WO2012028683A1 - Système de gel d'anticorps pour administration de médicament prolongée - Google Patents

Système de gel d'anticorps pour administration de médicament prolongée Download PDF

Info

Publication number
WO2012028683A1
WO2012028683A1 PCT/EP2011/065096 EP2011065096W WO2012028683A1 WO 2012028683 A1 WO2012028683 A1 WO 2012028683A1 EP 2011065096 W EP2011065096 W EP 2011065096W WO 2012028683 A1 WO2012028683 A1 WO 2012028683A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
gel
formulation
gel formulation
lyophilisate
Prior art date
Application number
PCT/EP2011/065096
Other languages
English (en)
Inventor
Margarida Rodrigues
Karin Schoenhammer
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Publication of WO2012028683A1 publication Critical patent/WO2012028683A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • 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
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators

Definitions

  • Monoclonal antibodies are typically formulated either in aqueous form ready for parenteral administration or as lyophilisates for reconstitution with a suitable buffer prior to administration. Such formulations provide immediate release of the mAb after administration; release is not sustained and the mAb is rapidly cleared from the site of administration.
  • reference 1 discloses the encapsulation of antibodies inside biodegradable poly(lactic-glycolic) acid microspheres. These are injected subcutaneously and provide sustained release.
  • Example 4 of reference 2 discloses a surfactant/solvent gel formulation for sustained delivery of particulate mAbs.
  • references 3 and 4 disclose topical delivery systems in which antibodies were dispersed within a poly(ethylene-co-vinyl acetate) matrix shaped as disks. After insertion into the body the disks release encapsulated antibody to local tissue over a sustained period.
  • Reference 5 discloses an intratumoral injectable gel drug delivery system for local delivery of radiolabelled immunotherapeutic mAbs.
  • a gel formulation of polyclonal antibodies for local delivery, using carboxymethylcellulose, is disclosed in reference 6
  • Reference 62 discloses an antibody gel formulation of an anti-sclerostin antibody.
  • the gel formulation therein was derived from lyophilized drug substance that was resuspended in a sodium phosphate buffer (PBS) to form a 150 mg/ml antibody-containing solution having a pH of 6.6.
  • PBS sodium phosphate buffer
  • the reconstituted lyophlisate formed a gel structure having a turbidity of 1350 NTU, which was capable of sustained release of active monomeric antibody in vitro.
  • the time required to achieve a turbidity of 1350 NTU was at least 20 minutes. Slow gellification of the mAb could allow dispersion of the antibody into underlying tissue and blood, resulting in reduced antibody concentration at the site of administration.
  • the present invention describes antibody formulations, e.g., anti-sclerostin antibody formulations, such as formulations of ANTIBODY 1, and methods for their manufacture, which (i) are capable of rapidly gelling and preserving an antibody in a native and therapeutically active state while achieving a depot effect without the use of polymers or other complex formulation reagents and (ii) provide high loading capacity, thus offering the potential for increased dose per application to patient.
  • anti-sclerostin antibody formulations such as formulations of ANTIBODY 1
  • methods for their manufacture which (i) are capable of rapidly gelling and preserving an antibody in a native and therapeutically active state while achieving a depot effect without the use of polymers or other complex formulation reagents and (ii) provide high loading capacity, thus offering the potential for increased dose per application to patient.
  • formulations take the form of a gel and are based on the surprising finding that rapid gellificaiton of certain monoclonal antibodies may be induced by using: 1) specific buffers to reconstitute a lyophilisate of the antibody or 2) specific buffers in combination with an aqueous antibody formulation that has not previously been lyophilized. Due to the rapid gellificaiton of the antibody, the resultant antibody gel formulations can be immediately administered to a patient as a depot (gel) system that releases active mAb over a sustained period of time without the use of current depot excipients.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising lyophilising a first aqueous formulation of ANTIBODY 1 at a first pH to give a ANTIBODY 1 lyophilisate; and reconstituting the lyophilisate with a MOPS or a HEPES buffer to provide a second aqueous formulation of ANTIBODY 1 having a second pH, the second pH being different from the first pH.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising lyophilising a first aqueous formulation of ANTIBODY 1 at a first pH to give a ANTIBODY 1 lyophilisate; and reconstituting the lyophilisate with a Tris buffer having a pH between about 7.8 and about 8.6 to provide a second aqueous formulation of ANTIBODY 1 having a second pH, the second pH being different from the first pH.
  • the first pH is lower than the second pH. In some embodiments of the aforementioned processes, the first pH is less than about 7.0. In some embodiments of the aforementioned processes, the first pH is in the range about 5.0 to about 6.0. In some embodiments of the aforementioned processes, the second pH and the first pH differ by at least one pH unit. In some embodiments of the aforementioned processes, the second pH is in the range about 6.0- about 8.0.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising reconstituting a ANTIBODY 1 lyophilisate with a MOPS or a HEPES buffer to provide an aqueous formulation of the ANTIBODY 1 antibody having a pH of about > 5.5 to about ⁇ 9.0
  • a process for preparing a gel formulation of ANTIBODY 1 comprising reconstituting a ANTIBODY 1 lyophilisate with a Tris buffer having a pH between about 7.8 and 8.6 to provide a gel formulation of ANTIBODY 1 having a pH of about > 5.5 to about ⁇ 9.0.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising reconstituting a ANTIBODY 1 lyophilisate with a MOPS or a HEPES buffer, wherein the a MOPS buffer has a pH between about 6.6 and about 7.4 and the HEPES buffer has a pH between about 7.0 and about 7.8; and allowing the reconstituted lyophilisate to form the gel formulation or changing the pH of the reconstituted lyophilisate to cause formation of the gel formulation.
  • gel formation occurs less than about 30 minutes after reconstituting. In some embodiments of the aforementioned processes, the step of reconstituting is carried out at a temperature between room temperature and 37°C.
  • kits comprising, a ANTIBODY 1 lyophilisate at a first pH value; and a MOPS or a HEPES buffer at a second pH, wherein said MOPS or HEPES buffer, when combined with said ANTIBODY 1 lyophilisate, is capable of providing a formulation of ANTIBODY 1 that spontaneously forms a gel.
  • kits comprising, a ANTIBODY 1 lyophilisate; and a Tris buffer having a pH between about 7.8 and 8.6, wherein mixing of the lyophilisate and the Tris buffer gives an aqueous formulation of ANTIBODY 1 which either spontaneously forms a gel, or is not a gel but will form a gel in vivo.
  • the lyophilisate includes one or more lyophilization stabilizers selected from the group consisting of: sugars, amino sugars, amino acids and/or surfactants.
  • the gel formulation can release antibody (e.g., ANTIBODY 1) in vivo for more than 7 days.
  • the gel formulation has a turbidity of about 1500 NTU to about 4000 NTU as measured by a HACH Tubidimeter 2100 AN. In other embodiments, the formulation has a turbidity of about 2000 NTU to about 3000 NTU.
  • the antibody e.g., ANTIBODY 1
  • the gel formulation has a turbidity of about 1500 NTU to about 4000 NTU as measured by a HACH Tubidimeter 2100 AN.
  • the formulation has a turbidity of about 2000 NTU to about 3000 NTU.
  • the antibody gel formulations of the invention, and formulations made using the methods and kits of the invention, typically do not include a gelling polymer.
  • gel formulations comprising, a sclerostin antibody (e.g., ANTIBODY 1) in about 80-90 mM, e.g., about 83mM, MOPS buffer, a sclerostin antibody (e.g., ANTIBODY 1) in about 80-90 mM, e.g., about 83mM, HEPES buffer and a sclerostin antibody (e.g., ANTIBODY 1) in about 80-90 mM, e.g., about 83mM, TRIS buffer.
  • a sclerostin antibody e.g., ANTIBODY 1
  • MOPS buffer e.g., MOPS buffer
  • a sclerostin antibody e.g., ANTIBODY 1
  • HEPES buffer e.g., HEPES buffer
  • a sclerostin antibody e.g., ANTIBODY 1
  • the gel formulation of the sclerostin antibody (e.g., ANTIBODY 1) is used in therapy, e.g., for use (a) in the treatment of bone injuries such as a bone fracture, or (b) in promoting osseointegration of a bone plate, pin, screw, prosthetic joint or dental implant.
  • the gel formulation of the sclerostin antibody (e.g., ANTIBODY 1) is used in the manufacture of a medicament for (a) the treatment of bone injuries such as a bone fracture, or (b) promoting osseointegration of a bone plate, pin, screw, prosthetic joint or dental implant.
  • the gel formulation reduces recovery time following injury or surgery.
  • a process for preparing a gel formulation of AANTIBODY 1 comprising adjusting a first aqueous formulation of ANTIBODY 1 at a first pH with a MOPS buffer to provide a second aqueous formulation of ANTIBODY 1 having a second pH, the second pH being different from the first pH.
  • the first pH is lower than the second pH. In some embodiments of the aforementioned processes, the first pH is ⁇ about 7.0. In some embodiments of the aforementioned processes, the first pH is in the range of about 5.0 to about 6.0. In some embodiments of the aforementioned processes, the second pH and the first pH differ by at least one pH unit. In some embodiments of the aforementioned processes, the second pH is in the range of about 6.0 to about 8.0. In some embodiments of the aforementioned processes, the first aqueous formulation of ANTIBODY 1 is an aqueous ANTIBODY 1 formulation that has not previously been lyophilized. In some embodiments of the aforementioned processes, gel formation occurs less than about 30 minutes after reconstituting. In some embodiments of the aforementioned processes, the adjusting is carried out at a temperature between room temperature and 37°C.
  • kits comprising, an aqueous formulation of ANTIBODY 1 at a first pH; and a MOPS buffer at a second H, wherein said MOPS buffer, when combined with said aqueous formulation of ANTIBODY 1, is capable of providing a formulation of ANTIBODY 1 that spontaneously forms a gel.
  • the gel formulation can release antibody (e.g., ANTIBODY 1) in vivo for more than 7 days.
  • the gel formulation has a turbidity of about 1500 NTU to about 4000 NTU as measured by a HACH Tubidimeter 2100 AN. In other embodiments, the gel formulation has a turbidity of about 2000 NTU to about 3000 NTU.
  • a gel formulation comprising, a sclerostin antibody in about 20-40 mM, e.g., 26mM to about 34mM, MOPS buffer.
