doctor's office, or other setting where the pharmaceutical formulations can be administered to a patient.
[0085] Suitably, the formulations are prepared in sterile water or water for injection at the desired volume.
[0086] In exemplary embodiments, the pharmaceutical formulations have a volume of about 0.1 mL to about 20.0 mL, more suitably about 0.5 mL to about 15.0 mL, about 0.5 mL to about 12.0 mL, about 1.0 mL to about 10.0 mL, about 1.0 mL to about 5.0 mL, about 1.0 mL to about 2.0 mL or about 0.5 mL, about 0.6 mL, about 0.7 mL, about 0.8 mL, about 0.9 mL, about 1.0 mL, about 1.1 mL, about 1.2 mL, about 1.3 mL, about 1.4 mL, about 1.5 mL, about 1.6 mL, about 1.7 mL, about 1.8 mL, about 1.9 mL, about 2.0 mL, about 2.1 mL, about 2.2 mL, about 2.3 mL, about 2.4 mL, about 2.5 mL, about 2.6 mL, about 2.7 mL, about 2.8 mL, about 2.9 mL, or about 3.0 mL, including any ranges or values within these ranges.
[0087] While in suitable embodiments, the pharmaceutical formulations described herein are liquid formulations, i.e., pharmaceutical formulations prepared in sterile water or water for injection (WFI), the pharmaceutical formulations can also be frozen formulations or lyophilized formulations.
[0088] In embodiments, it is desirable to prepare frozen formulations by providing a liquid pharmaceutical formulation as described herein, and freezing the formulation under appropriate conditions. For example, the frozen formulations can be provided by freezing the liquid formulations to less than 0° C, more suitably to about -20° C, about -40° C, about -60° C, or suitably to about -80° C. [0089] Suitable protocols and methods for preparing lyophilized pharmaceutical formulations from liquid and/or frozen formulations are known in the art and described herein.
Lyophilized Pharmaceutical Formulations
[0090] In further embodiments, lyophilized pharmaceutical formulations are provided.
As described herein, such formulations have been prepared from the liquid and/or frozen formulations provided throughout by removing water from the formulations to yield a freeze-dried formulation. The lyophilized pharmaceutical formulations provided herein are described as containing recited weight amounts of components, indicating dried formulations, rather than a liquid formulation.
[0091] In embodiments, lyophilized pharmaceutical formulations are provided which comprise a bispecific antibody, the bispecific antibody comprising a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen. Suitably the lyophilized pharmaceutical formulations comprise about 2 mg to about 10 mg of the bispecific antibody, about 15 mg to about 40 mg citrate, about 100 mg to about 200 mg trehalose and about 0.01 μg to about 0.08 μg polysorbate 80.
[0092] In suitable embodiments, the lyophilized pharmaceutical formulations comprise about 3 mg to about 10 mg of the bispecific antibody, or about 3 mg to about 8 mg, about 5 mg to about 8 mg, or about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg or about 10 mg of the bispecific antibody, including any ranges or values within these ranges. Suitably, the lyophilized formulations comprise about 20 mg to about 30 mg citrate, or about 20 mg, about 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg or about 30 mg citrate, including any ranges or values within these ranges. In embodiments, the lyophilized pharmaceutical formulations comprise about 100 mg to about 180 mg trehalose, or about 100 mg to about 150 mg, or about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg or about 150 mg trehalose, including any ranges or values within these ranges. Suitably the lyophilized pharmaceutical formulations comprise about 0.02 μg to about 0.08 μg polysorbate 80, or about 0.03 μg to about 0.06 μg, or about 0.02 μg, about 0.03 μg, about 0.04 μg, about 0.05 μg, about 0.06 μg, about 0.07 μg, or about 0.08 μg polysorbate 80, including any ranges or values within these ranges.
[0093] In suitable embodiments, the lyophilized pharmaceutical formulations comprise about about 2.5 mg to about 61 mg of the bispecific antibody, about 8.5 mg to about 204 mg of citrate, about 40 mg to about 954mg trehalose and about 0.15 μg to about 3.6 μg of polysorbate 80, more suitably about 7 mg of the bispecific antibody, about 25 mg or about 24.95 mg citrate, about 117 mg or about 116.5 mg trehalose, and about 0.4 μg polysorbate 80.
[0094] The lyophilized pharmaceutical formulations are designed to provide enhanced stability and limited aggregation upon reconstitution. A "reconstituted" formulation is one which has been prepared by dissolving a lyophilized pharmaceutical formulation in a diluent such that the bispecific antibody is dispersed in the reconstituted formulation (i.e., the act of reconstitution). The reconstituted formulation is suitable for administration (e.g. parenteral administration) to a patient to be treated with the bispecific antibody of interest and, in certain embodiments of the invention, may be one which is suitable for parenteral administration, including for example subcutaneous administration or injection. The "diluent" of interest herein is one which is pharmaceutically acceptable (safe and nontoxic for administration to a human) and is useful for the preparation of a liquid pharmaceutical formulation, such as a pharmaceutical formulation reconstituted after lyophilization. In some embodiments, diluents include, but are not limited to, sterile water, bacteriostatic water for injection (BWFI), water for injection (WFI), a pH buffered solution (e.g. phosphate -buffered saline), sterile saline solution, Ringer's solution or dextrose solution, though suitably water (sterile, WFI or BWFI) is used.
[0095] An "isotonic" pharmaceutical formulation is one which has essentially the same osmotic pressure as human blood. Isotonic pharmaceutical formulations generally have an osmotic pressure from about 250 to 350 mOsm. The term "hypotonic" describes a pharmaceutical formulation with an osmotic pressure below that of human blood. Correspondingly, the term "hypertonic" is used to describe a pharmaceutical formulation with an osmotic pressure above that of human blood. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer.
Stability of Pharmaceutical Formulations [0096] In exemplary embodiments, the lyophilized pharmaceutical formulations described herein are stable for extended periods of storage at room temperature. As used herein, room temperature is generally in the range of about 22-25° C. The lyophilized pharmaceutical formulations are also suitably stable after storage at 25° C for 6 months or more. In additional embodiments, the lyophilized pharmaceutical formulations are stable after storage at 2-8° C (e.g. 5° C) for 24 months or more. As used herein, the term "stable" for a period of storage (or "stability") is used to indicate that the bispecific antibodies in the lyophilized pharmaceutical formulations resist aggregation, degradation and/or fragmentation following reconstitution; or the bispecific antibodies in the liquid pharmaceutical formulations resist aggregation, degradation and/or fragmentation. The stability of the bispecific antibodies can be assessed by degrees of aggregation, degradation or fragmentation, as measured by high performance size exclusion chromatography (HPSEC), static light scattering (SLS), Fourier Transform Infrared Spectroscopy (FTIR), circular dichroism (CD), urea unfolding techniques, intrinsic tryptophan fluorescence, differential scanning calorimetry, and/or ANS binding techniques, compared to a reference.
[0097] The overall stability of a pharmaceutical formulation comprising bispecific antibodies can be assessed by various immunological assays including, for example, ELISA and radioimmunoassay using isolated antigen molecules.
[0098] The phrase "low to undetectable levels of aggregation" as used herein refers to pharmaceutical formulations containing no more than about 5%, no more than about 4%, no more than about 3%, no more than about 2%, no more than about 1%, or no more than about 0.5% aggregation by weight of protein as measured by high performance size exclusion chromatography (HPSEC) or static light scattering (SLS) techniques. Suitably, the lyophilized pharmaceutical formulations exhibit < 5.0% aggregation when reconstituted, more suitably < 4.0% aggregation when reconstituted, < 3.0% aggregation when reconstituted, < 2.0% aggregation when reconstituted, < 1.0% aggregation when reconstituted, or 0.5% aggregation when reconstituted. Suitably, the liquid pharmaceutical formulations exhibit < 5.0% aggregation, more suitably < 4.0% aggregation, < 3.0% aggregation, < 2.0% aggregation, < 1.0% aggregation, or 0.5% aggregation. [0099] The term "low to undetectable levels of fragmentation" as used herein refers to pharmaceutical formulations containing equal to or more than about 80%, about 85%, about 90%, about 95%, about 98% or about 99% of the total bispecific antibody, for example, in a single peak as determined by HPSEC, or reduced Capillary Gel Electrophoresis (rCGE), representing the non-degraded bispecific antibody or a non- degraded fragment thereof, and containing no other single peaks having more than about 5%, more than about 4%, more than about 3%, more than about 2%, more than about 1%, or more than about 0.5% of the total bispecific antibody in each.
