WO2012168491A1 - Formulations pharmaceutiques d'antagonistes de pcsk9 - Google Patents

Formulations pharmaceutiques d'antagonistes de pcsk9 Download PDF

Info

Publication number
WO2012168491A1
WO2012168491A1 PCT/EP2012/061045 EP2012061045W WO2012168491A1 WO 2012168491 A1 WO2012168491 A1 WO 2012168491A1 EP 2012061045 W EP2012061045 W EP 2012061045W WO 2012168491 A1 WO2012168491 A1 WO 2012168491A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
formulation
seq
amino acid
pcsk9
Prior art date
Application number
PCT/EP2012/061045
Other languages
English (en)
Inventor
Marion GIFFARD
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 WO2012168491A1 publication Critical patent/WO2012168491A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates of pharmaceutical formulations comprising antibody antagonists against PCSK9 for therapeutic use.
  • LDL-R low-density lipoprotein receptor
  • LDL-R low-density lipoprotein receptor
  • PCSK9 proprotein convertase subtilisin/kexin type 9a
  • Antibodies, as other protein therapeutics are complex molecules and in general, large amounts of antibodies have to be used in pharmaceutical formulations due to their therapeutically effective dose in mammals, particularly humans.
  • Liquid formulations of protein therapeutics should preserve intact the biologic activity of the protein therapeutics and protect the functional groups of the protein therapeutics from degradation during
  • liquid pharmaceutical formulations of protein therapeutics i.e. antibodies should be long-term stable, contain a safe and effective amount of the pharmaceutical compound.
  • a long appreciated problem with liquid formulations of protein therapeutics is that of aggregation, where protein molecules physically stick together, for example, resulting in the formation of opaque insoluble matter or precipitation, which may show undesired immunological reactions.
  • a major problem caused by the aggregate formation is that during the administration the formulation may block syringes or pumps and rendering it unsafe to patients.
  • the present invention addresses the above-identified need by providing a novel formulation comprising an antibody, free of protein aggregates, stable and having sufficiently low viscosity and which is therefore suitable for administration to mammalians, particularly human subjects.
  • a pharmaceutical formulation comprising an antibody that binds to proprotein convertase subtilisin/kexin type 9 (PCSK9), wherein the antibody:
  • a) does not block PCSK9 binding to the low density lipoprotein receptor (LDLR) and b) inhibits PCSK9-mediated degradation of LDLR.
  • LDLR low density lipoprotein receptor
  • the invention provides a stable, highly concentrated liquid pharmaceutical composition comprising at least about 10 mg/mL of said antibody.
  • the invention provides a lyophilized pharmaceutical composition which comprises at least about 10 mg/ml of said antibody after reconstitution in solution.
  • a formulation according to invention for administration to an individual having hypercholesterolemia or a high baseline LDL-C level.
  • the invention is directed to a dosage form comprising: a liquid or lyophilized pharmaceutical composition comprising: a) least about 10 mg/mL of said antibody and b) at least one pharmaceutically acceptable excipient.
  • the present invention provides a method of producing a lyophilized pharmaceutical composition comprising at least about 10 mg/mL of said antibody post reconstitution, by contacting a) said antibody with b) at least one pharmaceutically acceptable excipient.
  • Another aspect of the present invention relates to a method for producing said liquid or lyophilized formulation.
  • an "antibody” refers to a polypeptide of the immunoglobulin family or a polypeptide comprising fragments of an immunoglobulin that is capable of noncovalently, reversibly, and in a specific manner binding a corresponding antigen.
  • An exemplary antibody structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light” (about 25 kD) and one "heavy" chain (about 50-70 kD), connected through a disulfide bond.
  • immunoglobulin genes include the ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ constant region genes, as well as the myriad immunoglobulin variable region genes.
  • Light chains are classified as either ⁇ or ⁇ .
  • Heavy chains are classified as ⁇ , ⁇ , ⁇ , ⁇ , or ⁇ , which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively.
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms variable light chain (V L ) and variable heavy chain (V H ) refer to these regions of light and heavy chains respectively.
  • an “antibody” encompasses all variations of antibody and fragments thereof that possess a particular binding specifically, e.g., for PCSK9.