  • the sclerostin antibody is the ANTIBODY 1 antibody.
  • gel formulations of the invention, and formulations made using the methods and kits of the invention typically do not include a gelling polymer.
  • FIG. 1 shows ANTIBODY 1 release (%, as measured by SEC) in triplicate (vial 1-3) over time (hours) from a gel formed from ANTIBODY 1 lyophilisate (DP) reconstituted with a PBS buffer.
  • the release experiments were performed at 37°C in 0.1M PBS buffer.
  • Figure 2 shows the % of released aggregation products from a gel formed from lyophilisate (DP) reconstitued with a PBS buffer, measured by SEC.
  • Figure 3 shows the % of released degradation products from a gel formed from lyophilisate (DP) reconstitued with a PBS buffer, measured by SEC
  • Figure 4 shows the activity (%) at two timepoints of released ANTIBODY 1 from a gel formed of a lyophilisate (DP) reconstitued with a PBS buffer. At both timepoints the released mAb from the gelling system showed high bioactivity. The dotted line is the mean.
  • Figure 5 shows the turbidity (NTU) increase over time (minutes) of ANTIBODY 1 lyophilisate (DP) reconstitued with different buffers at different pH levels. Note: the upper limit of the turbidimeter measurements is 4000NTU.
  • Figure 6 shows the effect of adding MOPS buffer pH 7.0 to ANTIBODY 1 drug substrate (DS).
  • the first two images are 1 mL of ANTIBODY 1 drug substance at two different concentrations in 6R vials.
  • the second two images are ANTIBODY 1 drug substance 1 min after addition of 100 mM
  • an antibody gel formulation prepared from a liquid (e.g., aqueous) antibody formulation that has not previously been lyophilized (referred to herein as an antibody "drug substance" or "DS"), e.g., a liquid (e.g., aqueous) ANTIBODY 1 drug substance.
  • DS antibody "drug substance” or "DS”
  • the novel antibody drug substances have the desirable property of rapidly gellifying following a pH change.
  • the pH of an antibody drug substance can be changed to initiate gelation.
  • the pH change can be achieved by adding acid or base to the antibody drug substance, or by adding a diluent, e.g., a buffer (such as a MOPS buffer).
  • the pH may be changed by at least one pH unit e.g.
  • the post-dilution pH (e.g., following addition of the buffer, e.g., a MOPS buffer, acid or base) may be higher than the starting pH or lower than the starting pH, preferably higher than the starting pH.
  • the MOPS buffer may have a lower (e.g., pH 5.0) or higher (e.g., pH 7.0) pH than that of the aqueous antibody drug substance.
  • the ANTIBODY 1 drug substance is in a Histidine buffer (e.g., 6.4 mM Histidine buffer). In one embodiment the ANTIBODY 1 drug substance is in a Histidine buffer having a pH of > 5.0, e.g., 5.3+0.1.
  • the pH change of the antibody (e.g., ANTIBODY 1) drug substance is achieved by adding a MOPS buffer (e.g., about 1 mM to about 1 M MOPS buffer, preferably about 100 mM) having a pH of about 7.0.
  • a MOPS buffer e.g., about 1 mM to about 1 M MOPS buffer, preferably about 100 mM
  • the concentration of the MOPS buffer following addition to the antibody (e.g., ANTIBODY 1) drug substance is about 20mM to about 30mM, e.g., about 26mM, for lOOmg/ml protein concentration. In another embodiment, the concentration of the MOPS buffer following addition to the antibody (e.g., ANTIBODY 1) drug substance is about 30mM to about 40mM, e.g., about 34mM, for 50mg/ml protein concentration.
  • the pH change e.g., caused by adding MOPS buffer to a ANTIBODY 1 drug substance
  • gelation ideally occurs spontaneously.
  • the pH change does not cause gelation, but is used to provide a formulation that is less painful for patient administration, with gelation being initiated after administration.
  • the pH change does not immediately cause gelation, but is used to provide a formulation that may be induced to form a gel
  • the kinetics of gelation caused by pH change may vary and gelation may occur quickly or slowly. For example, in some embodiments it may be substantially simultaneous with the the pH change. In other embodiments it may occur rapidly after the pH change (e.g. ⁇ about 60 minutes after, such as ⁇ about 45 minutes, ⁇ about 30 minutes, ⁇ about 20 minutes, ⁇ about 15 minutes, ⁇ about 14 minutes,
  • the pH change should not be so extreme as to irreversibly denature the mAb.
  • Such denaturation is also easily detected, and so an appropriate pH window can readily be identified which ensures appropriate gelation while avoiding irreversible denaturation.
  • the diluent e.g., a MOPS buffer
  • the antibody e.g., anti-sclerostin antibody, e.g., ANTIBODY 1
  • the pH may be > about 5.5 (e.g. > about 6.0, > about 6.5, etc.), while typically not being above about 9.0 e.g.
  • the aqueous formulation formed in this manner may include active ingredients in addition to the mAb.
  • active ingredients in addition to the mAb.
  • further pharmacological agents may be included, such as chemotherapeutic compounds. These can be incorporated into a gel during its formation, facilitating co-delivery together with the mAb.
  • the pH following addition of the diluent, e.g., a MOPS buffer, to the antibody (e.g., ANTIBODY 1) drug substance is > 5.3, e.g., > 5.5 to ⁇ 8.0.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising adjusting a first aqueous formulation of ANTIBODY 1 at a first pH with a MOPS buffer to provide a second aqueous formulation of ANTIBODY 1 having a second pH, the second pH being different from the first pH.
  • the first pH is lower than the second pH. In some embodiments of the aforementioned processes, the first pH is ⁇ about 7.0. In some embodiments of the aforementioned processes, the first pH is in the range of about 5.0- about 6.0. In some embodiments of the aforementioned processes, the second pH and the first pH differ by at least one pH unit. In some embodiments of the aforementioned processes, the second pH is in the range of about 6.0 to about 8.0. In some embodiments of the aforementioned processes, the first aqueous formulation of ANTIBODY 1 is an aqueous ANTIBODY 1 formulation that has not previously been lyophilized. In some embodiments of the aforementioned processes, gel formation occurs less than about 30 minutes after reconstituting. In some embodiments of the aforementioned processes, the adjusting is carried out at a temperature between room temperature and 37°C.
  • kits comprising, an aqueous formulation of ANTIBODY 1 at a first pH; and a MOPS buffer at a second pH, wherein said MOPS buffer, when combined with said aqueous formulation of ANTIBODY 1, is capable of providing a formulation of ANTIBODY 1 that spontaneously forms a gel.
  • gel refers to a substance (e.g., a solution or formulation) having a turbidity of at least about 1000 NTU, e.g., about 1200 NTU, 1400 NTU, 1600 NTU, 1800 NTU, 2000 NTU, 2200 NTU, 2400 NTU, 2600 NTU, 2800 NTU, 3000 NTU, 3200 NTU, 3400 NTU, 3600 NTU, 3800 NTU, 3800 NTU, 4000 NTU, 4200 NTU, 4400 NTU, 4600 NTU, 4800 NTU, 5000 NTU, 5200 NTU.
  • a substance e.g., a solution or formulation
  • a gel solution of the disclosure has a turbidity of at least about 1000 NTU to about 4000 NTU, e.g., at least about 1000 NTU, at least about 1500 NTU, at least about 2000 NTU, at least about 3000 NTU, at least about 4000 NTU, etc. to .
  • the gel formulation has a turbidity of about 1500 NTU to about 4000 NTU as measured by a HACH Tubidimeter 2100 AN.
  • the gel formulation has a turbidity of about 2000 NTU to about 3000 NTU.
  • the term "gellify" is used to refer to the process whereby a formulation takes on gel properties.
  • the gel formulation can release antibody (e.g., ANTIBODY 1) in vivo for more than 7 days.
  • a gel formulation comprising, a sclerostin antibody in about 20 mM to about 40 mM, e.g., about 26mM to about 34mM, MOPS buffer.
  • the sclerostin antibody is the ANTIBODY 1 antibody.
  • gel formulations of the invention, and formulations made using the methods and kits of the invention typically do not include a gelling polymer.
  • liquid antibody formulations i.e., antibody drug substances
  • lyophilisate drug product
  • DP drug product
  • Techniques for lyophilisation of mAbs are well known in the art e.g. see references 7 to 15. For example, monoclonal antibody products SYNAGISTM, REMICADETM,
  • NEUTROSPECTM, RAPTIVATM, SIMULECTTM, XOLAIRTM and HERCEPTINTM are supplied as lyophilisates.
  • the pH of the antibody drug substance to be lyophilized (i.e., the pre-lyophilisation pH) should be selected or controlled to ensure that gelation does not occur prior to lyophilization, unless such is specifically desired. Gelation is easily detected and so it is simple to select appropriate pH conditions for any particular mAb.
  • the pre-lyophilisation pH of the drug substance will be ⁇ about 7.0 (e.g. ⁇ 6.5, ⁇ 6.0, ⁇ 5.5, etc.), while typically not being below about 4.5 e.g. in the range about 4.5- about 6.5 or about 5.0- about 6.0.
  • a pre-lyophilisation pH of 5.3+0.1 is suitable for antibody ANTIBODY 1.
  • the lyophilisate may include, in addition to the mAb, lyophilisation stabilisers such as sugars, amino sugars, amino acids and/or surfactants.
  • the lyophilisate may include one or more of: glycine, mannitol, sucrose, trehalose, hydroxyethyl starch and/or polyethylene glycol.
  • glycine, mannitol, sucrose, trehalose, hydroxyethyl starch and/or polyethylene glycol will be present in the pre-lyophilisation aqueous formulation.
  • Further components which may be present in the pre-lyophilisation aqueous formulation include buffers, salts, etc.
  • a formulation containing sucrose, arginine and polysorbate 80 has been shown to be suitable for lyophilisation of antibody ANTIBODY 1.
  • the lyophilisate may include active ingredients in addition to the mAb.
  • further pharmacological agents may be included, such as chemotherapeutic compounds.
  • methotrexate may be included, and it is known to include methotrexate sodium in lyophilisates.
  • the ANTIBODY 1 lyophilisate is 150 mg/ml ANTIBODY 1, 270 mM Sucrose, 51 mM Arg-HCl, 30 mM Histidine, 0.06% Tween 80, pH 5.3.
  • a lyophilisate Before a lyophilisate can be administered to a patient it should be reconstituted with an aqueous reconstituent. This step permits the antibody in the lyophilisate to re-dissolve.
  • Reconstitution of the lyophilisate using an aqueous reconstituent e.g., an aqueous solution such as a buffer
  • an aqueous antibody solution hereinafter referred to as an antibody "reconstituate”
  • the process of reconstitution can facilitates a change in the formulation pH, which can initiate the gelation process. This may be achieved by employing a reconstituent having a different pH than the lyophilisate pH.
  • Typical reconstituents for lyophilised mAbs include sterile water or buffer, optionally containing a preservative. Rather than reconstitute the anti-sclerositin (e.g., ANTIBODY 1) lyophilisate with water, however, the instant disclosure provides specific buffers as reconstituents. Buffered reconstituents are helpful in adjusting the pH of the antibody formulation to give a post- reconstitution pH that differs from the pre-lyophilisation pH.