[00100] In additional embodiments, the pharmaceutical formulations described herein consist of the recited components, such that no additional components beyond those recited are allowed. In further embodiments, the pharmaceutical formulations described herein consist essentially of the recited components. In such embodiments, the addition of components that alter the stability of the bispecific antibody are considered a material alteration to such pharmaceutical formulations and are thus excluded from such methods that consist essentially of the recited components.
[00101] As described throughout, it has been surprisingly discovered that the addition of citrate, at the recited amounts, to pharmaceutical formulations comprising bispecific antibodies (including, e.g., MEDI-565 and MT103) increases the stability, and suitably, lowers aggregation following reconstitution of a pharmaceutical formulation. This allows the preparation of pharmaceutical formulations containing amounts of bispecific antibodies in the 1.7 mg/L to 5 mg/mL range, and for example, bispecific antibodies (including, e.g., MEDI-565 and MT103) at 5 mg/mL, so as to allow for parenteral administration, including subcutaneous administration or injection if a large amount of bispecific antibody.
Methods of Preparation of Pharmaceutical Formulations
[00102] In further embodiments, methods are provided for preparing pharmaceutical formulations, as well as pharmaceutical formulations prepared by such methods. Such methods suitably comprise preparing a liquid pharmaceutical formulation in accordance with the various embodiments described herein, suitably comprising a bispecific antibody (e.g., at about 1.0 mg/mL to about 5.0 mg/mL of the bispecific antibody), citrate (e.g., about 15 mM to about 60 mM citrate), a lyoprotectant (e.g., about 25 mM to about 0.21 M of a lyoprotectant) and a surfactant (e.g., about 0.002% to about 0.1% of a surfactant). The liquid pharmaceutical formulation is then lyophilized to produce a lyophilized formulation. The lyophilized formulation is then reconstituted in a volume suitable for parenteral administration.
[00103] As described throughout, the methods of preparation can also further comprise freezing the liquid pharmaceutical formulation (e.g., to a temperature of less than 0° C, more suitably to about -20° C, about -40° C, about -60° C, or suitably to about -80° C), prior to lyophilizing the formulation. It should be noted that while the freezing and lyophilizing are suitably conducted in a single facility within a relatively close amount of time (hours to days to weeks), in embodiments, the pharmaceutical formulations can be frozen (e.g., to about -80 C) at a first facility, shipped in the frozen state (if desired), thawed and then lyophilized at a later time in the first facility or in a second facility (i.e., days to weeks to months or even a year).
[00104] The methods described herein suitably utilize a reconstituting volume, i.e., the volume of desired diluent such as sterile water or WFI, that is on the order of about 1/2 to about 1/3 of the volume of the initial, starting liquid pharmaceutical formulation that was prepared and then lyophilized. It has been surprisingly found that by utilizing the components and amounts recited herein in the pharmaceutical formulations, the lyophilized pharmaceutical formulations can be reconstituted in a reduced volume, i.e., about 1/2 to about 1/3 of the starting volume, so as to increase the final amount of bispecific antibody, yet still maintain the desired stability (i.e., low aggregation and/or fragmentation) and also still maintain a time of reconstitution on the order of minutes (e.g., 5-15 minutes), so as to allow for simple use in clinical settings and to increase patient and physician compliance.
[00105] In embodiments, the total amount of volume for the reconstitution of the lyophilized pharmaceutical formulations is on the order of about 0.5 mL to about 12.0 mL, for example, about 0.5 mL to about 10.0 mL, about 0.5 mL to about 8.0 mL, about 0.5 mL to about 6.0 mL, about 0.5 mL to about 5.0 mL, about 0.5 mL to about 4.0 mL, about 0.5 mL to about 3.0 mL, about 0.5 mL to about 2.0 mL, or about 0.5 mL, about 0.6 mL, about 0.7 mL, about 0.8 mL, about 0.9 mL, about 1.0 mL, about 1.1 mL, about 1.2 mL, about 1.3 mL, about 1.4 mL, about 1.5 mL, about 1.6 mL, about 1.7 mL, about 1.8 mL, about 1.9 mL, or about 2.0 mL total volume. In embodiments, the total volume for reconstitution is a volume suitable for parenteral administration, including subcutaneous administration or injection.
Bispecific Antibody Sequences
[00106] As described throughout, in suitable embodiments, the bispecific antibodies for use in the pharmaceutical formulations comprise a first binding domain that specifically binds the CD3 T cell surface antigen. In exemplary embodiments, the first binding domain comprises amino acids 283-525 of SEQ ID NO: 5 of WO 2009/070642, published June 4, 2009 corresponding to US 2010/0303827 (published December 2, 2010), (SEQ ID NO: 1 in the present disclosure), the entire disclosure of which is incorporated by reference herein for all purposes, including for the disclosure of the bispecific antibody sequences recited therein and in the Sequence Listing.
[00107] In suitable embodiments, the second binding domain of the bispecific antibodies used in the pharmaceutical formulations described herein specifically bind an antigen selected from the group consisting of CD19, CD20, CD22, EphA2, EphA4, INFR, ICOS, Ep-CAM, CEA, and IL-5 receptor. In suitable embodiments, the second binding domain specifically binds CEA, and comprises the amino acid sequence of any one of SEQ ID NOs: 28-44 and 46-51 of US 2013/0035249, published February 7, 2013 (SEQ ID NOs: 2-24 of the present disclosure), the entire disclosure of which is incorporated by reference herein for all purposes, including for the disclosure of the bispecific antibody sequences recited therein and in the Sequence Listing. In further embodiments, the second binding domain of the bispecific antibody comprises the amino acid sequence of residues 28-277 of SEQ ID NO:5 from WO2009/070642, corresponding to US 2010/0303827, (SEQ ID NO: 25 herein).
[00108] In exemplary embodiments, the bispecific antibody comprises the amino acid sequence of SEQ ID NO:26, also known as "MEDI-565," a bispecific single chain antibody of the BiTE class that includes an anti-CEA binding portion and an anti-CD3 binding portion. The anti-CEA binding portion is a humanized scFv derived from mouse monoclonal antibody A5B7. MEDI-565 is described and disclosed in WO07/071426, Lutterbuese et al., 2009, Journal of Immunother 32: 341-352, and Osada et al. 2010, British Journal of Cancer, 102: 124-33; each of which is incorporated by reference herein for all purposes. [00109] In additional embodiments, the bispecific antibody comprises the amino acid sequence of residues 28-525 of SEQ ID NO 5 from WO2009/070642, corresponding to US 2010/0303827, (SEQ ID NO: 27 herein), the entire disclosure of which is incorporated by reference herein for all purposes, including for the disclosure of the bispecific antibody sequences recited therein and in the Sequence Listing.
[00110] In a further embodiment, the formulations of the invention comprise a BiTE® molecule. In additional embodiments, a formulation of the invention comprises a BiTE® wherein the BiTE® molecule comprises an anti-CEA VL domain having the amino acid sequence of SEQ ID NO:21, an anti-CEA VH domain having the amino acid sequence of SEQ ID NO:22. In additional embodiments, a formulation of the invention comprises a BiTE® molecule wherein the BiTE® molecule comprises an anti-CD19 VL domain having the amino acid sequence of SEQ ID NO:28, an anti-CD19 VH domain having the amino acid sequence of SEQ ID NO:29. In additional embodiments, a formulation of the invention comprises a BiTE® molecule wherein the BiTE® molecule comprises an anti- CD3 VH domain having the amino acid sequence of SEQ ID NO:30 and an anti-CD3 VL domain having the amino acid sequence of SEQ ID NO:31.
[00111] The invention encompasses stable formulations comprising a single BiTE® molecule, for example, a BiTE® molecule comprising at least a first and a second binding site specific for the CD3 and CEA antigens, respectively; or at least a first and a second binding site specific for the CD3 and CD 19 antigens, respectively. The invention also encompasses formulations comprising two or more bispecific antibodies, for example, bispecific antibodies comprising at least a first and a second binding site specific for the CD3 and CEA antigens, respectively; or at least a first and a second binding site specific for the CD3 and CD19 antigens, respectively. In a specific embodiment, a formulation of the invention comprises MEDI-565 or MT103. In another embodiment, a formulation of the invention comprises two or more bispecific antibodies, wherein one of the bispecific antibodies is MEDI-565. In another embodiment, a formulation of the invention comprises two or more bispecific antibodies, wherein one of the bispecific antibodies is MT103.
[00112] The following table provides exemplary sequences of the first and second binding domains of the bispecific antibodies, as well as exemplary sequences for the entire bispecific antibody, for use in the pharmaceutical formulations described herein. Table 1: Sequences of Exemplary Bispecific Antibodies and/or Binding Domains
DESCRIPTION AMINO ACID SEQUENCE SEQ ID NO:
First Binding DIKLQQSGAELARPGASVKMSCKTSGYTFTR 1 Domain (amino YTMHWVKQRPGQGLEWIGYINPSRGYTNYN
acids 283-525 of QKFKDKATLTTDKSSSTAYMQLSSLTSEDSA
SEQ ID NO: 5 of VYYCARYYDDHYCLDYWGQGTTLTVSSVE