  • an “antibody” encompasses all variations of antibody and fragments thereof that possess a particular binding specifically, e.g., for PCSK9.
  • full length antibodies chimeric antibodies, humanized antibodies, single chain antibodies (ScFv), Fab, Fab', and multimeric versions of these fragments (e.g., F(ab') 2 ) with the same binding specificity.
  • CDRs complementary metal-oxide-semiconductor domains
  • VL and VH the hypervariable regions of VL and VH.
  • the CDRs are the target protein-binding site of the antibody chains that harbors specificity for such target protein.
  • CDRl-3 the target protein-binding site of the antibody chains that harbors specificity for such target protein.
  • CDRl-3 the target protein-binding site of the antibody chains that harbors specificity for such target protein.
  • CDRl-3 three CDRs (CDRl-3, numbered sequentially from the N-terminus) in each human VL or VH, constituting about 15-20% of the variable domains.
  • the CDRs are structurally complementary to the epitope of the target protein and are thus directly responsible for the binding specificity.
  • the remaining stretches of the V L or V H the so-called framework regions, exhibit less variation in amino acid sequence (Kuby, Immunology, 4th ed., Chapter 4. W.H. Freeman & Co., New York, 2000
  • the positions of the CDRs and framework regions are determined using various well known definitions in the art, e.g., Kabat, Chothia, international ImMunoGeneTics database (IMGT) (on the worldwide web at imgt.cines.fr/), and AbM (see, e.g., Johnson et al, Nucleic Acids Res., 29:205-206 (2001); Chothia and Lesk, J. Mol. Biol., 196:901-917 (1987); Chothia et al, Nature, 342:877-883 (1989); Chothia et al, J. Mol.
  • IMGT international ImMunoGeneTics database
  • binding specificity determinant or “BSD” interchangeably refer to the minimum contiguous or non-contiguous amino acid sequence within a complementarity determining region necessary for determining the binding specificity of an antibody.
  • a minimum binding specificity determinant can be within one or more CDR sequences. In some embodiments, the minimum binding specificity determinants reside within (i.e., are determined solely by) a portion or the full-length of the CDR3 sequences of the heavy and light chains of the antibody.
  • an "antibody light chain” or an “antibody heavy chain” as used herein refers to a polypeptide comprising the V L or V H , respectively.
  • the endogenous V L is encoded by the gene segments V (variable) and J (junctional), and the endogenous VH by V, D (diversity), and J.
  • Each of V L or V H includes the CDRs as well as the framework regions.
  • antibody light chains and/or antibody heavy chains may, from time to time, be collectively referred to as "antibody chains.” These terms encompass antibody chains containing mutations that do not disrupt the basic structure of VL or VH, as one skilled in the art will readily recognize.
  • Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)' 2 , a dimer of Fab' which itself is a light chain joined to V H -C H 1 by a disulfide bond.
  • the F(ab)' 2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)' 2 dimer into an Fab' monomer.
  • the Fab' monomer is essentially Fab with part of the hinge region. Paul, Fundamental Immunology 3d ed. (1993).
  • antibody While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology.
  • antibody also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies ⁇ e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al, Nature 348:552-554 (1990)).
  • phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind to selected antigens (see, e.g., McCafferty et al, supra; Marks et al, Biotechnology, 10:779-783, (1992)).
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as import residues, which are typically taken from an import variable domain. Humanization can be essentially performed following the method of Winter and co-workers ⁇ see, e.g., Jones et al, Nature 321 :522-525 (1986); Riechmann et al, Nature 332:323-327 (1988); Verhoeyen et al, Science 239:1534-1536 (1988) and Presta, Curr. Op. Struct. Biol.
  • humanized antibodies are chimeric antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some complementarity determining region ("CDR") residues and possibly some framework (“FR”) residues are substituted by residues from analogous sites in rodent antibodies.
  • a "chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, and drug; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • Antibodies or antigen-binding molecules of the invention further includes one or more immunoglobulin chains that are chemically conjugated to, or expressed as, fusion proteins with other proteins. It also includes bispecific antibody.
  • a bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Other antigen-binding fragments or antibody portions of the invention include bivalent scFv (diabody), bispecific scFv antibodies where the antibody molecule recognizes two different epitopes, single binding domains (dAbs), and minibodies.