  • Suitable reconstituent buffers include TRIS, MOPS, HEPES, ACES, PIPES, MOPSO, TES, DIPSO, BES, TAPSO, MES, citrate, maleate, histidine, carbonate, and phosphate buffer.
  • the buffer used for reconstitution of the lyophilisate is at a concentration of from about 1 mM to about 1 M, e.g., about 10 mM, about 100 mM, etc.
  • the buffer concentration e.g., MOPS, HEPES or Tris buffer concentration, is about 100 mM.
  • the final concentration of the buffer in the reconstitute is about 80-90 mM, e.g., 83mM.
  • the pH of the buffer used for reconstitution of the antibody lyophilisate is from about 6.8 to about 8.6, e.g., Tris buffer at about pH 7.8 to about pH 8.6, HEPES buffer at about pH 7.0 to about pH 7.8, and MOPS buffer at about pH 6.8 to about pH 7.4.
  • the aqueous reconstituent may include pharmacological agents, such as chemotherapeutic compounds, which can be incorporated into the gel during its formation, facilitating co-delivery together with the mAb.
  • pharmacological agents such as chemotherapeutic compounds
  • the pH of the antibody (e.g, ANTIBODY 1) drug substance i.e., the pre- lyophilization pH
  • the pH of the antibody (e.g., ANTIBODY 1) reconstituate i.e., the post-reconstitution pH
  • the pre-lyophilisation pH should ideally be selected or controlled to ensure that gelation does not occur prior to lyophilization, unless such is specifically desired. Gelation is easily detected and so it is simple to select appropriate pH conditions for any particular mAb.
  • the post-reconstitution pH should not be so extreme as to irreversibly denature the mAb, though. Such denaturation is also easily detected, and so an appropriate pH window for the pre- lyophilisate can readily be identified to avoid both gelation and irreversible denaturation.
  • the post-reconstitution pH of the antibody reconstituate typically differs from the pre-lyophilisation pH of the antibody drug substance by at least one pH unit e.g. a difference of >1.0, > 1.5, >2, >2.5, etc.
  • the post-reconstitution pH may be higher than the pre-lyophilisation pH or lower than the pre- lyophilisation pH, preferably it is higher than the pre-lyophilisation pH.
  • the post-reconstitution pH of the reconstituate will be > about 5.4 (e.g._> about 5.5, > about 6.0, > about
  • the pre-lyophilisation pH of the drug substance will be ⁇ about 7.0 (e.g. ⁇ 6.5, ⁇ 6.0, ⁇ 5.5, etc.), while typically not being below about 4.5 e.g. in the range about 4.5- about 6.5 or about 5.0- about 6.0.
  • a pre-lyophilisation pH of 5.3+0.1 is suitable for antibody ANTIBODY 1 drug substance.
  • a pH of about 6.6+0.1 is provided for an antibody reconstituate, e.g., a ANTIBODY1 reconstituate.
  • this is achieved by reconstitution of the lyophilisate (e.g, a ANTIBODY 1 lyophilisate) with Tris buffer at about pH 7.8 to about pH 8.6, HEPES buffer at about pH 7.0 to about pH 7.8, or MOPS buffer at about pH 6.8 to about pH 7.4, which causes gelation to occur rapidly after reconstitution.
  • the post-reconstitution pH of the reconstituate will depend on the pre- lyophilisation pH of the drug substance and the pH of the aqueous reconstituent.
  • Appropriate pH values can be selected for the drug substance and reconstituent according to the pH-related gelation properties of the antibody in question.
  • the reconstituent may have a pH below about 7.0 (e.g. below about 6.8, such as in the range about 5.0- about 6.8 or about 5.4- about 6.4) or a pH above about 7.0 (e.g. above about 7.2, such as in the range about 7.2- about 8.5 or about 7.4 to about 8.0).
  • the pre-lyophilisation pH is below pH 7.0 then a reconstituent with pH above about 7.0 will be used, and vice versa.
  • the anti-sclerositin antibody e.g., ANTIBODY 1
  • the post-reconstitution pH may be further changed (for example by at least one pH unit e.g. a difference of >1.0,_>1.5, >2, >2.5, etc.) by addition of acid or base.
  • the post-reconstitution pH may also be modified by administration of the formulation to a mammal, with the pH altering in vivo.
  • gelation is initiated spontaneously after reconstitution, or requires further alteration of the reconstituted formulation, the kinetics of gelation may vary. Thus gelation may occur quickly or slowly. For example, in some embodiments it may be substantially simultaneous with reconstitution.
  • a process for preparing a gel formulation of ANTIBODY 1, comprising lyophilising a first aqueous formulation of ANTIBODY 1 at a first pH to give a ANTIBODY 1 lyophilisate; and reconstituting the lyophilisate with a MOPS or a HEPES buffer to provide a second aqueous formulation of ANTIBODY 1 having a second pH, the second pH being different from the first pH.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising lyophilising a first aqueous formulation of ANTIBODY 1 at a first pH to give a ANTIBODY 1 lyophilisate; and reconstituting the lyophilisate with a Tris buffer having a pH between about 7.8 and about 8.6 to provide a second aqueous formulation of ANTIBODY 1 having a second pH, the second pH being different from the first pH.
  • the first pH is lower than the second pH. In some embodiments of the aforementioned processes, the first pH is less than about 7.0. In some embodiments of the aforementioned processes, the first pH is in the range about 5.0 to about 6.0. In some embodiments of the aforementioned processes, the second pH and the first pH differ by at least one pH unit. In some embodiments of the aforementioned processes, the second pH is in the range about 6.0- about 8.0.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising reconstituting a ANTIBODY 1 lyophilisate with a MOPS or a HEPES buffer to provide an aqueous formulation of the ANTIBODY 1 antibody having a pH of about > 5.5 to about ⁇ 9.0
  • a process for preparing a gel formulation of ANTIBODY 1 comprising reconstituting a ANTIBODY 1 lyophilisate with a Tris buffer having a pH between about 7.8 and 8.6 to provide a gel formulation of ANTIBODY 1 having a pH of about > 5.5 to about ⁇ 9.0.
  • a process for preparing a gel formulation of ANTIBODY 1 comprising reconstituting a ANTIBODY 1 lyophilisate with a MOPS or a HEPES buffer, wherein the a MOPS buffer has a pH between about 6.6 and about 7.4 and the HEPES buffer has a pH between about 7.0 and about 7.8; and allowing the reconstituted lyophilisate from step a) to form the gel formulation or changing the pH of the reconstituted lyophilisate from step a) to cause formation of the gel formulation.
  • gel formation occurs less than about 30 minutes after reconstituting. In some embodiments of the aforementioned processes, the step of reconstituting is carried out at a temperature between room temperature and 37°C.
  • kits comprising, a ANTIBODY 1 lyophilisate at a first pH value; and a MOPS or a HEPES buffer at a second pH, wherein said MOPS or HEPES buffer, when combined with said ANTIBODY 1 lyophilisate, is capable of providing a formulation of ANTIBODY 1 that spontaneously forms a gel.
  • kits comprising, a ANTIBODY 1 lyophilisate; and a Tris buffer having a pH between about 7.8 and 8.6, wherein mixing of the lyophilisate and the Tris buffer gives an aqueous formulation of ANTIBODY 1 which either spontaneously forms a gel, or is not a gel but will form a gel in vivo.
  • the lyophilisate includes one or more lyophilization stabilizers selected from the group consisting of: sugars, amino sugars, amino acids and/or surfactants.
  • the gel formulation can release antibody (e.g., ANTIBODY 1) in vivo for more than 7 days.
  • the gel formulation has a turbidity of about 1500 NTU to about 4000 NTU as measured by a HACH Tubidimeter 2100 AN. In other embodiments, the formulation has a turbidity of about 2000 NTU to about 3000 NTU.
  • the antibody e.g., ANTIBODY 1
  • the gel formulation has a turbidity of about 1500 NTU to about 4000 NTU as measured by a HACH Tubidimeter 2100 AN.
  • the formulation has a turbidity of about 2000 NTU to about 3000 NTU.
  • the antibody gel formulations of the invention, and formulations made using the methods and kits of the invention, typically do not include a gelling polymer.
  • gel formulations comprising, a sclerostin antibody (e.g., ANTIBODY 1) in about 80-90 mM, e.g., 83mM, MOPS buffer, a sclerostin antibody (e.g., ANTIBODY 1) in about 80- 90 mM, e.g., 83mM, HEPES buffer and a sclerostin antibody (e.g., ANTIBODY 1) in about 80-90 mM, e.g., 83mM, TRIS buffer.
  • a sclerostin antibody e.g., ANTIBODY 1
  • MOPS buffer e.g., MOPS buffer
  • a sclerostin antibody e.g., ANTIBODY 1
  • HEPES buffer e.g., HEPES buffer
  • a sclerostin antibody e.g., ANTIBODY 1
  • the gel formulation of the sclerostin antibody (e.g., ANTIBODY 1) is used in therapy, e.g., for use (a) in the treatment of bone injuries such as a bone fracture, or (b) in promoting osseointegration of a bone plate, pin, screw, prosthetic joint or dental implant.
  • the gel formulation of the sclerostin antibody (e.g., ANTIBODY 1) is used in the manufacture of a medicament for (a) the treatment of bone injuries such as a bone fracture, or (b) promoting osseointegration of a bone plate, pin, screw, prosthetic joint or dental implant.
  • the gel formulation reduces recovery time following injury or surgery.
  • the gel formulation reduces recovery time following injury or surgery.
  • the invention provides gel formulations of mAbs. These formulations can give sustained release of the mAb in vivo.
  • the gel formulation is physically distinct from mere antibody precipitates and opalescent turbid antibody suspensions, both of which are known in the art ⁇ e.g. see references 16 & 17).
  • the gels have not been subjected to detailed rheological analysis, once formed they are structurally stable e.g. they do not appreciably flow out of an inverted test tube, and water droplets will stay on the gel surface rather than penetrate it.
  • an advantage of the disclosed gel formulations is that they do not require the presence of the polymers, additives or excipients that are currently used for sustained mAb release.
  • the formulation does not have to include a gelling polymer, such as celluloses or polyacrylates or polyvinyl alcohols.
  • the capacity for gel formation is intrinsic in the mAb itself rather than in any non-mAb component in the formulation (including any non-mAb component which may be attached to the mAb).
  • it may be useful to include such gelling polymers ⁇ e.g. to slow down release of mAb from the gel), but their absence is preferred. The absence of extrinsic gelling components reduces the potential for adverse patient reactions.
  • Gel formulations of the invention are typically turbid.
  • they may have a turbidity above about 500 NTU (Nephelometric Turbidity Units) e.g. > about 750 NTU, > about 1000 NTU, > about 1250 NTU, > about 1500 NTU, > about 2000 NTU, > about 2500 NTU, > about 3000 NTU, > about 3500 NTU, > about 4000 NTU etc. when measured at 25°C and atmospheric pressure.
  • NTU Nephelometric Turbidity Units