WO 2009/070642) GGSGGSGGSGGSGGVDDIQLTQSPAIMSASP
GEKVTMTCRASSSVSYMNWYQQKSGTSPKR
WIYDTSKVASGVPYRFSGSGSGTSYSLTISSM
EAED AATYYCQQWS SNPLTFG AGTKLELK
Second Binding FYFDY 2 Domain (SEQ ID
NO: 28 of US
2013/0035249)
Second Binding DX1X2X3X4FYFDY 3 Domain (SEQ ID
NO: 29 of US "Xi", "X2", "X3" or " X4" represents any amino
2013/0035249) acid residue, and the amino acid residue "D"
corresponds to Kabat position 95 of CDR-H3 of
murine monoclonal antibody A5B7 and the amino
acid residues "FYFDY" correspond to Kabat
positions 100, 100a, 100b, 101, and 102,
respectively, of CDR-H3 of murine monoclonal
antibody A5B7.
Second Binding RFYFDY 4 Domain (SEQ ID
NO: 30 of US
2013/0035249)
Second Binding LRFYFDY 5 Domain (SEQ ID
NO: 31 of US
2013/0035249)
Second Binding GLRFYFDY 6 Domain (SEQ ID
NO: 32 of US
2013/0035249)
Second Binding RGLRFYFDY 7 Domain (SEQ ID
NO: 33 of US
2013/0035249)
Second Binding DRGLRFYFDY 8 Domain (SEQ ID
NO: 34 of US
2013/0035249)
Second Binding RGLR 9 Domain (SEQ ID NO: 35 of US
2013/0035249)
Second Binding SYWMH 10
Domain (SEQ ID
NO: 36 of US
2013/0035249)
Second Binding FIRNKANGGTTE YMS VKG 11
Domain (SEQ ID
NO: 37 of US
2013/0035249)
Second Binding FILNKANGGTTE YMS VKG 12
Domain (SEQ ID
NO: 38 of US
2013/0035249)
Second Binding TYAMH 13
Domain (SEQ ID
NO: 39 of US
2013/0035249)
Second Binding LISNDGSNKYYADSVKG 14
Domain (SEQ ID
NO: 40 of US
2013/0035249)
Second Binding TLRRGINVGAYSIY 15
Domain (SEQ ID
NO: 41 of US
2013/0035249)
Second Binding YKSDSDKQQGS 16
Domain (SEQ ID
NO: 42 of US
2013/0035249)
Second Binding MIWHSGASAV 17
Domain (SEQ ID
NO: 43 of US
2013/0035249)
Second Binding FILNKANGGTTEYAASVKG 18
Domain (SEQ ID
NO: 44 of US
2013/0035249)
Second Binding QAVLTQPASLSASPGASASLTCTLRRGINVGA 19
Domain (SEQ ID YSIYWYQQKPGSPPQYLLRYKSDSDKQQGSG
NO: 46 of US VSSRFSASKDASANAGILLISGLQSEDEADYY
2013/0035249) CMIWHSGASAVFGGGTKLTVLGGGGSGGGG
SGGGGSEVQLVESGGGLVQPGRSLRLSCAAS
GFTVSSYWMHWVRQAPGKGLEWVGFIRNK
ANGGTTEYAASVKGRFTISRDDSKNTLYLQM
NSLRAEDTAVYYCARDRGLRFYFDYWGQGT
TVTVS SS GGGGSD VQLVQSGAEVKKPGAS V
KVS CKAS G YTFTRYTMHW VRQAPGQGLEWI GYINPSRGYTNYADSVKGRFTITTDKSTSTAY
MELS S LRSEDT AT Y YC ARY YDDH YCLD YWG
QGTTVTVSSGEGTSTGSGGSGGSGGADDIVL
TQSPATLSLSPGERATLSCRASQSVSYMNWY
QQKPGKAPKRWI YDTS KVAS G VP ARFS GS GS
GTDYSLTINSLEAEDAATYYCQQWSSNPLTF
GGGTKVEIK
Second Binding FIRNKANGGTTEYAASVKG 20 Domain (SEQ ID
NO: 47 of US
2013/0035249)
Second Binding OAVLTOPASLSASPGASASLTCTLRRGINVGA 21 Domain (SEQ ID YSIYWYOOKPGSPPOYLLRYKSDSDKOOGSG
NO: 48 of US VSSRFSASKDASANAGILLISGLQSEDEADYY
2013/0035249) CMIWHSGASAVFGGGTKLTVL
Second Binding E VQLVES GGGL VQPGRS LRLS C A AS GFT VS S 22 Domain (SEQ ID YWMHWVROAPGKGLEWVGFIRNKANGGTT
NO: 49 of US EYAASVKGRFTISRDDSKNTLYLOMNSLRAE
2013/0035249) DTAVYYCARDRGLRFYFDYWGOGTTVTVS
S
Second Binding DVQLVQSGAEVKKPGASVKVSCKASGYTFT 23 Domain (SEQ ID RYTMHWVRQAPGQGLEWIGYINPSRGYTNY
NO: 50 of US ADSVKGRFTITTDKSTSTAYMELSSLRSEDTA
2013/0035249) TYYCARYYDDHYCLDYWGQGTTVTVSSGEG
TSTGSGGSGGSGGADDIVLTQSPATLSLSPGE
RATLSCRASQSVSYMNWYQQKPGKAPKRWI
YDTS KVAS G VP ARFS GS GS GTD YS LTINS LEA
EDAATYYCQQWSSNPLTFGGGTKVEIK
Second Binding E VQLVES GGGL VQPGRS LRLS C A AS GFT VS S 24 Domain (SEQ ID YWMHWVRQAPGKGLEWVGFILNKANGGTT
NO: 51 of US EYAASVKGRFTISRDDSKNTLYLQMNSLRAE
2013/0035249) DTAVYYCARDRGLRFYFDYWGQGTTVTVS
S
Second Binding DIQLTQSPASLAVSLGQRATISCKASQSVDYD 25 Domain (amino GDSYLNWYQQIPGQPPKLLIYDASNLVSGIPP acids 28-277 of RFS GS GS GTDFTLNIHP VEKVD A AT YHCQQS
SEQ ID NO: 5 TEDPWTFGGGTKLEIKGGGGS GGGGSGGGGS from Q VQLQQS GAELVRPGS S VKIS CKAS G YAFS S
WO2009/070642). YWMNWVKQRPGQGLEWIGQIWPGDGDTNY
NGKFKGKATLTADESSSTAYMQLSSLASEDS AVYFCARRETTTVGRYYYAMDYWGQGTTV TVSS
Bispecific QAVLTQPASLSASPGASASLTCTLRRGINVGA 26
Antibody YSIYWYQQKPGSPPQYLLRYKSDSDKQQGSG
Sequence VSSRFSASKDASANAGILLISGLQSEDEADYY
CMIWHSGASAVFGGGTKLTVLGGGGSGGGG
SGGGGSEVQLVESGGGLVQPGRSLRLSCAAS
GFTVSSYWMHWVRQAPGKGLEWVGFIRNK ANGGTTEYAASVKGRFTISRDDSKNTLYLQM
NSLRAEDTAVYYCARDRGLRFYFDYWGQGT
TVTVS SS GGGGSD VQLVQSGAEVKKPGAS V
KVS CKAS G YTFTRYTMHW VRQAPGQGLEWI
GYINPSRGYTNYADSVKGRFTITTDKSTSTAY
MELS S LRSEDT AT Y YC ARY YDDH YCLD YWG
QGTTVTVSSGEGTSTGSGGSGGSGGADDIVL
TQSPATLSLSPGERATLSCRASQSVSYMNWY
QQKPGKAPKRWI YDTS KVAS G VP ARFS GS GS
GTDYSLTINSLEAEDAATYYCQQWSSNPLTF
GGGTKVEIK
Bispecific DIQLTQSPASLAVSLGQRATISCKASQSVDYD 27
Antibody GDSYLNWYQQIPGQPPKLLIYDASNLVSGIPP
Sequence RFS GS GS GTDFTLNIHP VEKVD A AT YHCQQS
TEDPWTFGGGTKLEIKGGGGS GGGGSGGGGS
Q VQLQQS GAELVRPGS S VKIS CKAS G YAFS S
YWMNWVKQRPGQGLEWIGQIWPGDGDTNY
NGKFKGKATLTADESSSTAYMQLSSLASEDS
AVYFCARRETTTVGRYYYAMDYWGQGTTV
TVSSGGGGSDIKLQQSGAELARPGASVKMSC
KTSGYTFTRYTMHWVKQRPGQGLEWIGYIN
PSRGYTNYNQKFKDKATLTTDKSSSTAYMQL
SSLTSEDSAVYYCARYYDDHYCLDYWGQGT
TLTVSSVEGGSGGSGGSGGSGGVDDIQLTQSP
AIMSASPGEKVTMTCRASSSVSYMNWYQQK
S GTSPKRWIYDTS KVAS G VP YRFS GS GS GTS Y
SLTISSMEAEDAATYYCQQWSSNPLTFGAGT
KLELK
Second Binding DIQLTQSPASLAVSLGQRATISCKASQSVDYD 28 Domain (SEQ ID GDSYLNWYQQIPGQPPKLLIYDASNLVSGIPP
NO:l from RFS GS GS GTDFTLNIHP VEKVD A AT YHCQQS
WO2009/070642). TEDPWTFGGGTKLEIK
Second Binding Q VQLQQS GAELVRPGS S VKIS CKAS G Y AFS S 29 Domain (SEQ ID YWMNWVKQRPGQGLEWIGQIWPGDGDTNY
NO:2 from NGKFKGKATLTADESSSTAYMQLSSLASEDS
WO2009/070642). AVYFCARRETTTVGRYYYAMDYWGQGTTV
TVSS
First Binding DIKLQQSGAELARPGASVKMSCKTSGYTFTR 30 Domain (SEQ ID YTMHWVKQRPGQGLEWIGYINPSRGYTNY
NO: 3 from NQKFKDKATLTTDKSSSTAYMQLSSLTSEDS
WO2009/070642). A V Y YC ARY YDDH YCLD YWGQGTTLT VS S
First Binding DIQLTQSPAIMSASPGEKVTMTCRASSSVSY 31 Domain (SEQ ID MNWYQQKS GTS PKRWI YDTS KV ASG VP YR
NO: 4 from FSGSGSGTSYSLTISSMEAEDAATYYCQQWS
WO2009/070642). SNPLTFGAGTKLELK Methods of Treatment/Administration
[00113] Also provided herein are methods of treating a subject by administering a therapeutically effective amount of the pharmaceutical formulations to the subject. Suitably, the pharmaceutical formulations are administered to a subject parenterally, i.e. no involving the gastrointestinal tract, and are suitably administered subcutaneously (i.e, directly below the dermis and epidermis of the skin) or by injection. Subcutaneous administration can include various pumps or other methods of administration below the skin surface.
[00114] As used herein, the term "subject" includes any human or nonhuman animal. The term "nonhuman animal" includes all vertebrates, for example, but not limited to, mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
[00115] By the terms "treat," "treating" or "treatment of (or grammatically equivalent terms) it is meant that the severity of the subject's condition is reduced or at least partially improved or ameliorated and/or that some alleviation, mitigation or decrease in at least one clinical symptom is achieved and/or there is an inhibition or delay in the progression of the condition and/or prevention or delay of the onset of a disease or illness. Thus, the terms "treat," "treating" or "treatment of refer to both prophylactic and therapeutic treatment regimes. A "therapeutically effective" amount as used herein is an amount that provides some improvement or benefit to the subject. Stated in another way, a "therapeutically effective" amount is an amount that provides some alleviation, mitigation, and/or decrease in at least one clinical symptom. Clinical symptoms associated with the disorders that can be treated by the methods of the invention are well- known to those skilled in the art. Further, those skilled in the art will appreciate that the therapeutic effects need not be complete or curative, as long as some benefit is provided to the subject. Appropriate dosages and timing of such dosages for administration of the pharmaceutical formulations can be readily determined by those of ordinary skill in the art, and are described for example in U.S. 2013/0035249 {see e.g., paragraphs [0236- 0245]), which is incorporated by reference herein in its entirety. Methods for Buffer or Excipient Selection
[00116] In further embodiments, methods for selecting a buffer or an excipient for use in a protein formulation are provided. Such methods can also be utilized for selecting a buffer or excipient suitable to reduce or prevent protein aggregation in a protein formulation.
[00117] The terms "polypeptide," "peptide," "protein," and "protein fragment" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. A protein formulation refers to a composition comprising one or more proteins (may be the same or different) and one or more appropriate buffers and/or excipients. As used herein, "polypeptide," "peptide," and "protein" refer to amino acid polymers including antibodies or antigen-binding fragments thereof.
[00118] In embodiments, the methods comprise providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient. Suitably, the concentration of the protein is the same in each buffer solution, and the concentration of the buffer or the excipient in each buffer solution is the same, but the buffer or the excipient in each buffer solution is different, so as to provide a method for investigation of the effect of the type of buffer or excipient on the protein formulation.
[00119] The methods comprise measuring a light scattering intensity of each of the plurality of buffer solutions. Light scattering intensity can be measured using various methods and instrumentations as described herein or otherwise known in the art.
[00120] From the light scattering intensity, a Debye plot is constructed. Relative scattering intensities of the buffer solutions in excess of a background (solvent) are converted into absolute scattering intensities and plotted as a function of concentration to obtain B22 and weight average molecular weight from the Debye working equation:
KC/R90=1/M+ 2B22C
[00121] Where 'K' is an optical constant, R90 is Rayleigh ratio, M (kDa) is the weight average molecular weight, C (mg/mL) is protein concentration and B22 (mol»mL/g2) is the second virial coefficient [00122] From the Debye plot, the second virial coefficient (B22) of each of the plurality of buffer solutions is determined. The second virial coefficients of each of the plurality of buffer solutions are then compared, for example graphically. As a result of the comparison, a more positive second virial coefficient (i.e., less negative) indicates a buffer or excipient for use in the protein formulation.
[00123] The more positive the second virial coefficient in comparison to another buffer or excipient, the more desirable the buffer or excipient is for use in a protein formulation. As described herein, selection of buffers and/or excipients with more positive second virial coefficients is an indication of the suitability of the buffer or excipient for use in protein formulations, by providing a more stable protein formulation with reduced aggregation and/or fragmentation of protein.
[00124] In further embodiments, methods are provided for selecting a concentration of a buffer or an excipient for use in a protein formulation. Methods are also provided for selecting a buffer concentration or excipient concentration suitable to reduce or prevent protein aggregation in a protein formulation.
[00125] The methods suitably comprise providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient. In embodiments, the concentration of the protein is the same in each buffer solution, and the concentration of the buffer or the excipient in each buffer solution is different, but the buffer or the excipient in each buffer solution is the same, so as to provide a method for investigation of the effect of the buffer concentration or excipient concentration on the protein formulation.
[00126] The light scattering intensity of each of the plurality of buffer solutions is measured, and a Debye plot is constructed. After determining the second virial coefficient of each of the plurality of buffer solutions, the second virial coefficients of each of the plurality of buffer solutions are compared. A more positive second virial coefficient indicates the concentration of the buffer or the excipient for use in the protein formulation. As described herein, selection of a concentration of buffers and/or excipients with more positive second virial coefficients is an indication of the suitability of the concentration of buffer or excipeint for use in protein formulations, by providing a more stable protein formulation with reduced aggregation and/or fragmentation of protein. [00127] The methods provided herein are suitably used with any protein formulation, including formulations where the protein is an antibody or antigen-binding fragment thereof, a peptide or a fusion protein. In exemplary embodiments, the antibody investigated in the methods described herein is a bispecific antibody comprising a first and a second binding domain, suitably where the first binding domain specifically binds the CD3 T cell surface antigen.
[00128] Any desired buffer or excipient, as well as concentration of buffer or excipient, can be selected for the investigation. In embodiments, the buffer includes, but is not limited to, citrate, succinate, phosphate, histidine, acetate, TRIS and combinations thereof. Suitably, the concentration of the buffer or the excipient is from about 5 mM to about 200 mM, suitably less than about 100 mM. Examples of excipients include, but are not limited to, proteins (for example, but not limited to, serum albumin), amino acids (for example, but not limited to, aspartic acid, glutamic acid, lysine, arginine, glycine), surfactants (for example, but not limited to, SDS, Tween 20, Tween 80, polysorbate, polysorbate 80 and nonionic surfactants), sugars and saccharides (for example, but not limited to, glucose, sucrose, maltose and trehalose), polyols (for example, but not limited to, mannitol and sorbitol), fatty acids and phospholipids (for example, but not limited to, alkyl sulfonates and caprylate).
[00129] In embodiments, the methods for selection of a buffer or excipient or concentration of a buffer or excipient are carried out with bispecific antibody formulations, including formulations which include the various bispecific antibodies and binding domains described throughout.
[00130] The methods described herein for determining and comparing the second virial coefficient to select an appropriate buffer and/or excipient have been found to reduce the time required to otherwise select appropriate buffer systems from 2-3 weeks to 1 week or less. This significant reduction leads to savings in costs, man-power and materials.
[00131] It will be readily apparent to one of ordinary skill in the relevant arts that other suitable modifications and adaptations to the methods and applications described herein can be made without departing from the scope of any of the embodiments. The following examples are included herewith for purposes of illustration only and are not intended to be limiting. Examples
EXAMPLE 1 - Materials & Methods
The following materials and methods were utilized in Examples 1-8.
Materials:
[00132] MEDI-565/BiTE® comprising SEQ ID NO: 26 was prepared according to standard methods. Other bispecific antibodies are readily prepared by those in the field using well-known methods.
[00133] Buffer components, including sodium citrate, citric acid, sodium phosphate, sodium succinate, L-histidine and L-histidine hydrochloride were purchased from
J.T.Baker. (Center Valley, PA).
[00134] Lyoprotectant, α,α-trehalose dihydrate and surfactant, polysorbate 80, were purchased from Ferro Pfanstiehl (Waukegan, IL) and J.T.Baker (Center Valley, PA) respectively.
Measurement of Second Virial Coefficient
[00135] Second virial coefficient (B22) measurements were performed using a DAWN® HELEOS® Multi-Angle Light Scattering (MALS) detector coupled to OPTILAB® REX™ refractive index detector (Wyatt Technologies, Santa Barbara, CA) to obtain virial coefficients (B22) and apparent molecular weight. Protein and buffer solutions were filtered through 0.22 μιη filters and delivered into the light scattering cell and refractive index detector using a CALYPSO® dispensing system, (Wyatt Technologies, Santa Barbara, CA). Relative scattering intensities in excess of the background (solvent) are converted into absolute scattering intensities and plotted as a function of concentration to obtain B22 and weight average molecular weight from the Debye working equation:
KC/R90=1/M+ 2B22C
Where 'K' is an optical constant, R90 is Rayleigh ratio, M (kDa) is the weight average molecular weight, C (mg/mL) is protein concentration and B22 (mol- mL/g2) is the second virial coefficient.
Differential Scanning Calorimetry
[00136] The conformational stability of bispecific antibodies as described herein in the presence of different excipients and excipient concentrations were determined by VP- DSC ultrasensitive differential scanning calorimeter (Microcal, Northampton, MA) using a 96 well plate at a protein concentration of ~1 mg/mL. Samples were heated from 20- 90°C at a rate of 90°C per hour. Normalized heat capacity (Cp) data were corrected for buffer baseline. The onset of melting transition (Tml) was used to determine bispecific antibody conformational stability in different formulations.
Stability Studies
[00137] Bispecific antibody liquid formulations were subjected to 5°C, 25°C/60 relative humidity (RH) and 30°C. The aggregation of bispecific antibody increased at elevated temperature, thus 25°C/60 RH and 30°C were selected as accelerated temperature conditions to evaluate the impact of excipients and excipient concentrations on antibody aggregation. To evaluate stability of frozen formulations, samples were incubated at -80°C and/or -40°C.
[00138] Pharmaceutical formulations were investigated comprising different buffers at varying concentrations (generally 10 mM to 100 mM), with different lyoprotectants at a concentration range of about 50 mM to 150 mM, and polysorbate 80 (0.002 to 0.05 )
[00139] Lyophilized bispecific antibody formulations were subjected to 5°C, 25°C/60 RH and 40°C/75 RH to evaluate stability.
[00140] Both liquid formulations and reconstituted lyophilized formulations were tested for aggregation by high performance size exclusion chromatography (HP-SEC), sub- visible particles by MFI and activity by bioassay.
Freeze Drying:
[00141] Freeze drying was performed using a laboratory scale VirTis freeze dryer.
Formulations were freeze dried in Schott (Mullheim, Germany) 6R or 10R Fiolax clear glass vials using the following protocol:
Freezing - 1° C/min to -40° C for 120min
• Annealing - Γ C/min to -16° C for 150min
Freezing - 1° C/min to -40° C for 120min
Primary Drying - 0.3° C/min to -30° C at >100 mTorr
Secondary Drying - 0.3° C/min to 20° C at >100 mTorr [00142] The chamber pressure was set to >100mTorr. The end point of primary and secondary drying was determined based on a pirani pressure gauge. Thermocouples were placed in selected vials to measure the product temperature.
High Performance Size Exclusion Chromatography (HP-SEC)
[00143] HP-SEC samples were analyzed using Agilent (Santa Clara, CA) 1100 HP-SEC, equipped with a YMC® DIOL (5μιη, 8x300mm; 200A) size exclusion column with an FLD detector (FLD method) or Agilent 1200 series HPLC TSK-Gel G3000SWXL (5μιη, 7.8x300mm; 250A) size exclusion column with a UV-vis detector (DAD method).