  • the various antibodies or antigen-binding fragments described herein can be produced by enzymatic or chemical modification of the intact antibodies, or synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv), or identified using phage display libraries (see, e.g., McCafferty et al., Nature 348:552-554, 1990).
  • minibodies can be generated using methods described in the art, e.g., Vaughan and Sollazzo, Comb Chem High Throughput Screen. 4:417-30 2001.
  • Bispecific antibodies can be produced by a variety of methods including fusion Of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin.
  • Single chain antibodies can be identified using phage display libraries or ribosome display libraries, gene shuffled libraries. Such libraries can be constructed from synthetic, semi-synthetic or native and immunocompetent sources.
  • a "chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • a mouse anti-PC SK9 antibody can be modified by replacing its constant region with the constant region from a human immunoglobulin. Due to the replacement with a human constant region, the chimeric antibody can retain its specificity in recognizing human PCSK9 while having reduced antigenicity in human as compared to the original mouse antibody.
  • antibody binding molecule or “non-antibody ligand” refers to antibody mimics that use non-immunoglobulin protein scaffolds, including adnectins, avimers, single chain polypeptide binding molecules, and antibody-like binding peptidomimetics.
  • variable region or "V-region” interchangeably refer to a heavy or light chain comprising FR1 -CDR 1 -FR2-CDR2-FR3 -CDR3 -FR4.
  • An endogenous variable region is encoded by immunoglobulin heavy chain V-D-J genes or light chain V-J genes.
  • a V- region can be naturally occurring, recombinant or synthetic.
  • variable segment or “V-segment” interchangeably refer to a subsequence of the variable region including FR1-CDR1-FR2-CDR2-FR3.
  • An endogenous V-segment is encoded by an immunoglobulin V-gene.
  • a V-segment can be naturally occurring, recombinant or synthetic.
  • J-segment refers to a subsequence of the variable region encoded comprising a C-terminal portion of a CDR3 and the FR4.
  • An endogenous J-segment ' is encoded by an immunoglobulin J-gene.
  • a J-segment can be naturally occurring, recombinant or synthetic.
  • a "humanized” antibody is an antibody that retains the reactivity of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody with their human counterparts. See, e.g., Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851- 6855 (1984); Morrison and Oi, Adv. Immunol, 44:65-92 (1988); Verhoeyen et al, Science, 239:1534-1536 (1988); Padlan, Molec. Immun., 28:489-498 (1991); Padlan, Molec. Immun., 31(3):169-217 (1994).
  • corresponding human germline sequence refers to the nucleic acid sequence encoding a human variable region amino acid sequence or subsequence that shares the highest determined amino acid sequence identity with a reference variable region amino acid sequence or subsequence in comparison to all other all other known variable region amino acid sequences encoded by human germline immunoglobulin variable region sequences.
  • the corresponding human germline sequence can also refer to the human variable region amino acid sequence or subsequence with the highest amino acid sequence identity with a reference variable region amino acid sequence or subsequence in comparison to all other evaluated variable region amino acid sequences.
  • the corresponding human germline sequence can be framework regions only, complementarity determining regions only, framework and complementarity determining regions, a variable segment (as defined above), or other combinations of sequences or subsequences that comprise a variable region.
  • Sequence identity can be determined using the methods described herein, for example, aligning two sequences using BLAST, ALIGN, or another alignment algorithm known in the art.
  • the corresponding human germline nucleic acid or amino acid sequence can have at least about 90%, 92%, 94%, 96%, 98%, 99% sequence identity with the reference variable region nucleic acid or amino acid sequence.
  • Corresponding human germline sequences can be determined, for example, through the publicly available international ImMunoGeneTics database (IMGT) (on the worldwide web at imgt.cines.fr/) and V-base (on the worldwide web at vbase.mrc-cpe.cam.ac.uk).
  • IMGT international ImMunoGeneTics database
  • a biological sample e.g., a blood, serum, plasma or tissue sample.