  • a useful gel formulation of antibody ANTIBODY 1 may have a turbidity of about 1350 NTU to about 4000 NTU, about 1500 NTU to about 4000 NTU, or about 2000 NTU to about 3000 NTU, e.g., about at least 1350 NTU or about at least 1500 NTU.
  • gel formulations of the invention are their ability to release antibody in active form into surrounding aqueous media.
  • the gel can be contacted with an aqueous medium (whether in vitro or in vivo) and antibodies can transfer passively from the gel into the medium in active form. After release they can interact with target antigens, either locally or remotely.
  • Gel formulations of the invention may be able to release antibody for more than about 2 days e.g. > about 3 days, > about 4 days, > about 5 days, > about 6 days, > about 7 days, > about 10 days, > about 14 days, > about 21 days, > about 28 days, etc. Release typically occurs at a higher initial rate which decreases over time.
  • Gel formulations of the invention are pharmaceutically acceptable and are suitable for administration to a patient.
  • mAb and water may include further components, including those typical of pharmaceutical formulations buffers, salts, amino acids, glycerol, alcohols, preservatives, surfactants, etc.
  • buffers including those typical of pharmaceutical formulations buffers, salts, amino acids, glycerol, alcohols, preservatives, surfactants, etc.
  • preservatives including those typical of pharmaceutical formulations buffers, salts, amino acids, glycerol, alcohols, preservatives, surfactants, etc.
  • surfactants etc.
  • mAbs as the active ingredient of pharmaceuticals is now widespread, including the products HERCEPTINTM (trastuzumab), RITUXANTM (ntuximab), SYNAGISTM (palivizumab), etc.
  • Techniques for purification of mAbs to a pharmaceutical grade are well known in the art.
  • the gel formulation will usually be sterile, at least at the time of its formation.
  • the composition will usually be non-pyrogenic e.g. containing ⁇ about 1 EU (endotoxin unit, a standard measure) per dose, and preferably ⁇ about 0.1 EU per dose.
  • the composition is preferably gluten free.
  • reconstitution gives a mAb concentration of at least about 50 mg/rriL is typical e.g. > about 100 mg/rriL, > about 150 mg/rriL, > about 200 mg/rriL, > about 250 mg/rriL, etc. These concentrations are achievable in aqueous formulations e.g. SYNAGISTM is provided for reconstitution to give a mAb concentration of 100 mg/mL.
  • the pH change is initiated by adding a specific diluent (e.g., a buffer) to an antibody (e.g., ANTIBODY 1) drug substance
  • a specific diluent e.g., a buffer
  • an antibody e.g., ANTIBODY 1
  • the mAb concentration is at least about 50 mg/mL, e.g. > about 100 mg/mL, > about 150 mg/mL, > about 200 mg/mL, > about 250 mg/mL, etc.
  • a mAb preferably make up at least about 80% by weight ⁇ e.g. at least about 90%, about 95%, about 97%, about 98%, about 99% or more) of the total protein in the formulation.
  • the mAb is thus in purified form.
  • Gel formulations of the invention can be used to treat or prevent a variety of diseases or disorders.
  • the gel is suitable for treatment of bone injuries.
  • the gel can be formed at the site of the bone injury and can stay in local contact with it while releasing its active mAb ingredient.
  • an anti-sclerostin antibody such as ANTIBODY 1
  • the gel may be applied at the site of a bone fracture. Such an application would reduce healing time. This embodiment would be particularly useful for the treatment of open fractures, complete fractures, spiral fractures or multi-fragmentary fractures.
  • a gel comprising an anti-sclerostin antibody such as ANTIBODY 1 may be used as a slow-release depot system for the treatment of osteoporosis.
  • the gel may also be applied at a site where a bone prosthesis is used, to promote osseointegration.
  • the gel may be applied at the site where a bone plate, pin or screw is located.
  • plates, pins or screws may be used to assist with fracture healing.
  • the gel may be coated onto the plate, pin or screw, prior to fixation to the bone.
  • the gel may be applied subsequent to fixation of the plate, pin or screw.
  • the plates, pins and screws may be made out of various materials, or combinations of materials such as stainless steel, titanium, ceramic, collagen or plastic.
  • Various types of plates, pins and screws used with bone and fracture healing are known in the art, and various types are summarised in reference 19.
  • the gel may be applied at a site of joint replacement, to promote osseointegration of the prosthesis.
  • joint replacements typically include hip, knee, shoulder and elbow replacements.
  • the gel may be placed into the bone marrow cavity prior to fixation of the artificial joint.
  • the gel may be used as a filler following fixation of the artificial joint.
  • the gel is also suitable for treatment of dental disorders and for improving the success of dental implants.
  • a mAb e.g. an anti-sclerostin antibody
  • the implant may be a re-implantation of a subject's own tooth (e.g. lost through trauma) or a prosthetic implant (made of, for example, plastic, ceramic, metal or from stem cells as described in WO2004/074464).
  • the gel is also suitable for treatment of respiratory diseases.
  • Topical treatment of lung disease e.g. COPD
  • mAbs is known in the art, such as by delivery of an anti-inflammatory mAb.
  • the gel is useful for treatment of osteo- or psoriatic- or rheumatoid arthritis.
  • Arthritis therapy by mAbs is well known in the art e.g. using adalimumab (HUMIRATM) or infliximab (REMICADETM).
  • the gel is also useful for local treatment of tumours.
  • Tumour therapy by mAbs is known in the art e.g. using trastuzumab (HERCEPTINTM), rituximab (RITUXANTM or MABTHERATM).
  • the gel is also useful for topical treatment of skin to aid healing and/or regeneration.
  • Skin treatment by mAbs is known in the art e.g. using efalizumab (RAPTIVATM).
  • a gel formulation of the invention may form in vitro and then be administered to a patient, or it may form in vivo after its ingredients have been administered. Administration will typically be via a syringe.
  • Patients will receive an effective amount of the mAb active ingredient i.e. an amount that is sufficient to detect, treat, ameliorate, or prevent the disease or disorder in question. Therapeutic effects may also include reduction in physical symptoms.
  • the optimum effective amount and concentration of mAb in a gel for any particular subject will depend upon various factors, including the patient's age, size, health and/or gender, the nature and extent of the condition, the activity of the particular mAb, the rate of its clearance by the body, and also on any possible further therapeutic(s) administered in combination with the mAb.
  • the effective amount delivered for a given situation can be determined by routine experimentation and is within the judgment of a clinician.
  • an effective dose may be from about 0.01 mg/kg to about 50 mg/kg, or about 0.05 mg/kg to about 30 mg/kg, e.g., 10 mg/kg.
  • HERCEPTINTM is administered with an initial loading dose of 4 mg/kg body weight and a weekly maintenance dose of 2 mg/kg body weight; RITUXANTM is administered weekly at 375 mg/m 2 ; SYNAGISTM is administered intramuscularly at 15 mg/kg body weight; etc.
  • the invention provides a method for delivering a monoclonal antibody to a mammal (e.g. a human), comprising a step of administering to the patient a gel formulation of the invention.
  • the invention also provides a method for delivering a monoclonal antibody to a mammal, comprising steps of: (i) preparing an aqueous formulation of the monoclonal antibody ⁇ e.g. as described above), wherein the aqueous formulation will form a gel after x minutes of its preparation; and (ii) administering the aqueous formulation to the patient within x minutes of its preparation.
  • the invention also provides a method for delivering a monoclonal antibody to a mammal, comprising steps of: (i) preparing an aqueous formulation of the monoclonal antibody (e.g.
  • the invention also provides formulations of the invention for use as medicaments e.g. for use in delivering a monoclonal antibody to a mammal.
  • the mammal is preferably a human but may also be, for example, a horse or a cow or a dog or a cat.
  • the mAb will ideally be chosen to match the target species e.g. a human antibody for human administration, an equine antibody for horses, a canine antibody for dogs, etc. If native host antibodies are not available then transfer of antibody specificity from one species to another can be achieved by transfer of CDR residues (and typically, in addition, one or more framework residues) from a donor antibody into a recipient framework from the host species e.g. as in humanisation. Equinised, bovinised, caninised, camelised and felinised antibodies are known in the art.
  • mAb ANTIBODY 1 these methods and uses may be for treating a bone injury. Dosage can be by a single dose schedule or a multiple dose schedule.
  • Ingredients for forming gels may be supplied in hermetically-sealed containers.
  • the monoclonal antibody is the monoclonal antibody
  • the invention concerns the formulation of monoclonal antibodies.
  • monoclonal as originally used in relation to antibodies referred to antibodies produced by a single clonal line of immune cells, as opposed to “polyclonal” antibodies that, while all recognizing the same target protein, were produced by different B cells and would be directed to different epitopes on that protein.
  • the word “monoclonal” does not imply any particular cellular origin, but refers to any population of antibodies that display a single binding specificity and affinity for a particular epitope in the same target protein. This usage is normal e.g.
  • a mAb may be produced using any suitable protein synthesis system, including immune cells, non-immune cells, acellular systems, etc.
  • a mAb can thus be produced by a variety of techniques, including conventional monoclonal antibody methodology ⁇ e.g. the standard somatic cell hybridization technique of Kohler & Milstein), by viral or oncogenic transformation of B lymphocytes, by combinatorial synthesis, by phage display, etc.
  • Antibodies used with the invention can take various forms. For instance, they may be native antibodies, as naturally found in mammals. Native antibodies are made up of heavy chains and light chains. The heavy and light chains are both divided into variable domains and constant domains. The ability of different antibodies to recognize different antigens arises from differences in their variable domains, in both the light and heavy chains. Light chains of native antibodies in vertebrate species are either kappa ( ⁇ ) or lambda ( ⁇ ), based on the amino acid sequences of their constant domains. The constant domain of a native antibody's heavy chains will be ⁇ , ⁇ , ⁇ , ⁇ or ⁇ , giving rise respectively to antibodies of IgA, IgD, IgE, IgG, or IgM class.
  • Classes may be further divided into subclasses or isotypes e.g. IgGl, IgG2, IgG3, IgG4, IgA, IgA2, etc.