[00144] Samples analyzed using DAD method were run undiluted to achieve a target column load of 170-250 μg whereas samples analyzed using the FLD method were diluted (with phosphate buffered saline) to achieve a target load of 0.5 μg. For both methods, samples were incubated in the auto-sampler (5°C) for approximately 1 hour prior to analysis. The chilled samples were injected onto respective columns and eluted isocratically with 0.1 M disodium phosphate containing 0.1 M sodium sulfate and 0.05% sodium azide, pH 6.8, at a flow rate of 1.0 ml/minute (DAD method) or 0.2 M disodium phosphate containing 0.2 M sodium sulfate, pH 7, at a flow rate of 0.75 ml/minute (FLD method). The eluted protein was detected using UV absorbance at 280 nm (DAD method) and fluorescence detection with excitation at 280nm and emission at 345nm (FLD method). The results are reported as the area percent of the product monomer peak. Peaks eluting earlier than the monomer are recorded as percent aggregate and peaks eluting after the monomer are recorded as percent fragment/other.
Post Re constitution Stability Study:
[00145] The stability of lyophilized formulations was determined post reconstitution by adding 1.4 mL water for injection (WFI) to achieve target protein concentration of ~ 5mg/mL. The post reconstituted formulation was subjected to 5° C and room temperature for 24 hours and 4 hours respectively. Aggregation and sub-visible particles were evaluated during an in use stability study. Sub-visible Particle Analysis:
[00146] Sub-visible particle analysis was performed using a Micro-flow Imager
(proteinsimple, Santa Clara, CA). 0.8mL of an undiluted sample was analyzed using 100 μιη, 1.6 mm flow cell. Deionized water was used for background measurements and aspect ratio filter was used to eliminate any spherical particles with aspect ratio >0.85.
T-Cell Mediated Apoptosis Bioassay:
[00147] A T-Cell mediated apoptosis bioassay was used to measure the ability of a test sample to induce apoptotic cell death of a CEA-expressing human colon tumor cell line (LS174T). Once one domain of a bispecific antibody binds CEA on the target cells and the other domain binds to CD3 on effector T-cells (MCI 5), the transient ligation of the effector cell and the target cell via the bispecific antibody activates the effector cells to release cytotoxic granules (Granzyme B), which enter the target cells through Perforin- mediated tunnels in the cell membrane. This process induces the LS174T cells to undergo apoptosis via Caspase 3/7 activation that can be measured by the Caspase-Glo™ 3/7 Assay System. The amount of luminescence proportional to the amount of apoptosis was quantified in a luminometer after reaction with a Caspase 3/7 luciferase substrate. LS174T and MC15 cells were plated in the cell growth media (10% fetal bovine serum (FBS) in RPMI) in a 96-well plate. A series of sample dilutions were added to the plates. After incubation for 23 hours, the Caspase 3/7 reagent was added to the plate and the luminescence was measured on a plate reader. The data was analyzed in Softmax Pro using a four-parameter logistical curve fit to determine the EC50 values of the samples. The potency of the test sample was determined by dividing the EC50 of Reference Standard by the EC50 of the test sample.
EXAMPLE 2: EFFECT OF BUFFER ON STABILITY
[00148] To investigate the effect of different buffers on the stability of a single chain bispecific antibody at a target concentration of about 5 mg/mL, stability studies at 30° C, for 0-14 days were performed on a pharmaceutical formulation comprising 5 mg/mL of MEDI-565 (SEQ ID NO: 26). FIG. 2 shows the effect of buffer on stability of the bispecific antibody MEDI-565, examining aggregation percent for the antibody at 5 mg/mL, at 30° C for 15 days as measured by HPSEC, using the methods described in Example 1. The study examined four buffers: histidine (circle), succinate (square), citrate (triangle) and phosphate (cross), each at 30 mM. Citrate buffer demonstrated the lowest amount of aggregation over the time-course of the study.
EXAMPLE 3: MEASUREMENT OF SECOND VIRIAL COEFFICIENT
[00149] One of the main drivers for aggregation of protein molecules, including single chain bispecific antibodies, is from protein-protein interactions (PPI). PPI's are often dictated by colloidal (weak PPI) and conformational stability of a given molecule. The second virial coefficient (B22) is a thermodynamic parameter known to reflect PPI in dilute solution conditions and can be utilized to determine the colloidal stability. To further investigate the role of citrate, succinate, phosphate and histidine to control aggregation rates, the second virial coefficient (B22) was determined for samples comprising 30 mM buffer, 5 mg/mL of MEDI-565 (SEQ ID NO: 26) at pH 6.0. As shown in Figure 3, a strong correlation between the observed aggregation rates per day at 30°C and the colloidal stability (B22) for the single chain bispecific antibody was observed. From the data in Figure 3 we also concluded that intermolecular interactions are also affected in the order of Hofmeister series with citrate being the most stabilizing excipient (lowest aggregation rate and weakly attractive PPI) and histidine being the lowest stabilizing excipient (high aggregation rate and strong attractive PPI). Conformational stability was also studied using Differential Scanning Calorimetry (DSC) and Differential Scanning Fluorometry (DSF), however differences in these stabilities were noticeable only at higher salt concentration. Historical studies have indicated that the Hofmeister series usually impacts protein stability at higher salt concentrations, however, this work also shows significant stabilization by these ions at lower salt concentration (<0.1M). For single chain antibodies, optimization of colloidal stability through formulation development was crucial for developing a stable formulation.
[00150] FIG. 3 further demonstrates that the single chain bispecific antibody in citrate showed a surprisingly low amount of aggregation as compared to the other buffers. Also shown on the second Y axis is the second virial coefficient for the formulations. As described, the most positive, i.e., least negative, value for B22 corresponded to citrate, which also demonstrated the lowest aggregation rate, evidence of the weakly attractive protein-protein interaction in citrate compared to other buffers. EXAMPLE 4: IMPACT OF CITRATE CONCENTRATION ON STABILITY
[00151] To identify the optimal citrate concentration, the effect of citrate concentration over the range 10 mM to 100 mM on the stability and second viral coefficient of a 5 mg/mL formulation of MEDI-565 at pH 6.0 was examined. As shown in FIG. 4A with increasing citrate concentration, the aggregation rate at 30°C of the bispecific antibody was reduced, reaching a relative plateau after approximately 30 mM, and an observed significant drop in aggregation above 20 mM citrate. Thus, stability of the single chain antibody formulation was highly dependent on citrate concentration citrate with an increasing concentration of greater than about 20mM or greater than about 30 mM significantly improving antibody stability.
[00152] The lowest aggregation rate also corresponded to the formulation having the most positive second virial coefficient. See FIG. 4B. Increasing the citrate ion concentration lowered the aggregation rate demonstrating improved colloidal stability, with higher citrate concentrations showing a net protein-protein interactions change from attractive to repulsive, likely explaining the increased colloidal stability also observed at high ion concentrations. No significant changes to conformation stability by DSF & DSC were observed as a function of citrate concentration. For concentrations above approximately 60-100 mM citrate, subject discomfort associated at the injection site of a pharmaceutical formulation is often considered a limiting factor for using higher citrate concentrations. Thus, these experiments support selecting a target citrate concentration of about 60 mM for a single chain bispecific antibody formulation comprising about 5.0 mg/mL of the bispecific antibody to maximize antibody stability while minimizing potential injection site discomfort.
EXAMPLE 5: IMPACT OF PROTEIN CONCENTRATION
[00153] To investigate the effect of protein concentration on the stability of the pharmaceutical formulations, the following were prepared and the aggregation at 25° C was observed over the time course of 12 days by HPSEC.
Formulations: 1.7 mg/mL MEDI-565, 30 mM citrate, 70 mM trehalose, 0.01 % polysorbate-80, pH 6.0; 2.5 mg/mL MEDI-565, 30 mM citrate, 80 mM trehalose, 0.01 % polysorbate-80, pH 6.0; and
5.0 mg/mL MEDI-565, 60 mM citrate, 150 mM trehalose, 0.02 % polysorbate-80, pH 6.0.
[00154] As shown in FIG. 5, at a concentration of 1.7 mg/mL (circle) and 2.5 mg/mL (square), the addition of 30 mM citrate provided significant stability to the formulations, demonstrating about 2% aggregation increase over the time course of the study when held at 25° C. Increasing the concentration of the protein to 5 mg/mL (triangle) showed an increase in the aggregation and thus lower stability, reaching above 5% aggregation after about 10 days. Therefore, as described in Example 6, it was determined that in order to provide a formulation capable of being delivered to a patient at 5 mg/mL which is stable when held at 25 °C in preparation for and during administration, a lower concentration of protein was necessary during preparation of the liquid pharmaceutical formulations prior to lyophilization, and subsequent reconstitution in a volume that provides the final 5 mg/mL of protein for subject administration.