  • Specific binding to an antibody or binding agent under such conditions may require the antibody or agent to have been selected for its specificity for a particular protein. As desired or appropriate, this selection may be achieved by subtracting out antibodies that cross-react with, e.g., PCSK9 molecules from other species (e.g., mouse) or other PCSK subtypes.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select antibodies specifically immunoreactive with a protein (see, e.g., Harlow & Lane, Using Antibodies, A Laboratory Manual (1998), for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity).
  • a specific or selective binding reaction will produce a signal at least twice over the background signal and more typically at least than 10 to 100 times over the background.
  • Equilibrium dissociation constant (K D , M) refers to the dissociation rate constant (kd, time “1 ) divided by the association rate constant (k a , time "1 , M “1 ). Equilibrium dissociation constants can be measured using any known method in the art.
  • the antibodies of the present invention generally will have an equilibrium dissociation constant of less than about 10 "7 or 10 "8 M, for example, less than about 10 "9 M or 10 "10 M, in some embodiments, less than about 10 "11 M, 10 "12 M or 10 "13 M.
  • the term "antigen-binding region” refers to a domain of the PCSK9- binding molecule of this invention that is responsible for the specific binding between the molecule and PCSK9.
  • An antigen-binding region includes at least one antibody heavy chain variable region and at least one antibody light chain variable region. There are at least one such antigen-binding regions present in each PCSK9-binding molecule of this invention, and each of the antigen-binding regions may be identical or different from the others. In some embodiments, at least one of the antigen-binding regions of a PCSK9-binding molecule of this invention acts as an antagonist of PCSK9.
  • an antagonist refers to an agent that is capable of specifically binding and inhibiting the activity of the target molecule.
  • an antagonist of PCSK9 specifically binds to PCSK9 and fully or partially inhibits PCSK9- mediated degradation of the LDLR. Inhibiting PCSK9-mediated degradation of the LDLR may or may not interfere with PCSK9 binding to the LDLR.
  • a PCSK9 antagonist can be identified by its ability to bind to PCSK9 and inhibit binding of PCSK9 to the LDLR.
  • Inhibition occurs when PCSK9-mediated degradation of the LDLR, when exposed to an antagonist of the invention, is at least about 10% less, for example, at least about 25%, 50%, 75% less, or totally inhibited, in comparison to PCSK9-mediated degradation in the presence of a control or in the absence of the antagonist.
  • a control can be exposed to no antibody or antigen binding molecule, an antibody or antigen binding molecule that specifically binds to another antigen, or an anti-PCSK9 antibody or antigen binding molecule known not to function as an antagonist.
  • An "antibody antagonist" refers to the situation where the antagonist is an inhibiting antibody.
  • PCSK9 or "proprotein convertase subtilisin/kexin type 9a"
  • PCSK9 is synthesized as a soluble zymogen that undergoes autocatalytic intramolecular processing in the endoplasmic reticulum, and is thought to function as a proprotein convertase.
  • PCSK9 plays a role in cholesterol homeostasis and may have a role in the differentiation of cortical neurons.
  • PCSK9 has been associated with a form of autosomal dominant familial hypercholesterolemia. See, e.g., Burnett and Hooper, Clin Biochem Rev (2008) 29(1):11-26.
  • the nucleic acid and amino acid sequences of PCSK9 are known, and have been published in GenBank Accession Nos. NM l 74936.2 and NP 777596.2, respectively.
  • a PCSK9 polypeptide functionally binds to LDLR and promotes the degradation of LDLR.
  • a PCSK9 amino acid sequence has at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with the amino acid sequence of GenBank accession no. NP 777596.2.
  • a PCSK9 nucleic acid sequence has at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with the nucleic acid sequence of GenBank accession no.
  • PCSK9 gain-of-function mutation refers to natural mutations occurring in PCSK9 genes that are associated with and/or causative of the familial hypercholesterolemia phenotype, accelerated atherosclerosis and premature coronary heart disease, e.g., due to enhanced LDLR degradation and a reduction of LDLR levels.
  • the allele frequency of PCSK9 gain-of-function mutations is rare. See, Burnett and Hooper, Clin Biochem Rev. (2008) 29(1):11-26.
  • Exemplary PCSK9 gain-of-function mutations include D129N, D374H, N425S and R496W.
  • Activity of a polypeptide of the invention refers to structural, regulatory, or biochemical functions of a polypeptide in its native cell or tissue.