  • Antibodies may also be classified by allotype e.g. a j heavy chain may have Glm allotype a, f, x or z, G2m allotype n, or G3m allotype bO, bl, b3, b4, b5, c3, c5, gl, g5, s, t, u, or v; a ⁇ light chain may have a Km(l), Km(2) or Km(3) allotype.
  • a native IgG antibody has two identical light chains (one constant domain C L and one variable domain V L ) and two identical heavy chains (three constant domains C H I , C H 2 & C H 3 and one variable domain V H ), held together by disulfide bridges.
  • the domain and three-dimensional structures of the different classes of native antibodies are well known.
  • an antibody of the invention has a light chain with a constant domain, it may be a ⁇ or ⁇ light chain.
  • an antibody of the invention may be an a, ⁇ , ⁇ , ⁇ or ⁇ heavy chain.
  • Heavy chains in the ⁇ class i.e. IgG antibodies are preferred.
  • Antibodies of the invention may be fragments of native antibodies that retain antigen binding activity. For instance, papain digestion of native antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual "Fc” fragment without antigen-binding activity. Pepsin treatment yields a "F(ab') 2 " fragment that has two antigen-binding sites. "Fv” is the minimum fragment of a native antibody that contains a complete antigen-binding site, consisting of a dimer of one heavy chain and one light chain variable domain. Thus an antibody of the invention may be Fab, Fab', F(ab') 2 , Fv, or any other type, of fragment of a native antibody.
  • An antibody of the invention may be a "single-chain Fv" (“scFv” or “sFv”), comprising a VH and VL domain as a single polypeptide chain [20-22].
  • scFv single-chain Fv
  • the VH and VL domains are joined by a short polypeptide linker (e.g. >12 amino acids) between the VH and VL domains that enables the scFv to form the desired structure for antigen binding.
  • a typical way of expressing scFv proteins, at least for initial selection is in the context of a phage display library or other combinatorial library [23-25]. Multiple scFvs can be linked in a single polypeptide chain [26].
  • An antibody of the invention may be a "diabody” or “triabody” etc. [27-30], comprising multiple linked Fv (scFv) fragments.
  • scFv linked Fv
  • VH and VL domains that is too short to allow them to pair with each other (e.g. ⁇ 12 amino acids)
  • they are forced instead to pair with the complementary domains of another Fv fragment and thus create two antigen-binding sites.
  • These antibodies may include CH and/or CL domains.
  • An antibody of the invention may be a single variable domain or VHH antibody.
  • Antibodies naturally found in camelids (e.g. camels and llamas) and in sharks contain a heavy chain but no light chain. Thus antigen recognition is determined by a single variable domain, unlike a mammalian native antibody [31-33].
  • the constant domain of such antibodies can be omitted while retaining antigen-binding activity.
  • One way of expressing single variable domain antibodies, at least for initial selection, is in the context of a phage display library or other combinatorial library [34].
  • An antibody of the invention may be a "domain antibody” (dAb).
  • dAbs are based on the variable domains of either a heavy or light chain of a human antibody and have a molecular weight of approximately 13 kDa (less than one-tenth the size of a full antibody).
  • dAbs are cleared from the body quickly and so benefit from a sustained release system, but can additionally be sustained in circulation by fusion to a second dAb that binds to a blood protein (e.g. to serum albumin), by conjugation to polymers (e.g. to a polyethylene glycol), or by other techniques.
  • a blood protein e.g. to serum albumin
  • polymers e.g. to a polyethylene glycol
  • the antibody may have a scaffold which is based on the fibronectin type ⁇ domain, as disclosed in reference 35 e.g. an adnectin or trinectin.
  • the fibronectin-based scaffold is not an immunoglobulin, although the overall fold is closely related to that of the smallest functional antibody fragment. Because of this structure the non-immunoglobulin antibody mimics antigen binding properties that are similar in nature and affinity to those of natural antibodies.
  • the Fnffl domain has 7 or 8 beta strands which are distributed between two beta sheets, which themselves pack against each other to form the core of the protein, and further containing loops (analogous to antibody CDRs) which connect the beta strands to each other and are solvent exposed.
  • the Fnlll loops can be replaced with immunoglobulin CDRs using standard cloning techniques, and can be used in a loop randomization and shuffling strategy in vitro that is similar to the process of affinity maturation of antibodies in vivo.
  • the Fnlll scaffold may be based on the tenth module of fibronectin type III ⁇ i.e. 10Fn3).
  • the term "antibody” as used herein encompasses a range of proteins having diverse structural features, but usually including at least one immunoglobulin domain, having an all- ⁇ protein fold with a 2-layer sandwich of anti-parallel ⁇ -strands arranged in two ⁇ -sheets. In all embodiments, however, the mAb has the ability to form a gel as described herein. Although not all mAbs will have this inherent gelation property, it is simple to determine if it is possessed by any particular mAb e.g. by detecting physicochemical changes after lyophilisation and reconstitution as described above.
  • Antibodies used with the invention may include a single antigen-binding site ⁇ e.g. as in a Fab fragment or a scFv) or multiple antigen-binding sites ⁇ e.g. as in a F(ab') 2 fragment or a diabody or a native antibody). Where an antibody has more than one antigen-binding site then advantageously it can result in cross-linking of antigens.
  • the antibody may be mono-specific ⁇ i.e. all antigen-binding sites recognize the same antigen) or it may be multi-specific ⁇ i.e. the antigen- binding sites recognise more than one antigen).
  • An antibody of the invention may include a non-protein substance e.g. via covalent conjugation.
  • an antibody may include a radio-isotope e.g. the ZEVALINTM and BEXXARTM products include 90 Y and 13 l l isotopes, respectively.
  • an antibody may include a cytotoxic molecule e.g. MYLOTARGTM is linked to N-acetyl-y-calicheamicin, a bacterial toxin.
  • an antibody may include a covalently-attached polymer e.g. attachment of polyoxyethylated polyols or polyethylene glycol (PEG) has been reported to increase the circulating half-life of antibodies.
  • an antibody can include one or more constant domains ⁇ e.g. including C H or C L domains).
  • the constant domains may form a ⁇ or ⁇ light chain or an ⁇ , ⁇ , ⁇ , ⁇ or ⁇ heavy chain.
  • an antibody may be a native constant domain or a modified constant domain.
  • a heavy chain may include either three (as in ⁇ , ⁇ , ⁇ classes) or four (as in ⁇ , ⁇ classes) constant domains.
  • Constant domains are not involved directly in the binding interaction between an antibody and an antigen, but they can provide various effector functions, including but not limited to: participation of the antibody in antibody-dependent cellular cytotoxicity
  • ADCC ADCC
  • Clq binding complement dependent cytotoxicity
  • Fc receptor binding Fc receptor binding
  • phagocytosis phagocytosis
  • the constant domains can form a "Fc region", which is the C-terminal region of a native antibody's heavy chain.
  • an antibody of the invention may be a native Fc region or a modified Fc region.
  • a Fc region is important for some antibodies' functions e.g. the activity of HERCEPTINTM is Fc-dependent.
  • the boundaries of the Fc region of a native antibody may vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226 or Pro230 to the heavy chain's C-terminus.
  • the Fc region will typically be able to bind one or more Fc receptors, such as a FcyRI (CD64), a FcyRII ⁇ e.g.
  • FcyRIIA, FcyRIIBl, FcyRIIB2, FcyRIIC a FcyRIII (e.g. FcyRIIIA, FcyRIIIB), a FcRn, FcaR (CD89), Fc5R, Fc ⁇ R, a FcsRI ⁇ e.g. Fc8RIa y 2 or FcsRIor ⁇ ), FcsRII ⁇ e.g. FcsRIIA or FcsRIIB), etc.
  • the Fc region may also or alternatively be able to bind to a complement protein, such as Clq. Modifications to an antibody's Fc region can be used to change its effector function(s) e.g.
  • reference 36 reports that effector functions may be modified by mutating Fc region residues 234, 235, 236, 237, 297, 318, 320 and/or 322.
  • reference 37 reports that effector functions of a human IgGl can be improved by mutating Fc region residues (EU Index Kabat numbering) 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434,
  • Fc residues 322, 329 and/or 331 Modification of Fc residues 322, 329 and/or 331 is reported in reference 38 for modifying Clq affinity of human IgG antibodies, and residues 270, 322, 326, 327, 329, 331, 333 and/or 334 are selected for modification in reference 39. Mapping of residues important for human IgG binding to FcRI, FcRII, FcRIII, and FcRn receptors is reported in reference 40, together with the design of variants with improved FcR-binding properties.
  • Whole C H domains can be substituted between isotypes e.g.
  • reference 41 discloses antibodies in which the C H 3 domain (and optionally the C H 2 domain) of human IgG4 is substituted by the C H 3 domain of human IgGl to provide suppressed aggregate formation.
  • Reference 41 also reports that mutation of arginine at position 409 (EU index Kabat) of human IgG4 to e.g. lysine shows suppressed aggregate formation. Mutation of the Fc region of available monoclonal antibodies to vary their effector functions is known e.g.
  • reference 42 reports mutation studies for RITUXANTM to change Clq-binding
  • reference 43 reports mutation studies for NUMAXTM to change FcR-binding, with mutation of residues 252, 254 and 256 giving a 10-fold increase in FcRn-binding without affecting antigen-binding.
  • Antibodies will typically be glycosylated. N-linked glycans attached to the C H 2 domain of a heavy chain, for instance, can influence Clq and FcR binding [40], with aglycosylated antibodies having lower affinity for these receptors.
  • the glycan structure can also affect activity e.g.
  • complement-mediated cell death differences in complement-mediated cell death may be seen depending on the number of galactose sugars (0, 1 or 2) at the terminus of a glycan's biantennary chain.
  • An antibody's glycans preferably do not lead to a human immunogenic response after administration.
  • Antibodies can be prepared in a form free from products with which they would naturally be associated. Contaminant components of an antibody's natural environment include materials such as enzymes, hormones, or other host cell proteins.
  • Useful antibodies have nanomolar or picomolar affinity constants for their target antigens ⁇ e.g. 10 "9 M, 10 "10 M, 10 "n M, 10 "12 M, 10 "13 M or tighter). Such affinities can be determined using conventional analytical techniques e.g. using surface plasmon resonance techniques as embodied in BIAcoreTM instrumentation and operated according to the manufacturer's instructions. Radioimmunoassay using radiolabeled target antigen (hemagglutinin) is another method by which binding affinity may be measured.