EXAMPLE 6: DRUG SUBSTANCE AND DRUG PRODUCT STABILITY
[00155] As demonstrated in Examples 2-5, stability of the single chain bispecific antibody was dependent on protein concentration and citrate concentration. To mitigate the effect of protein concentration, two possible options were investigated: (1) 2.5 mg/ml single chain antibody formulation + 1/2 reconstitution volume to yield a 5 mg/ml post reconstitution antibody concentration or (2) 1.7 mg/ml single chain antibody formulation + ½ reconstitution volume to yield a 5 mg/ml post reconstitution antibody concentration. Three suitable pharmaceutical formulations of MEDI-565 were further examined as described in Table 2. It should be noted that all three formulations are exemplary candidates for clinical use. In addition, other bispecific antibodies are also expected to demonstrate improved stability properties when formulated as described herein. The applicability of the formulations provided below to other bispecific antibodies, as well as optimization of such formulations, is readily performed by those working in the field, and can be accelerated using the methods described herein that rely on measurement and comparison of second virial coefficients. TABLE 2
Figure imgf000046_0001
[00156] For each formulation, 0.01% polysorbate 80 was also included, and the formulation pH was set at 6.0. The amount of polysorbate over the range of 0.002% to 0.05% was found to give similar results as far as reduced aggregation upon shaking. DS osmolality refers to the osmolality of the drug substance (DS), or liquid formulation, prior to lyophilization. DP osmolality refers to the osmolality of the lyophilized drug product (DP) following reconstitution in a volume of 1.4 mL (WFI).
[00157] Liquid formulations 1-3 were examined for stability over the course of 12 days at 25° C by measuring % aggregation by HSPEC.
[00158] As shown in FIG. 6, all three formulation remain stable at 25°C over a 12 day period, with formulations 2 (squares) and 3 (triangle) providing improved stability over formulation 1 (circles). Formulation 1 provided significant stability to the protein, particularly up to 10 days of storage.
[00159] Based on the results of FIG. 6, no significant differences were observed in formulation 2 (squares) and formulation 3 (triangles) thus in order to achieve lower post reconstitution citrate concentration (about 60 mM for formulation 2 compared to about 90 mM for formulation 3) while maintaining post reconstitution stability, formulation 2 was selected as the lead formulation.
[00160] FIG. 7 A shows the stability (% Aggregate at 5° C as measured by HPSEC) of 3 bispecific antibody (MEDI-565) lyophilized formulations at 5° C. The study examined the three formulations described in Table 2. As demonstrated, all 3 formulations were stable when stored at 5° C.
[00161] FIG. 7B shows the stability (% Aggregate at 40° C as measured by HPSEC) of 3 bispecific antibody (MEDI-565) lyophilized formulations at an accelerated temperature. The study examined the three formulations described in Table 2. As demonstrated, no significant increase in aggregation was observed for all 3 formulations at 40° C. [00162] All three formulations (1, 2 and 3 described in Table 2) were lyophilization suitable with the trehalose content optimized to improve cake appearance (about 60- 80mM) as described in Example 1. All three formulations had the following characteristics: (1) Reconstitution time < 5min; (2) Moisture content <1.3%, and (3) an acceptable cake quality and acceptable shrinkage observed following a visual inspection. In addition, all three formulations were stable at 5°C, 25 °C and 40°C with no significant increase in aggregation observed in accelerated stability studies performed at 40°C.
[00163] As described in FIG. 9A and 9B, 3 lyophilized MEDI-565 pharmaceutical formulations (as described in Table 2) were reconstituted (as follows: lyophilized Formulation 1 was reconstituted 1/2 volume for a final liquid formulation = 5mg/ml MEDI-565, 60mM Citrate, 0.16M Trehalose, 0.02% PS-80, pH 6; lyophilized Formulation 2 was reconstituted 1/3 volume for a final liquid formulation = 5mg/ml MEDI-565, 59mM Citrate, 0.21M Trehalose, 0.03% PS-80, pH 6; and lyophilized Formulation 3 was reconstituted 1/3 volume for a final liquid formulation = 5mg/ml MEDI-565, 89mM Citrate, 0.18M Trehalose, 0.03% PS-80, pH 6), and percent aggregation was determined at 4 hours (at room temperature) and 24 hours (at 5° C). Virtually no increase in aggregation was observed further evidencing the suitability of the three reconstituted formulations.
EXAMPLE 7: EFFECT OF SUCCINATE BUFFER ON STABILITY &
MEASUREMENT OF SECOND VIRIAL COEFFICIENT
[00164] A study was performed to identify the impact of succinate concentration over the range 10 mM to 100 mM on the stability and second viral coefficient of a 5 mg/mL formulation of MEDI-565 at pH 6.0. Additional excipients can be readily examined using these methods as well (see, e.g., Examples 2-3). As shown in FIG. 8A with increasing succinate concentration, the aggregation rate at 30°C of the bispecific antibody was reduced. Thus, similar to our results with citrate (see Example 2), stability of the single chain antibody formulation was highly dependent on succinate concentration.
[00165] Similar to our results with citrate (see Example 3), the lowest aggregation rate also corresponded to the formulation having the most positive second virial coefficient. See FIG. 8B. Increasing the succinate ion concentration lowered the aggregation rate demonstrating improved colloidal stability, with higher succinate concentrations showing a net protein-protein interactions change from strong attractive to weak attractive, likely explaining the increased colloidal stability also observed at high ion concentrations.
EXAMPLE 8: FROZEN DRUG SUBSTANCE STABILITY STUDIES
[00166] The liquid formulations described in Example 6 / Table 2 were also studied for long term storage stability at -80° C. FIG. 10 demonstrates percent aggregation of liquid MEDI-565 pharmaceutical formulations comprising the amounts of bispecific antibody, citrate and trehalose, following storage at -80° C for 3 months for formulations 1 and 3 and 27 months for formulation 2 (and then thawing and measurement). In all cases, aggregation % did not change significantly and was maintained below 0.5%, and virtually no sub-particles larger than 2 μιη were observed. Stability was demonstrated for periods of up to 2 years after storage at -80° C. In addition, virtually no particles larger than 2 μιη for the pharmaceutical formulations after three cycles of freeze-thaw (-80° C and room temperature) were observed.
EXAMPLE 9: EFFECT OF pH ON ANTIBODY AGGREGATION
[00167] FIG. 11 shows the results of an investigation of pH on aggregation for a MEDI- 565 formulation (1.7 mg/mL, 20 mM citrate, 70 mM trehalose, 0.01% polysorbate-80). No significant effects on aggregation were noted, which suggests that MEDI-565 can be formulation at a pH range of 6 + 1 until while maintaining stability.
[00168] In summary, the Examples demonstrate that a 1.7mg/ml single chain bispecific antibody, 20mM Sodium Citrate/Citric Acid, 70mM Trehalose Dihydrate, 0.01% PS80, pH 6.0 pharmaceutical formulation and a 5mg/ml single chain bispecific antibody, 59mM Sodium Citrate/Citric Acid, 0.21M Trehalose Dihydrate, 0.03% PS80, pH 6.0 (having an osmolality of 436mOsm/kg) pharmaceutical formulation are each suitable for parenteral administration of the single chain bispecific antibody, including subcutaneous administration or injection. These formulations provided good stability at 5° C, 25° C and -80° C with combined aggregation < 2.0%. EXAMPLE 10: INVITRO ACTIVITY (BIOASSAY) OF DRUG SUBSTANCE AND
DRUG PRODUCT
[00169] FIG. 12 shows the results of in vitro activity assay (bioassay) of frozen drug substance (circle), lyophilized drug product stored at 2-8°C (square) and lyophilized drug product stored at 40°C (triangle) relative to a reference standard.
[00170] The drug substance shown in FIG. 12 was composed of 1.7 mg/mL MEDI-565, 20 mM citrate, 70 mM trehalose, 0.01% polysorbate-80, pH 6 and the corresponding post reconstitution drug product contained 5mg/mL MEDI-565, 59 mM citrate, 0.21 M trehalose, 0.03% polysorbate 80, pH 6.0.
[00171] As shown in FIG. 12 there was no significant change in % relative activity of the drug substance when stored frozen (-80°C) and the drug product when stored at 2-8°C for 27 months. Similarly no significant changes were observed for the drug product stored at 40°C for 3 months, which suggests that both frozen drug substance and the drug product retain their activities for an extended period.
***
[00172] All documents, patents, journal articles and other materials cited in the present application are hereby incorporated by reference.
[00173] Although the present invention has been fully described in conjunction with several embodiments thereof with reference to the accompanying drawings, it is to be understood that various changes and modifications can be apparent to those skilled in the art. Such changes and modifications are to be understood as included within the scope of the present invention as defined by the appended claims, unless they depart there from.