  • Examples of activity of a polypeptide include both direct activities and indirect activities.
  • Exemplary direct activities of PCSK9 are the result of direct interaction with the polypeptide, including binding to LDLR and PCSK9-mediated degradation of LDLR.
  • Exemplary indirect activities in the context of PCSK9 are observed as a change in phenotype or response in a cell, tissue, organ or subject to a polypeptide's directed activity, e.g., reducing increased liver LDLR, reduced plasma HDL-C, decreased plasma cholesterol, enhances sensitivity to statins.
  • nucleic acid or protein when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It is preferably in a homogeneous state. It can be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. In particular, an isolated gene is separated from open reading frames that flank the gene and encode a protein other than the gene of interest. The term "purified" denotes that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. Particularly, it means that the nucleic acid or protein is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure.
  • nucleic acid refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
  • DNA deoxyribonucleic acids
  • RNA ribonucleic acids
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed- base and/or deoxyinosine residues (Batzer et ah, Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al, J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al, Mol. Cell. Probes 8:91-98 (1994)).
  • 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 polymer.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, ⁇ - carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refer to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a-carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Constantly modified variants applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
  • the following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (e.g., a polypeptide of the invention), which does not comprise additions or deletions, for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same sequences.
  • Two sequences are “substantially identical” if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity over a specified region, or, when not specified, over the entire sequence of a reference sequence), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
  • the invention provides polypeptides or polynucleotides that are substantially identical to the polypeptides or polynucleotides, respectively, exemplified herein (e.g., the variable regions exemplified in any one of SEQ ID NOS:l-5, 15-19 and 40- 41; the variable segments exemplified in any one of SEQ ID NOS:27-31; the CDRs exemplified in any one of SEQ ID NOS:6-14, 20-26; the FRs exemplified in any one of SEQ ID NOs: 32-39; and the nucleic acid sequences exemplified in any on of SEQ ID NOS:42- 45).
  • the variable regions exemplified in any one of SEQ ID NOS:l-5, 15-19 and 40- 41 the variable segments exemplified in any one of SEQ ID NOS:27-31
  • the identity exists over a region that is at least about 15, 25 or 50 nucleotides in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides in length, or over the full length of the reference sequence.
  • identity or substantial identity can exist over a region that is at least 5, 10, 15 or 20 amino acids in length, optionally at least about 25, 30, 35, 40, 50, 75 or 100 amino acids in length, optionally at least about 1 0, 200 or 250 amino acids in length, or over the full length of the reference sequence.
  • shorter amino acid sequences e.g., amino acid sequences of 20 or fewer amino acids
  • substantial identity exists when one or two amino acid residues are conservatively substituted, according to the conservative substitutions defined herein.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well known in the art.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.
  • BLAST and BLAST 2.0 algorithms Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nuc. Acids Res. 25:3389-3402, and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al, supra).
  • initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci.
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873- 5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
  • nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below.
  • a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions.
  • Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below.
  • Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.
  • link when used in the context of describing how the antigen-binding regions are connected within a PCSK9-binding molecule of this invention, encompasses all possible means for physically joining the regions.
  • the multitude of antigen-binding regions are frequently joined by chemical bonds such as a covalent bond (e.g., a peptide bond or a disulfide bond) or a non-covalent bond, which can be either a direct bond (i.e., without a linker between two antigen-binding regions) or indirect bond (i.e., with the aid of at least one linker molecule between two or more antigen-binding regions).
  • the terms "subject,” “patient,” and “individual” interchangeably refer to a mammal, for example, a human or a non-human primate mammal.
  • the mammal can also be a laboratory mammal, e.g., mouse, rat, rabbit, hamster.
  • the mammal can be an agricultural mammal (e.g., equine, ovine, bovine, porcine, camelid) or domestic mammal (e.g., canine, feline).
  • a therapeutically acceptable amount or “therapeutically effective dose” interchangeably refer to an amount sufficient to effect the desired result (i.e., a reduction in plasma non-HDL-C, hypercholesterolemia, atherosclerosis, coronary heart disease). In some embodiments, a therapeutically acceptable amount does not induce or cause undesirable side effects.