  • the monoclonal antibody used with the invention may be a human antibody, a humanized antibody, a chimeric antibody or (particularly for veterinary purposes) a non-human antibody.
  • the antibodies are human mAbs. These can be prepared by various means.
  • human B cells producing an antigen of interest can be immortalized e.g. by infection with Epstein Barr Virus (EBV), optionally in the presence of a polyclonal B cell activator [44 & 45].
  • EBV Epstein Barr Virus
  • Human monoclonal antibodies can also be produced in non-human hosts by replacing the host's own immune system with a functioning human immune system e.g. into Scid mice or Trimera mice.
  • Transgenic and transchromosomic mice have been successfully used for generating human monoclonal antibodies, including the "humab mouse” from Medarex and the "xeno-mouse” from Abgenix [46], collectively referred to herein as "human Ig mice”.
  • variable domains of these human antibodies are fully human (in particular the framework regions of the variable domains are fully human, in addition to the complementarity determining regions [CDRs]) and so will not elicit an immune response directed against the variable domain framework regions when administered to humans (except, potentially, for any anti-idiotypic response).
  • Human antibodies do not include any sequences that do not have a human origin.
  • the antibodies are humanised mAbs, CDR-grafted mAbs or chimeric mAbs.
  • Non-human monoclonal antibody DNA encoding the non-human heavy and light chain immunoglobulins can be obtained and engineered to contain human immunoglobulin sequences using standard molecular biology techniques.
  • the murine variable regions can be linked to human constant regions using methods known in the art.
  • the murine CDR regions can be inserted into a human framework [48-53].
  • one or more non-CDR variable framework residue(s) is also altered.
  • the HI, H2 and H3 CDRs may be transferred together into an acceptor VH domain, but it may also be adequate to transfer only one or two of them [51]. Similarly, one two or all three of the LI, L2 and L3 CDRs may be transferred into an acceptor VL domain.
  • Preferred antibodies will have 1, 2, 3, 4, 5 or all 6 of the donor CDRs. Where only one CDR is transferred, it will typically not be the L2 CDR, which is usually the shortest of the six.
  • the donor CDRs will all be from the same human antibody, but it is also possible to mix them e.g. to transfer the light chain CDRs from a first antibody and the heavy chain CDRs from a second antibody.
  • the antibodies are non-human mAbs. These can be prepared by various means e.g. the original Kohler & Milstein technique for preparing murine mAbs.
  • the antibody has a variable domain with an isoelectric point (pi) in the range of 5.0 to 8.0.
  • pi isoelectric point
  • a preferred antibody for use with the invention is an IgG2.
  • a preferred antibody for use with the invention is an anti-sclerostin antibody such as ANTIBODY 1 disclosed in reference 54 (the complete contents of which are incorporated herein by reference).
  • ANTIBODY 1 has a VH domain with amino acid SEQ ID NO: 1 and a VL domain with amino acid SEQ ID NO: 2.
  • Other anti-sclerostin antibodies useful with the present invention may include one or more (1, 2, 3, 4, 5 or 6) CDRs from ANTIBODY 1.
  • the CDRs in the heavy chain are SEQ ID NOs: 3, 4 & 5.
  • the CDRs in the light chain are SEQ ID NOs: 6, 7 & 8.
  • the ANTIBODY 1 heavy and light chains may be expressed as SEQ ID NOs: 9 and 10 to give a functional antibody.
  • composition “comprising” encompasses “including” as well as “consisting” e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X + Y.
  • the term “about” in relation to a numerical value x is optional and means, for example, x+10%.
  • ANTIBODY 1 recognises sclerostin and is disclosed in reference 54. It is a human IgGlk mAb obtained via phage display. ANTIBODY 1 heavy and light chains are shown in SEQ ID NOs: 9 and 10.
  • the following Examples provide improved gel formulations for anti-sclerositin antibodies, e.g., ANTIBODY 1, and methods for their preparation.
  • the gel formulation is prepared from an aqueous antibody formulation that has not previously been lyophilized, e.g., an aqueous formulation of ANTIBODY 1 drug substance (DS).
  • the gel formulation is prepared from an antibody lyophilisate (DP).
  • the novel formulations have the desirable property of rapidly gellifying following a pH change.
  • ANTIBODY 1 bulk drug substance is supplied at > 70 mg/mL in histidine buffer, pH 5.3 (used in Example 1, 3 and 4).
  • ANTIBODY 1 DS at a concentration of 135.94 mg/mL (in 6.4 mM Histidine buffer, pH 5.3) and 76.11 mg/mL (in 7.8 mM Histidine buffer, pH 5.3) were used in Example 5.
  • the preparation of 0.15 M Arginine-HCl solution was performed by adding 1.58g of Arginine HC1 solid powder and adjusting to 50 mL with water.
  • the preparation of 0.1 M stock solutions at different pH levels was performed for the following buffers: MOPS 3-(N-morpholino)propanesulfonic acid (pH 6.6, 7.0, 7.4), HEPES 2-[4-(2-hydroxyethyl)piperazin-l-yl]ethanesulfonic acid (pH 7.0, 7.4, 7.8) and TRIS 2- Amino-2-hydroxymethyl-propane-l,3-diol (pH 7.8, 8.2, 8.6).
  • MOPS 3-(N-morpholino)propanesulfonic acid pH 6.6, 7.0, 7.4
  • HEPES 2-[4-(2-hydroxyethyl)piperazin-l-yl]ethanesulfonic acid pH 7.0, 7.4, 7.8
  • a 0.1 M phosphate buffered saline (PBS) pH 7.4 solution was prepared by mixing 19 g of 0.2 M Na-dihydrogen phosphate, 81 g of 0.2 M Di-Na-Hydrogen phosphate and adjusting to 200 g with water.
  • a 0.1 M potassium phosphate buffer pH 7.4 solution was prepared by mixing 9.9 mL of 1 M potassium phosphate monobasic, 40.1 mL of 1 M potassium phosphate dibasic and adjusting to 500 g with water.
  • a 0.1 M potassium phosphate buffer pH 7.0 solution was prepared by mixing 7.7 mL of 1 M potassium phosphate monobasic, 12.3 mL of 1 M potassium phosphate dibasic and adjusting to 100 g with water.
  • a citrate phosphate buffer pH 7.0 solution was prepared by mixing 2.45 mL of 0.1 M citric acid and 22.55 mL 0.2 M sodium phosphate dibasic.
  • SEC Size exclusion chromatography
  • ANTIBODY 1 bulk drug substance was lyophilized to generate a lyophilisate containing 150 mg ANTIBODY 1, 30 mM histidine, 270 mM sucrose, 51 mM arginine, 0.06 % polysorbate 80 at pH 5.3.
  • the lyophilisate was reconstituted with 1 mL 0.1 M PBS pH 7.4. The gelling behavior and change in turbidity was monitored.
  • ANTIBODY 1 bulk drug substance was lyophilised to generate a lyophilisate containing 150 mg ANTIBODY 1, 30 mM histidine, 270 mM sucrose, 51 mM arginine, 0.06 % polysorbate 80 at pH 5.3.
  • the lyophilisate was reconstituted with 1 mL 0.1 M PBS pH 7.4 (Case #1); and mixtures of 0.1 M PBS pH 7.4 and 0.15 M Arginine-HCl solutions (Cases #2, #3 and #4 - see details in Table 1). The gelling behaviour and change in turbidity was monitored. Table 1 - Set-up of the reconstitution experiments.
  • PBS sodium phosphate buffer
  • the bioassay method determines the potency and specificity (identity) of a ANTIBODY 1 sample is in a fluorescence resonance energy transfer (FRET) assay that measures the capacity of ANTIBODY 1 to inhibit binding of recombinant human SOST to soluble recombinant human LRP6-IgG(Fc) chimera (LRP6(Fc)).
  • FRET fluorescence resonance energy transfer
  • the assay uses LRP6(Fc) labeled with a Eu3+ chelate as the donor fluorophore and SOST labeled with Cy5 as the acceptor fluorophore.
  • the Eu3+ chelate is excited at 340 nm and emits light at 615 nm.
  • ANTIBODY 1 competes with the binding of Cy5-SOST to Eu3+-LRP6(Fc) and thus reduces the fluorescence emission at 665 nm in a concentration-dependent manner.
  • the potency of a ANTIBODY 1 test sample is quantified by comparing its ability to inhibit binding of SOST to LRP6 to that of a reference standard.
  • ANTIBODY 1 lyophilizate as described in Example 1 was reconstituted with 1 mL of various buffers having different pH levels: MOPS (pH 6.6, 7.0, 7.4); HEPES (pH 7.0, 7.4, 7.8); and TRIS (pH 7.8, 8.2, 8.6).
  • MOPS pH 6.6, 7.0, 7.4
  • HEPES pH 7.0, 7.4, 7.8
  • TRIS pH 7.8, 8.2, 8.6
  • Potassium phosphate buffer, sodium phosphate buffer (PBS) and citrate phosphate buffer at pH 7.4 were used as a reference for gellification.
  • the time for the solution to gellify at room temperature was monitored visually and by turbidimetry. A visual check of the time for gellification of the solutions at 37°C was also performed.
  • ANTIBODY 1 DS at a concentration of 135.94 mg/ml (in 6.4 mM Histidine buffer, pH 5.3) and 76.11 mg/ml (in 7.8 mM Histidine buffer, pH 5.3) were diluted with 100 mM MOPS buffer pH 7.0 to a concentration of 100 mg/ml and 50 mg/ml, respectively. The time for gellification of the diluted DS solution was monitored visually.
  • Example 6 Example 6:
  • ANTIBODY 1 DS at a concentration of 73.0 mg/ml (in water) was diluted with different combinations of buffers plus excipients (either NaCl or Arginine-HCl) to a concentration of 15 mg/ml.
  • the set up of the different combinations tested is listed in the Table 2. The turbidity of these combinations along time was monitored at 25°C.
  • ANTIBODY 1 DS was diluted to a concentration of 15 mg/ml with different buffer/pH combinations. The viscosity and the zeta potential of these combinations were determined at 25°C. The theoretical ionic strength (IS) of the buffers was accurately adjusted to 15mM and 150mM using NaCl. The set up of the different combinations tested is listed in the Table 3.
  • the ANTIBODY 1 formulation reconstituted within 3 minutes at RT, resulting in a clear, colorless, liquid solution having a pH 6.6.
  • the turbidity of the reconstituted formulation 5 min after reconstitution was 325 NTU.
  • the solution turned into a solid gel structure with a turbidity of 1350 NTU.
  • the ANTIBODY 1 formulation reconstituted within 3 minutes at RT, resulting in a clear, colourless, liquid solution having a pH 6.6. After further incubation at 22.5 °C (room temperature), the solution turned into a solid gel structure. The turbidity of the reconstituted formulation increased very fast to the upper detection limit of 4000 NTU of the turbidimeter. When adding increasing concentrations of Arginine-HCl the turbidity increase was less pronounced as well as the gellification process ( Figure 1). Note that, for case #4 when 112.5 mM Arginine-HCl are present the turbidity reached a maximum of approximately 1000 NTU after incubation at RT for 21h (see Figure 1).
  • Example 4 The time to gellify at room temperature for lyophilisates reconstituted in various buffers is shown in Table 4.
  • ANTIBODY 1 antibody drug substance when diluted with MOPS buffer pH 7.0 (to a concentration of 50 mg/mL or 100 mg/mL) gellified rapidly within 1 minute at room temperature. This rapid gellification was independent of the concentration of the antibody formulation prior to dilution. However, the effect was more immediate for the 100 mg/mL sample (Figure 7).