Claims

WHAT IS CLAIMED IS:
1. A pharmaceutical formulation comprising: a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises:
a) about 1.0 mg/mL to about 5.0 mg/mL of the bispecific antibody;
b) about 15 mM to about 60 mM citrate;
c) about 25 mM to about 0.21 M of a lyoprotectant; and
d) about 0.002% to about 0.1% of a surfactant,
wherein the formulation has a pH of about 5.0 to about 7.0.
2. The pharmaceutical formulation of claim 1, comprising about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody.
3. The pharmaceutical formulation of claim 1 or claim 2, comprising about 1.7 mg/mL or about 2.5 mg/mL of the bispecific antibody.
4. The pharmaceutical formulation of any one of claims 1-3, comprising about 20 mM to about 30 mM citrate.
5. The pharmaceutical formulation of any one of claims 1-4, comprising about 20 mM citrate or about 30 mM citrate.
6. The pharmaceutical formulation of any one of claims 1-5, comprising about 60 mM to about 80 mM of the lyoprotectant.
7. The pharmaceutical formulation of any one of claims 1-6, comprising about 70 mM of the lyoprotectant.
8. The pharmaceutical formulation of any one of claims 1-7, comprising about 0.008% to about 0.012% of the surfactant.
9. The pharmaceutical formulation of any one of claims 1-8, comprising about 0.01% of the surfactant.
10. The pharmaceutical formulation of any one of claims 1-9, wherein the lyoprotectant is selected from the group consisting of trehalose, lactose, mannitol, raffinose and sucrose.
11. The pharmaceutical formulation of any one of claims 1-10, wherein the surfactant is selected from the group consisting of polysorbate-20, polysorbate-40, polysorbate-60 and polysorbate-80.
12. A pharmaceutical formulation comprising: a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises:
a) about 1.7 mg/mL to about 2.5 mg/mL of the bispecific antibody;
b) about 20 mM to about 30 mM citrate;
c) about 60 mM to about 80 mM trehalose; and
d) about 0.002% to about 0.1 % polysorbate 80,
wherein the formulation has a pH of about 5.0 to about 7.0.
13. A pharmaceutical formulation comprising: a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises:
a) about 1.7 mg/mL of the bispecific antibody;
b) about 20 mM citrate;
c) about 70 mM trehalose; and
d) about 0.01 % polysorbate 80,
wherein the formulation has a pH of about 6.0.
14. A pharmaceutical formulation comprising: a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises:
a) about 5.0 mg/mL of the bispecific antibody;
b) about 60 mM citrate;
c) about 0.21 M trehalose; and
d) about 0.03% polysorbate 80,
wherein the formulation has a pH of about 6.0.
15. The pharmaceutical formulation of claim 14, having a volume of about 0.5 mL to about 12.0 mL.
16. The pharmaceutical formulation of claim 14 or claim 15 having a volume of about 1.4 mL.
17. The pharmaceutical formulation of any one of claims 14-16, wherein the formulation is administered to a subject parenterally.
18. The pharmaceutical formulation of claim 17, wherein the formulation is administered to a subject subcutaneously or by injection.
19. The pharmaceutical formulation of any one of claims 1-16, wherein the formulation is a liquid formulation, a frozen formulation, a lyophilized formulation or a reconstituted formulation.
20. A lyophilized pharmaceutical formulation comprising: a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises:
a) about 2.5 mg to about 61mg of the bispecific antibody;
b) about 8.5 mg to about 204mg of citrate;
c) about 40 mg to about 954mg trehalose; and
d) about 0.15 μg to about 3^g of polysorbate 80.
21. A lyophilized pharmaceutical formulation comprising: a bispecific antibody, wherein the bispecific antibody comprises a first and a second binding domain, wherein the first binding domain specifically binds the CD3 T cell surface antigen, and wherein the formulation comprises:
a) about 7 mg of the bispecific antibody;
b) about 25 mg citrate;
c) about 117 mg trehalose; and
d) about 0.4 μg polysorbate 80.
22. The lyophilized pharmaceutical formulation of claim 20 or claim 21, wherein following storage at 25°C for 6 months, or storage at 2-8°C for 24 months, the formulation exhibits < 2.0% aggregation when reconstituted.
23. A method of preparing a pharmaceutical formulation comprising:
a) preparing a liquid pharmaceutical formulation according to the pharmaceutical formulation of any one of claims 1-18;
b) lyophilizing the liquid pharmaceutical formulation of a) to produce a lyophilized formulation; and c) reconstituting the lyophilized formulation of b) in a volume suitable for parenteral administration.
24. The method of claim 23, further comprising freezing the liquid pharmaceutical formulation of a) prior to the lyophilizing in b).
25. The method of claim 24, wherein the freezing the liquid pharmaceutical formulation of a) comprises freezing to at least about -40° C or at least about -80° C.
26. The method of any one of claims 23-25, wherein the reconstituting in c) comprises reconstituting the lyophilized formulation of b) in about 1/2 to about 1/3 volume of the liquid pharmaceutical formulation of a).
27. The method of any one of claims 23-26, wherein the reconstituting in c) comprises reconstituting in a volume of about 0.5 mL to about 12.0 mL.
28. The method of any one of claims 23-27, wherein the reconstituting in c) comprises reconstituting in a volume of about 1.4 mL.
29. The method of any one of claims 23-28, wherein the reconstituting in c) comprises reconstituting in a volume suitable for subcutaneous administration or injection.
30. A pharmaceutical formulation prepared according to any of the methods of claims 23- 29.
31. The pharmaceutical formulation of any one of claims 1-19 and 30, or the lyophilized pharmaceutical formulation of any one of claims 20-21, wherein the first binding domain comprises SEQ ID NO: 1, SEQ ID NO: 30 or SEQ ID NO: 31.
32. The pharmaceutical formulation of any one of claims 1-19 and 30, or the lyophilized pharmaceutical formulation of any one of claims 20-21, wherein the second binding domain specifically binds an antigen selected from the group consisting of: CD 19, CD20, CD22, EphA2, EphA4, INFR, ICOS, Ep-CAM, CEA, and IL-5 receptor.
33. The pharmaceutical formulation of claim 32, wherein the second binding domain specifically binds CEA.
34. The pharmaceutical formulation of claim 33, wherein the second binding domain comprises the amino acid sequence of any one of SEQ ID NOs: 2-24 and 28-29.
35. The pharmaceutical formulation of claim 33, wherein the bispecific antibody comprises SEQ ID NO: 26.
36. The pharmaceutical formulation of claim 32, wherein the second binding domain specifically binds CD 19.
37. The pharmaceutical formulation of claim 36, wherein the second binding domain comprises SEQ ID NO: 25.
38. The pharmaceutical formulation of claim 36, wherein the bispecific antibody comprises SEQ ID NO: 27
39. A method for selecting a buffer or an excipient for use in a protein formulation or for selecting a buffer or excipient suitable to reduce or prevent protein aggregation in a protein formulation, the method comprising:
a) providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient,
i) wherein the concentration of the protein is the same in each buffer solution, and
ii) wherein the concentration of the buffer or the excipient in each buffer solution is the same, but the buffer or the excipient in each buffer solution is different;
b) measuring a light scattering intensity of each of the plurality of buffer solutions; c) constructing a Debye plot;
d) determining a second virial coefficient of each of the plurality of buffer solutions; and
e) comparing the second virial coefficient of each of the plurality of buffer solutions, wherein a more positive second virial coefficient indicates a buffer or excipient for use in the protein formulation.
40. A method for selecting a concentration of a buffer or an excipient for use in a protein formulation, or for selecting a buffer or excipient concentration suitable to reduce or prevent protein aggregation in a protein formulation, the method comprising:
a) providing a plurality of buffer solutions, each buffer solution consisting of a concentration of a protein and a concentration of a buffer or an excipient,
i) wherein the concentration of the protein is the same in each buffer solution, and
ii) wherein the concentration of the buffer or the excipient in each buffer solution is different, but the buffer or the excipient in each buffer solution is the same;
b) measuring a light scattering intensity of each of the plurality of buffer solutions; c) constructing a Debye plot;
d) determining a second virial coefficient of each of the plurality of buffer solutions; e) comparing the second virial coefficient of each of the plurality of buffer solutions, wherein a more positive second virial coefficient indicates the concentration of the buffer or the excipient for use in the protein formulation.
41. The method of claim 39 or claim 40, wherein the protein is an antibody or antigen- binding fragment thereof, a peptide or a fusion protein.
42. The method of claim 41, wherein the antibody is a bispecific antibody comprising a first and a second binding domain, and wherein the first binding domain specifically binds the CD3 T cell surface antigen.
43. The method of any one of claims 39-42, wherein the buffer is selected from the group consisting of citrate, succinate, phosphate, histidine, acetate, TRIS and combinations thereof.
44. The method of any one of claims 39-43, wherein the concentration of the buffer or the excipient is from about 5 mM to about 200 mM.
45. The method of any one of claims 39-43, wherein the concentration of the buffer or the excipient is less than about 100 mM.
46. The method of any one of claims 39-45, wherein the excipient is a sugar.
47. The method of claim 42, wherein first binding domain comprises SEQ ID NO: l, SEQ ID NO: 30 or SEQ ID NO: 31.
48. The method of claim 42, wherein the second binding domain specifically binds an antigen selected from the group consisting of: CD19, CD20, CD22, EphA2, EphA4, INFR, ICOS, Ep-CAM, CEA, and IL-5 receptor.
49. The method of claim 48, wherein the second binding domain specifically binds CEA.
50. The method of claim 49, wherein the second binding domain comprises the amino acid sequence of any one of SEQ ID NOs: 2-24 and 28-29.
51. The method of claim 49, wherein the bispecific antibody comprises SEQ ID NO: 26.
52. The method of claim 48, wherein the second binding domain specifically binds CD19.
53. The method of claim 52, wherein the second binding domain comprises SEQ ID NO:
25.
54. The method of claim 52, wherein the bispecific antibody comprises SEQ ID NO: 27.
PCT/US2015/047843 2014-09-02 2015-09-01 Formulations of bispecific antibodies WO2016036678A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462044544P 2014-09-02 2014-09-02
US62/044,544 2014-09-02

Publications (1)

Publication Number Publication Date
WO2016036678A1 true WO2016036678A1 (en) 2016-03-10

Family

ID=55440297

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/047843 WO2016036678A1 (en) 2014-09-02 2015-09-01 Formulations of bispecific antibodies

Country Status (1)

Country Link
WO (1) WO2016036678A1 (en)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9914776B2 (en) 2014-08-04 2018-03-13 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
CN108264559A (en) * 2016-12-30 2018-07-10 上海近岸生物科技有限公司 Three functional moleculars and its application of a kind of combination CD19, CD3 and the positive costimulatory molecules of T cell
WO2018141910A1 (en) * 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
US10087250B2 (en) 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
US10155815B2 (en) 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10174124B2 (en) 2013-12-17 2019-01-08 Genentech, Inc. Anti-CD3 antibodies and methods of use
WO2019020745A1 (en) * 2017-07-28 2019-01-31 F. Hoffmann-La Roche Ag Bispecific antibody formulation
US10208285B2 (en) 2016-10-07 2019-02-19 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10323094B2 (en) 2015-06-16 2019-06-18 Genentech, Inc. Humanized and affinity matured antibodies to FcRH5 and methods of use
US10358473B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10501545B2 (en) 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
WO2019234576A1 (en) 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Bispecific antibodies against ceacam5 and cd47
WO2020056008A1 (en) 2018-09-12 2020-03-19 Silverback Therapeutics, Inc. Compositions for the treatment of disease with immune stimulatory conjugates
WO2020053301A1 (en) * 2018-09-11 2020-03-19 Ichnos Sciences S.A. Compositions comprising a bispecific antibody, bufffer and one or more stabilizing agents
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
WO2020257407A1 (en) 2019-06-19 2020-12-24 Silverback Therapeutics, Inc. Anti-mesothelin antibodies and immunoconjugates thereof
US10882918B2 (en) 2016-09-30 2021-01-05 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
WO2021028669A1 (en) * 2019-08-09 2021-02-18 Arecor Limited Novel composition comprising antibodies
WO2021053587A1 (en) 2019-09-18 2021-03-25 Klaus Strein Bispecific antibodies against ceacam5 and cd3
WO2021035028A3 (en) * 2019-08-20 2021-04-08 Regeneron Pharmaceuticals, Inc. Formulation optimization for bispecific antibodies
CN112789028A (en) * 2018-10-01 2021-05-11 美国安进公司 Methods for reducing aggregation of bispecific antibodies
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
EP3831849A1 (en) 2019-12-02 2021-06-09 LamKap Bio beta AG Bispecific antibodies against ceacam5 and cd47
US11084877B2 (en) 2014-09-12 2021-08-10 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US11179473B2 (en) 2020-02-21 2021-11-23 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
US11242376B2 (en) 2016-08-02 2022-02-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11242390B2 (en) 2016-03-22 2022-02-08 Hoffmann-La Roche Inc. Protease-activated T cell bispecific molecules
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
CN114246944A (en) * 2020-09-24 2022-03-29 盛禾(中国)生物制药有限公司 Pharmaceutical composition of bispecific antibody and application thereof
EP3878461A4 (en) * 2018-11-09 2022-08-17 Jiangsu Hengrui Medicine Co., Ltd. TGF-ß RECEPTOR FUSION PROTEIN PHARMACEUTICAL COMPOSITION AND USE THEREOF
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
WO2022212885A1 (en) * 2021-04-02 2022-10-06 Regeneron Pharmaceuticals, Inc. Stabilized formulations containing anti-muc16 x anti-cd3 bispecific antibodies
US11466094B2 (en) 2016-11-15 2022-10-11 Genentech, Inc. Dosing for treatment with anti-CD20/anti-CD3 bispecific antibodies
US11541126B1 (en) 2020-07-01 2023-01-03 Silverback Therapeutics, Inc. Anti-ASGR1 antibody TLR8 agonist comprising conjugates and uses thereof
WO2023062188A1 (en) * 2021-10-15 2023-04-20 Amgen Research (Munich) Gmbh Subcutaneous administration of cd19-binding t cell engaging antibodies
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
WO2023170474A1 (en) 2022-03-07 2023-09-14 Novimmune Sa Cd28 bispecific antibodies for targeted t cell activation
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
US11845799B2 (en) 2019-12-13 2023-12-19 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
US11958906B2 (en) 2022-04-13 2024-04-16 Genentech, Inc. Pharmaceutical compositions of mosunetuzumab and methods of use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070123479A1 (en) * 2003-05-31 2007-05-31 Micromet Ag Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
US20070178013A1 (en) * 2005-07-26 2007-08-02 Kalonia Devendra S Dual-detector systems and methods having utility in biomolecular measurements
US20070291265A1 (en) * 2006-04-13 2007-12-20 Mississippi State University Optical apparatus for simultaneously measuring the scattering and concentration of signals of macromolecules in a flow cell
US20100303827A1 (en) * 2007-11-28 2010-12-02 One Medimmune Way Protein Formulation
US20130035249A1 (en) * 2009-12-01 2013-02-07 Medimmune, Llc Methods and compositions for detecting and treating cea-expressing cancers