  • a therapeutically acceptable amount can be determined by first administering a low dose, and then incrementally increasing that dose until the desired effect is achieved.
  • a “prophylactically effective dosage,” and a “therapeutically effective dosage,” of a PCSK9 antagonizing antibody of the invention can prevent the onset of, or result in a decrease in severity of, respectively, disease symptoms associated with the presence of PCSK9 (e.g., hypercholesterolemia).
  • a “prophylactically effective dosage,” and a “therapeutically effective dosage,” can also prevent or ameliorate, respectively, impairment or disability due to the disorders and diseases resulting from activity of PCSK9.
  • co-administer refers to the simultaneous presence of two active agents in the blood of an individual. Active agents that are co-administered can be concurrently or sequentially delivered.
  • the phrase “consisting essentially of” refers to the genera or species of active pharmaceutical agents included in a method or composition, as well as any excipients inactive for the intended purpose of the methods or compositions. In some embodiments, the phrase “consisting essentially of expressly excludes the inclusion of one or more additional active agents other than an antagonist anti PCSK9 antibody of the invention. In some embodiments, the phrase “consisting essentially of expressly excludes the inclusion of one or more additional active agents other than an antagonist anti PCSK9 antibody of the invention and a second co-administered agent. [0054]
  • the term “statin” refers to a class of pharmacological agents that are competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase.
  • Figure 1 shows thawing and pooling of drug substance bottles.
  • Figure 2 shows the manufacture of 2% Polysorbate 20.
  • Figure 3 shows the manufacture of dilution solution.
  • Figure 4 shows the manufacture of drug product.
  • Figure 5 illustrates the heavy (SEQ ID NO:2) and light (SEQ ID NO: 16) chain amino acid sequences of parent mouse monoclonal antibody NVP-LGT209.
  • the sequences of CDR1 , CDR2 and CDR3 are underlined and in bold.
  • formulations of therapeutic antibodies that bind to and antagonize the function of proprotein convertase subtilisin/kexin type 9 (PCSK9) (e.g., SEQ ID NO:47).
  • PCSK9 proprotein convertase subtilisin/kexin type 9
  • antibody formulation is provided which is stable upon storage and delivery.
  • a stable formulation is a formulation wherein the antibody therein essentially retains its physical and chemical stability and integrity upon storage.
  • the stability of the antibody formulation may be measured using biological activity assays and wherein the biological activity upon storage is of about 80-125% of the original activity.
  • compositions comprising the present anti- PCSK9 antibodies or antigen-binding molecules formulated together with a pharmaceutically acceptable carrier.
  • the compositions can additionally contain other therapeutic agents that are suitable for treating or preventing a given disorder.
  • Pharmaceutically carriers enhance or stabilize the composition, or to facilitate preparation of the composition.
  • Pharmaceutically acceptable carriers include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • a pharmaceutical composition of the present invention can be administered by a variety of methods known in the art.
  • the route and/or mode of administration vary depending upon the desired results. It is preferred that administration be intravenous, intramuscular, intraperitoneal, or subcutaneous, or administered proximal to the site of the target.
  • the pharmaceutically acceptable carrier should be suitable for intravenous, intramuscular, subcutaneous, parenteral, intranasal, inhalational, spinal or epidermal administration (e.g., by injection or infusion).
  • the active compound i.e., antibody, bispecific and multispecific molecule, may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • the antibodies can be made into aerosol formulations (i.e., they can be "nebulized") to be administered via inhalation.
  • Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • the composition is sterile and fluid. Proper fluidity can be maintained, for example, by use of coating such as lecithin, by maintenance of required particle size in the case of dispersion and by use of surfactants.
  • isotonic agents for example, sugars, polyalcohols such as mannitol or sorbitol, and sodium chloride in the composition. Long-term absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • compositions are preferably manufactured under GMP conditions.
  • a therapeutically effective dose or efficacious dose of the anti-PCSK9 antibody is employed in the pharmaceutical compositions of the invention.
  • the anti-PCSK9 antibodies are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Dosage regimens are adjusted to provide the desired response (e.g., a therapeutic response).
  • a therapeutically or prophylactically effective dose a low dose can be administered and then incrementally increased until a desired response is achieved with minimal or no undesired side effects. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level depends upon a variety of pharmacokinetic factors including the activity of the particular
  • compositions of the present invention employed, or the ester, salt or amide thereof the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors.