Abstract

La présente invention concerne des formulations d'anticorps, et des procédés pour leur fabrication, qui (i) sont capables de conserver un anticorps dans un état natif et thérapeutiquement actif tout en produisant un effet de dépôt sans l'utilisation de polymères ou d'autres réactifs de formulation complexes et (ii) produisent une capacité de chargement très élevée, de manière à offrir la possibilité d'une dose augmentée par application au patient. Ces formulations prennent la forme d'un gel et sont basées sur l'observation surprenante que des anticorps monoclonaux ont une propriété de gélification dans des conditions appropriées.
PCT/EP2011/065096 2010-09-02 2011-09-01 Système de gel d'anticorps pour administration de médicament prolongée WO2012028683A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37952210P 2010-09-02 2010-09-02
US61/379,522 2010-09-02

Publications (1)

Publication Number Publication Date
WO2012028683A1 true WO2012028683A1 (fr) 2012-03-08

Family

ID=44534438

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/065096 WO2012028683A1 (fr) 2010-09-02 2011-09-01 Système de gel d'anticorps pour administration de médicament prolongée

Country Status (1)

Country Link
WO (1) WO2012028683A1 (fr)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8623367B2 (en) 2008-12-10 2014-01-07 Novartis Ag Antibody formulation
US8637643B2 (en) 2005-05-03 2014-01-28 Ucb Pharma, S.A. Sclerostin binding antibody
US8986685B2 (en) 1998-11-27 2015-03-24 Ucb Pharma S.A. Compositions and methods for increasing bone mineralization
US9089553B2 (en) 2005-05-03 2015-07-28 Amgen Inc. Method for inhibiting bone resorption
US9133272B2 (en) 2011-03-01 2015-09-15 Amgen Inc. Bispecific binding agents
US9145457B2 (en) 2011-03-25 2015-09-29 Amgen Inc. Sclerostin antibody crystals and formulations thereof
US9352043B2 (en) 2010-05-14 2016-05-31 Amgen Inc. High concentration antibody formulations
US9657090B2 (en) 2011-12-28 2017-05-23 Amgen Inc. Method of treating alveolar bone loss through the use of anti-sclerostin antibodies
US9822173B2 (en) 2012-11-21 2017-11-21 Amgen Inc. Heterodimeric immunoglobulins
US9925260B2 (en) 2012-07-05 2018-03-27 Ucb Pharma S.A. Treatment for bone diseases
WO2019020069A1 (fr) 2017-07-27 2019-01-31 江苏恒瑞医药股份有限公司 Composition pharmaceutique d'anticorps sost et utilisations de celle-ci
US10538584B2 (en) 2011-08-04 2020-01-21 Amgen Inc. Methods for treating bone gap defects
US11466079B2 (en) 2018-03-30 2022-10-11 Amgen Inc. C-terminal antibody variants
US11576970B2 (en) 2016-03-10 2023-02-14 UCB Biopharma SRL Pharmaceutical formulations
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
US11801300B2 (en) 2014-12-22 2023-10-31 Novartis Ag Pharmaceutical products and stable liquid compositions of IL-17 antibodies
US11851483B2 (en) 2014-12-12 2023-12-26 Amgen Inc. Anti-sclerostin antibodies and their use to treat bone disorders as part of a regimen

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992015331A1 (fr) 1991-03-08 1992-09-17 Mitsui Toatsu Chemicals, Inc. Preparation lyophilisee d'anticorps monoclonaux
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
US5591828A (en) 1989-06-22 1997-01-07 Behringwerke Aktiengesellschaft Bispecific and oligospecific mono-and oligovalent receptors, the preparation and use thereof
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
WO1998022136A2 (fr) 1996-11-19 1998-05-28 Roche Diagnostics Gmbh Preparations pharmaceutiques lyophilisees stables d'anticorps monoclonaux ou polyclonaux
WO2003009817A2 (fr) 2001-07-25 2003-02-06 Protein Design Labs, Inc. Formulation pharmaceutique lyophilisee stable d'anticorps igg
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6538124B1 (en) 1998-04-02 2003-03-25 Genentech, Inc. Polypeptide variants
WO2003041637A2 (fr) 2001-11-09 2003-05-22 Centocor, Inc. Compositions lyophilisees d'anticorps monoclonaux
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2004074464A1 (fr) 2003-02-21 2004-09-02 Odontis Limited Generation de dents a partir de cellules de moelle osseuse
WO2004076677A2 (fr) 2003-02-26 2004-09-10 Institute For Research In Biomedicine Production d'anticorps monoclonaux par transformation de lymphocytes b par le virus d'epstein barr
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2005048930A2 (fr) 2003-11-14 2005-06-02 Alza Corporation Gel a base de tensioactif en tant que support d'administration de medicament injectable a effet prolonge
WO2006033386A1 (fr) 2004-09-22 2006-03-30 Kirin Beer Kabushiki Kaisha Anticorps igg4 humains stabilises
WO2007074880A1 (fr) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Préparation stabilisatrice contenant des anticorps
WO2008029908A1 (fr) 2006-09-07 2008-03-13 Kyowa Hakko Kirin Co., Ltd. Préparation pharmaceutique lyophilisée stable comprenant un anticorps
WO2008071394A1 (fr) * 2006-12-11 2008-06-19 F. Hoffmann-La Roche Ag Formulation parentérale d'anticorps anti-peptide abêta
WO2008116103A2 (fr) 2007-03-22 2008-09-25 Imclone Llc Formulations d'anticorps stables
WO2009047356A1 (fr) 2007-10-12 2009-04-16 Novartis Ag Compositions et procédés pour l'utilisation d'anticorps contre la sclérostine
WO2009131553A2 (fr) 2006-12-29 2009-10-29 Osteogenex Inc. Procédés de modification de croissance osseuse par administration d'antagoniste ou d'agoniste sost ou wise
WO2010100179A2 (fr) * 2009-03-05 2010-09-10 Novartis Ag Système de gel autoformé pour administration de médicament à libération lente

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5591828A (en) 1989-06-22 1997-01-07 Behringwerke Aktiengesellschaft Bispecific and oligospecific mono-and oligovalent receptors, the preparation and use thereof
WO1992015331A1 (fr) 1991-03-08 1992-09-17 Mitsui Toatsu Chemicals, Inc. Preparation lyophilisee d'anticorps monoclonaux
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
WO1998022136A2 (fr) 1996-11-19 1998-05-28 Roche Diagnostics Gmbh Preparations pharmaceutiques lyophilisees stables d'anticorps monoclonaux ou polyclonaux
US20080286280A1 (en) 1996-11-19 2008-11-20 Roche Diagnostics Gmbh Stable Lyophilized Pharmaceutical Preparations of Monoclonal or polyclonal antibodies
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6538124B1 (en) 1998-04-02 2003-03-25 Genentech, Inc. Polypeptide variants
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2003009817A2 (fr) 2001-07-25 2003-02-06 Protein Design Labs, Inc. Formulation pharmaceutique lyophilisee stable d'anticorps igg
WO2003041637A2 (fr) 2001-11-09 2003-05-22 Centocor, Inc. Compositions lyophilisees d'anticorps monoclonaux
WO2004074464A1 (fr) 2003-02-21 2004-09-02 Odontis Limited Generation de dents a partir de cellules de moelle osseuse
WO2004076677A2 (fr) 2003-02-26 2004-09-10 Institute For Research In Biomedicine Production d'anticorps monoclonaux par transformation de lymphocytes b par le virus d'epstein barr
WO2005048930A2 (fr) 2003-11-14 2005-06-02 Alza Corporation Gel a base de tensioactif en tant que support d'administration de medicament injectable a effet prolonge
WO2006033386A1 (fr) 2004-09-22 2006-03-30 Kirin Beer Kabushiki Kaisha Anticorps igg4 humains stabilises
WO2007074880A1 (fr) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Préparation stabilisatrice contenant des anticorps
WO2008029908A1 (fr) 2006-09-07 2008-03-13 Kyowa Hakko Kirin Co., Ltd. Préparation pharmaceutique lyophilisée stable comprenant un anticorps
WO2008071394A1 (fr) * 2006-12-11 2008-06-19 F. Hoffmann-La Roche Ag Formulation parentérale d'anticorps anti-peptide abêta
WO2009131553A2 (fr) 2006-12-29 2009-10-29 Osteogenex Inc. Procédés de modification de croissance osseuse par administration d'antagoniste ou d'agoniste sost ou wise
WO2008116103A2 (fr) 2007-03-22 2008-09-25 Imclone Llc Formulations d'anticorps stables
WO2009047356A1 (fr) 2007-10-12 2009-04-16 Novartis Ag Compositions et procédés pour l'utilisation d'anticorps contre la sclérostine
WO2010100179A2 (fr) * 2009-03-05 2010-09-10 Novartis Ag Système de gel autoformé pour administration de médicament à libération lente

Non-Patent Citations (44)