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070123479A1 (en) * 2003-05-31 2007-05-31 Micromet Ag Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
US20070178013A1 (en) * 2005-07-26 2007-08-02 Kalonia Devendra S Dual-detector systems and methods having utility in biomolecular measurements
US20070291265A1 (en) * 2006-04-13 2007-12-20 Mississippi State University Optical apparatus for simultaneously measuring the scattering and concentration of signals of macromolecules in a flow cell
US20100303827A1 (en) * 2007-11-28 2010-12-02 One Medimmune Way Protein Formulation
US20130035249A1 (en) * 2009-12-01 2013-02-07 Medimmune, Llc Methods and compositions for detecting and treating cea-expressing cancers

Cited By (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
US10087250B2 (en) 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10155815B2 (en) 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10781257B2 (en) 2013-02-26 2020-09-22 Roche GlyeArt AG Bispecific T cell activating antigen binding molecules
US10781258B2 (en) 2013-02-26 2020-09-22 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US11186650B2 (en) 2013-12-17 2021-11-30 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10174124B2 (en) 2013-12-17 2019-01-08 Genentech, Inc. Anti-CD3 antibodies and methods of use
US11530275B2 (en) 2013-12-17 2022-12-20 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10640572B2 (en) 2013-12-17 2020-05-05 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10865251B2 (en) 2013-12-17 2020-12-15 Genentech, Inc. Anti-CD3 antibodies and methods of use
US11732054B2 (en) 2013-12-17 2023-08-22 Genentech, Inc. Anti-CD3 antibodies and methods of use
US11117965B2 (en) 2014-08-04 2021-09-14 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US9914776B2 (en) 2014-08-04 2018-03-13 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10611841B2 (en) 2014-08-04 2020-04-07 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10611840B2 (en) 2014-08-04 2020-04-07 Hoffman-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11084877B2 (en) 2014-09-12 2021-08-10 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US11613587B2 (en) 2014-11-20 2023-03-28 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US11028142B2 (en) 2015-05-18 2021-06-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10442849B2 (en) 2015-05-18 2019-10-15 Tcr2 Therabeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11965012B2 (en) 2015-05-18 2024-04-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10358474B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10358473B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10501545B2 (en) 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
US10323094B2 (en) 2015-06-16 2019-06-18 Genentech, Inc. Humanized and affinity matured antibodies to FcRH5 and methods of use
US11466087B2 (en) 2015-06-16 2022-10-11 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
US11192950B2 (en) 2015-06-16 2021-12-07 Genentech, Inc. Humanized and affinity matured antibodies to FcRH5 and methods of use
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
US11242390B2 (en) 2016-03-22 2022-02-08 Hoffmann-La Roche Inc. Protease-activated T cell bispecific molecules
US11242376B2 (en) 2016-08-02 2022-02-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10882918B2 (en) 2016-09-30 2021-01-05 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11085021B2 (en) 2016-10-07 2021-08-10 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11377638B2 (en) 2016-10-07 2022-07-05 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10208285B2 (en) 2016-10-07 2019-02-19 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11466094B2 (en) 2016-11-15 2022-10-11 Genentech, Inc. Dosing for treatment with anti-CD20/anti-CD3 bispecific antibodies
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
CN108264559B (en) * 2016-12-30 2021-08-10 惠和生物技术(上海)有限公司 Tri-functional molecule combined with CD19, CD3 and T cell positive co-stimulatory molecule and application thereof
CN108264559A (en) * 2016-12-30 2018-07-10 上海近岸生物科技有限公司 Three functional moleculars and its application of a kind of combination CD19, CD3 and the positive costimulatory molecules of T cell
WO2018141910A1 (en) * 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
CN110582297A (en) * 2017-02-02 2019-12-17 安进研究(慕尼黑)有限责任公司 Low pH pharmaceutical compositions comprising T cell conjugated antibody constructs
JP2020506924A (en) * 2017-02-02 2020-03-05 アムジェン リサーチ (ミュニック) ゲゼルシャフト ミット ベシュレンクテル ハフツング Low pH pharmaceutical compositions containing T cell engage antibody constructs
JP7189141B2 (en) 2017-02-02 2022-12-13 アムジェン リサーチ (ミュニック) ゲゼルシャフト ミット ベシュレンクテル ハフツング Low pH Pharmaceutical Compositions Containing T Cell Engaging Antibody Constructs
WO2019020745A1 (en) * 2017-07-28 2019-01-31 F. Hoffmann-La Roche Ag Bispecific antibody formulation
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
WO2019234576A1 (en) 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Bispecific antibodies against ceacam5 and cd47
WO2020053301A1 (en) * 2018-09-11 2020-03-19 Ichnos Sciences S.A. Compositions comprising a bispecific antibody, bufffer and one or more stabilizing agents
WO2020056008A1 (en) 2018-09-12 2020-03-19 Silverback Therapeutics, Inc. Compositions for the treatment of disease with immune stimulatory conjugates
CN112789028A (en) * 2018-10-01 2021-05-11 美国安进公司 Methods for reducing aggregation of bispecific antibodies
EP3860567A4 (en) * 2018-10-01 2022-07-06 Amgen Inc. Methods for reducing aggregation of bispecific antibodies
EP3878461A4 (en) * 2018-11-09 2022-08-17 Jiangsu Hengrui Medicine Co., Ltd. TGF-ß RECEPTOR FUSION PROTEIN PHARMACEUTICAL COMPOSITION AND USE THEREOF
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
WO2020257407A1 (en) 2019-06-19 2020-12-24 Silverback Therapeutics, Inc. Anti-mesothelin antibodies and immunoconjugates thereof
WO2021028669A1 (en) * 2019-08-09 2021-02-18 Arecor Limited Novel composition comprising antibodies
CN114206391A (en) * 2019-08-09 2022-03-18 艾瑞克有限公司 Novel compositions comprising antibodies
WO2021035028A3 (en) * 2019-08-20 2021-04-08 Regeneron Pharmaceuticals, Inc. Formulation optimization for bispecific antibodies
WO2021053587A1 (en) 2019-09-18 2021-03-25 Klaus Strein Bispecific antibodies against ceacam5 and cd3
WO2021110647A1 (en) 2019-12-02 2021-06-10 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
EP3831849A1 (en) 2019-12-02 2021-06-09 LamKap Bio beta AG Bispecific antibodies against ceacam5 and cd47
US11845799B2 (en) 2019-12-13 2023-12-19 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
US11179473B2 (en) 2020-02-21 2021-11-23 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
US11541126B1 (en) 2020-07-01 2023-01-03 Silverback Therapeutics, Inc. Anti-ASGR1 antibody TLR8 agonist comprising conjugates and uses thereof
CN114246944A (en) * 2020-09-24 2022-03-29 盛禾(中国)生物制药有限公司 Pharmaceutical composition of bispecific antibody and application thereof
WO2022212885A1 (en) * 2021-04-02 2022-10-06 Regeneron Pharmaceuticals, Inc. Stabilized formulations containing anti-muc16 x anti-cd3 bispecific antibodies
WO2023062188A1 (en) * 2021-10-15 2023-04-20 Amgen Research (Munich) Gmbh Subcutaneous administration of cd19-binding t cell engaging antibodies
WO2023170474A1 (en) 2022-03-07 2023-09-14 Novimmune Sa Cd28 bispecific antibodies for targeted t cell activation
US11958906B2 (en) 2022-04-13 2024-04-16 Genentech, Inc. Pharmaceutical compositions of mosunetuzumab and methods of use

Similar Documents

Publication Publication Date Title
WO2016036678A1 (en) Formulations of bispecific antibodies
US20210290525A1 (en) Formulations of antibody
AU2018227036B2 (en) Formulations of monoclonal antibodies
JP7446356B2 (en) Anti-RSV monoclonal antibody preparation
RU2763787C2 (en) Stable aqueous compositions based on antibodies
KR20210104736A (en) Antibody formulation
EP3911298B1 (en) Formulations

Legal Events

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

Ref document number: 15837389

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15837389

Country of ref document: EP

Kind code of ref document: A1