* Cited by examiner, † Cited by third party
Title
ADAMS, SCHIER, JLMMUNOL METHODS., vol. 231, no. 1-2, 1999, pages 249 - 60
AUSUBEL ET AL.: "Short protocols in molecular biology", 2002, CURRENT PROTOCOLS
BIRDI, K. S.: "Handbook of Surface and Colloidal Chemistry", 1997, CRC PRESS
D.M. WEIR AND C.C. BLACKWELL,: "Handbook of Experimental Immunology", vol. I-IV, 1986, BLACKWELL SCIENTIFIC PUBLICATIONS
DALL'ACQUA ET AL., JBIOL CHEM, vol. 281, no. 33, 2006, pages 23514 - 24
DUMOULIN ET AL., PROTEIN SCI., vol. 11, no. 3, 2002, pages 500 - 15
EWERT ET AL., METHODS, vol. 34, no. 2, 2004, pages 184 - 99
GENNARO: "Remington: The Science and Practice ofPharmacy. 20th edition", 2000
GREEN, JIMMUNOL METHODS., vol. 231, no. 1-2, 1999, pages 11 - 23
GRUBER ET AL., JIMMUNOL, vol. 152, no. 11, 1994, pages 5368 - 74
HALLBORN, CARLSSON, BIOTECHNIQUES, 2002, pages 30 - 7
HARN N ET AL: "Highly concentrated monoclonal antibody solutions: Direct analysis of physical structure and thermal stability", JOURNAL OF PHARMACEUTICAL SCIENCES, AMERICAN PHARMACEUTICAL ASSOCIATION, WASHINGTON, US, vol. 96, no. 3, 1 March 2007 (2007-03-01), pages 532 - 546, XP002626716, ISSN: 0022-3549, [retrieved on 20061102], DOI: 10.1002/JPS.20753 *
HOLLINGER ET AL., PROC. NATL. ACAD SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOMAYOUN ET AL., J PHARMACY PHARMACOL, vol. 55, 2003, pages 933 - 8
HUDSON, KORTT, JIMMUNOL METHODS, vol. 231, 1999, pages 177 - 89
IDUSOGIE ET AL., J IMMUNOL, vol. 164, no. 8, 2000, pages 4178 - 84
IWAHASHI ET AL., MOL IMMUNOL., vol. 36, no. 15-16, 1999, pages 1079 - 91
KOTZ ET AL., EUR BIOCHEM., vol. 271, no. 9, 2004, pages 1623 - 9
KUO ET AL., NATURE BIOTECHNOL, vol. 16, 1998, pages 163 - 7
LIU JUN ET AL: "Reversible self-association increases the viscosity of a concentrated monoclonal antibody in aqueous solution", JOURNAL OF PHARMACEUTICAL SCIENCES, AMERICAN PHARMACEUTICAL ASSOCIATION, WASHINGTON, US, vol. 94, no. 9, 1 September 2005 (2005-09-01), pages 1928 - 1940, XP002578902, ISSN: 0022-3549, [retrieved on 20050728], DOI: 10.1002/JPS.20347 *
LO, METHODS MOL BIOL., vol. 248, 2004, pages 135 - 59
MANCINI ET AL., NEW MICROBIOL., vol. 27, no. 4, 2004, pages 315 - 28
MATHEUS ET AL., J PHARM SCI DOI 10.1002/JPS.21526, 2008
MOORE J M R ET AL: "Kinetics and Thermodynamics of Dimer Formation and Dissociation for a Recombinant Humanized Monoclonal Antibody to Vascular Endothelial Growth Factor", BIOCHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 38, no. 42, 28 September 1999 (1999-09-28), pages 13960 - 13967, XP007901441, ISSN: 0006-2960, DOI: 10.1021/BI9905516 *
MUYLDERMANS, JBIOTECHNOL, vol. 74, no. 4, 2001, pages 277 - 302
NEWTON & GRAHAM: "PCR (Introduction to Biotechniques Series), 2nd ed.", 1997, SPRINGER VERLAG
NING ET AL., RADIOTHER ONCOL, vol. 39, 1996, pages 179 - 89
NING S ET AL: "Intratumoral radioimmunotherapy of a human colon cancer xenograft using a sustained-release gel.", RADIOTHERAPY AND ONCOLOGY : JOURNAL OF THE EUROPEAN SOCIETY FOR THERAPEUTIC RADIOLOGY AND ONCOLOGY MAY 1996 LNKD- PUBMED:8735485, vol. 39, no. 2, May 1996 (1996-05-01), pages 179 - 189, XP002663929, ISSN: 0167-8140 *
O'BRIEN, JONES, METHODS MOL BIOL., vol. 207, 2003, pages 81 - 100
PINI & BRACCI, CURR PROTEIN PEPT SCI, vol. 1, no. 2, 2000, pages 155 - 69
POELSTRA ET AL., JBIOMEDMATER RES, vol. 60, 2002, pages 206 - 15
RADORNKSY ET AL., BIOL REPROD, vol. 47, 1992, pages 133 - 40
REAM ET AL.,: "Molecular Biology Techniques: An Intensive Laboratory Course", 1998, ACADEMIC PRESS
REY, MAY, FREEZE-DRYING/LYOPHILIZATION OF PHARMACEUTICAL & BIOLOGICAL PRODUCTS, 2004
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
S. COLOWICK AND N. KAPLAN: "Methods In Enzymology", ACADEMIC PRESS, INC.
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual, 3rd edition", 2001, COLD SPRING HARBOR LABORATORY PRESS
SHIELDS ET AL., J BIOL CHEM, vol. 276, 2001, pages 6591 - 604
SIDHU ET AL., JMOL BIOL., vol. 338, no. 2, 2004, pages 299 - 310
SUKUMAR ET AL., PHARM RES, vol. 21, no. 7, 2004, pages 1087 - 93
TRAGGIAI ET AL., NAT MED., vol. 10, no. 8, 2004, pages 871 - 5
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
WALTER ET AL., COMB CHEM HIGH THROUGHPUT SCREEN., vol. 4, no. 2, 2001, pages 193 - 205
WORN, PLUCKTHUN, J MOL BIOL., vol. 305, no. 5, 2001, pages 989 - 1010

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8986685B2 (en) 1998-11-27 2015-03-24 Ucb Pharma S.A. Compositions and methods for increasing bone mineralization
US9791462B2 (en) 1998-11-27 2017-10-17 Ucb Pharma, S.A. Compositions and methods for increasing bone mineralization
US9296812B2 (en) 2005-05-03 2016-03-29 Amgen Inc. Sclerostin binding antibodies
US9089553B2 (en) 2005-05-03 2015-07-28 Amgen Inc. Method for inhibiting bone resorption
US11939372B2 (en) 2005-05-03 2024-03-26 Amgen Inc. Binding agents
US8637643B2 (en) 2005-05-03 2014-01-28 Ucb Pharma, S.A. Sclerostin binding antibody
US10562964B2 (en) 2005-05-03 2020-02-18 Amgen Inc. Methods for isolating antibodies that bind sclerostin
US10273293B2 (en) 2005-05-03 2019-04-30 Amgen Inc. Method for inhibiting bone resorption
US11091537B2 (en) 2007-09-17 2021-08-17 Amgen Inc. Method for inhibiting bone resorption
US8623367B2 (en) 2008-12-10 2014-01-07 Novartis Ag Antibody formulation
US10064946B2 (en) 2010-05-14 2018-09-04 Amgen Inc. High concentration antibody formulations
US9352043B2 (en) 2010-05-14 2016-05-31 Amgen Inc. High concentration antibody formulations
US11040102B2 (en) 2010-05-14 2021-06-22 Amgen Inc. High concentration antibody formulations
US9133272B2 (en) 2011-03-01 2015-09-15 Amgen Inc. Bispecific binding agents
US9617333B2 (en) 2011-03-25 2017-04-11 Amgen Inc. Sclerostin antibody crystals and formulations thereof
US9145457B2 (en) 2011-03-25 2015-09-29 Amgen Inc. Sclerostin antibody crystals and formulations thereof
US9920114B2 (en) 2011-03-25 2018-03-20 Amgen Inc. Antibody formulations
US10538584B2 (en) 2011-08-04 2020-01-21 Amgen Inc. Methods for treating bone gap defects
US9657090B2 (en) 2011-12-28 2017-05-23 Amgen Inc. Method of treating alveolar bone loss through the use of anti-sclerostin antibodies
US9913900B2 (en) 2011-12-28 2018-03-13 Amgen Inc. Method of treating alvelor bone loss through the use of anti-sclerostin antibodies
US11896667B2 (en) 2012-07-05 2024-02-13 Ucb Pharma S.A. Treatment for bone diseases
US9925260B2 (en) 2012-07-05 2018-03-27 Ucb Pharma S.A. Treatment for bone diseases
US10799583B2 (en) 2012-07-05 2020-10-13 Ucb Pharma, S.A. Treatment for bone diseases
US11466078B2 (en) 2012-11-21 2022-10-11 Amgen Inc. Heterodimeric immunoglobulins
US9822173B2 (en) 2012-11-21 2017-11-21 Amgen Inc. Heterodimeric immunoglobulins
US10233237B2 (en) 2012-11-21 2019-03-19 Amgen Inc. Heterodimeric immunoglobulins
US11851483B2 (en) 2014-12-12 2023-12-26 Amgen Inc. Anti-sclerostin antibodies and their use to treat bone disorders as part of a regimen
US11801300B2 (en) 2014-12-22 2023-10-31 Novartis Ag Pharmaceutical products and stable liquid compositions of IL-17 antibodies
US11576970B2 (en) 2016-03-10 2023-02-14 UCB Biopharma SRL Pharmaceutical formulations
EP3659586A4 (fr) * 2017-07-27 2020-08-12 Jiangsu Hengrui Medicine Co., Ltd. Composition pharmaceutique d'anticorps sost et utilisations de celle-ci
US11498961B2 (en) 2017-07-27 2022-11-15 Jiangsu Hengrui Medicine Co., Ltd. SOST antibody pharmaceutical composition and uses thereof
WO2019020069A1 (fr) 2017-07-27 2019-01-31 江苏恒瑞医药股份有限公司 Composition pharmaceutique d'anticorps sost et utilisations de celle-ci
US11466079B2 (en) 2018-03-30 2022-10-11 Amgen Inc. C-terminal antibody variants
US11858983B2 (en) 2018-03-30 2024-01-02 Amgen Inc. C-terminal anti-sclerostin antibody variants
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation

Similar Documents

Publication Publication Date Title
WO2012028683A1 (fr) Système de gel d'anticorps pour administration de médicament prolongée
WO2010100179A2 (fr) Système de gel autoformé pour administration de médicament à libération lente
US20100226928A1 (en) Lyophilised Antibody Formulation
ES2881642T3 (es) Anticuerpos anti-pro-miostatina/miostatina latente y usos de los mismos
US20230399388A1 (en) Methods for treating inflammation using antibodies to kallidin and des-arg10-kallidin
JP6845012B2 (ja) 遺伝性血管浮腫の治療における血漿カリクレイン結合タンパク質およびその使用
US8999343B2 (en) Antibodies that bind myostatin, compositions and methods
US20180155444A1 (en) Antibodies to Plasminogen Activator Inhibitor-1 (PAI-1) and Uses Thereof
JP7237848B2 (ja) 免疫原性の低下のための方法及び組成物
TW201200152A (en) Novel antibody formulation
CA2920890A1 (fr) Anticorps diriges contre l'inhibiteur des activateurs du plasminogene de type 1 (pai-1) et leurs utilisations
US11427632B2 (en) Antibodies with low immunogenicity and uses thereof
KR20180094028A (ko) 혈장 칼리크레인 저해제 및 유전성 혈관부종 발작을 치료하기 위한 이의 용도
AU2015228454A1 (en) Anti-MCAM antibodies and associated methods of use
CA3110689A1 (fr) Inhibiteurs de la kallicreine plasmatique et leurs utilisations pour traiter une crise d'angio-ƒdeme hereditaire
CA2993329A1 (fr) Compose ciblant le facteur d'activation d'il-23 et des lymphocytes b (baff) et utilisations associees
CN108602881A (zh) 抗-因子d抗体制剂
CN111201036A (zh) 包含抗α(V)β(6)抗体的药物组合物和剂量方案
US20230406942A1 (en) Igf1r antibodies
EP4339207A1 (fr) Molécule de liaison à l'antigène se liant spécifiquement à rankl et ngf, et son utilisation médicale
WO2023241389A1 (fr) Anticorps monoclonal contre tfpi et son utilisation
KR20220044286A (ko) 항 pd-1/her2 이중특이성 항체를 포함하는 제제, 이의 제조 방법 및 용도
Li et al. Antibodies to kallidin and des-Arg 9-kallidin
CN117586390A (zh) 抗cgrp抗体及用途

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: 11749851

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: 11749851

Country of ref document: EP

Kind code of ref document: A1