  • composition comprising an antibody that binds to proprotein convertase subtilisin/kexin type 9 (PCSK9), wherein the antibody:
  • a) does not block PCSK9 binding to the low density lipoprotein receptor (LDLR) and b) inhibits PCSK9-mediated degradation of LDLR, and wherein said antibody is present in amount higher than 10 mg/ml.
  • the amount of antibody present in the liquid formulation suitable for administration to a patient can be 20-300 mg/ml, 30-200 mg/ml, 90-200 mg/ml, or 90-180 mg/ml. In one embodiment the antibody concentration is between 100 and 170 mg/ml, or 150, 160, 170, 180, 190, 200, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 mg/ ml.
  • the antibody or antigen binding molecule binds to at least one amino acid within residue positions 680-692 of human PCSK9.
  • the antibody or antigen binding molecule binds to an epitope of PCSK9 within the amino acid sequence RSRHLAQASQELQ (SEQ ID NO:49).
  • the antibody or antigen binding molecule binds to human PCSK9 with an equilibrium dissociation constant (KD) of about 500 pM or less.
  • KD equilibrium dissociation constant
  • the antibody or antigen binding molecule binds to human PCSK9 with an equilibrium dissociation constant (KD) of about 400 pM, 300 pM, 250 pM, 200 pM, 190 pM, 180 pM, 170 pM, 160 pM, 150 pM, 140 pM, or less.
  • the antibody or antigen binding molecule has an in vivo half- life of at least about 7 days. In some embodiments the antibody or antigen binding molecule has an in vivo half-life of at least about 3, 4, 5, 6, 7, 8, 9, 10 days. In some embodiments, the antibody or antigen binding molecule has an in vivo cholesterol lowering effect of at least about 2 weeks, for example, 2, 3, 4 weeks or longer. Preferably, the in vivo half-life is determined in a human subject.
  • the antibody comprises
  • a heavy chain variable region comprising a human heavy chain V-segment, a heavy chain complementarity determining region 3 (CDR3), and a heavy chain framework region 4 (FR4), and
  • the heavy chain CDR3 comprises the amino acid sequence SYYYY(A/N)MD(A/F/S/V/Y) (SEQ ID NO:14);
  • the light chain CDR3 variable region comprises the amino acid sequence LQWSSDPPT (SEQ ID NO:26).
  • the antibody comprises
  • a heavy chain variable region comprising a human heavy chain V-segment, a heavy chain complementarity determining region 3 (CDR3), and a heavy chain framework region 4 (FR4), and
  • the heavy chain CDR3 comprises the amino acid sequence SYYYYNMDY (SEQ ID NO: 12);
  • the light chain CDR3 variable region comprises the amino acid sequence LQWSSDPPT (SEQ ID NO:26).
  • the antibody comprises
  • a heavy chain variable region comprising a human heavy chain V-segment, a heavy chain complementarity determining region 3 (CDR3), and a heavy chain framework region 4 (FR4), and
  • the heavy chain CDR3 comprises the amino acid sequence SYYYYAMDY (SEQ ID NO: 13);
  • the light chain CDR3 variable region comprises the amino acid sequence LQWSSDPPT (SEQ ID NO:26).
  • the heavy chain CDR3 comprises the amino acid sequence selected from the group consisting of SEQ ID NO: 12 and SEQ ID NO: 13; and the light chain CDR3 comprises the amino acid sequence of SEQ ID NO:26.
  • the heavy chain V-segment has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO:28, and wherein the light chain V segment has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO:31.
  • the heavy chain V-segment has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO:27, and wherein the light chain V segment has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to the amino acid selected from the group consisting of SEQ ID NO:29 and SEQ ID NO:30.
  • the heavy chain FR4 is a human germline FR4. In some embodiments, the heavy chain FR4 is SEQ ID NO:35.
  • the light chain FR4 is a human germline FR4. In some embodiments, the light chain FR4 is SEQ ID NO:39.
  • the heavy chain V-segment and the light chain V-segment each comprise a complementarity determining region 1 (CDR1) and a complementarity determining region 2 (CDR2); wherein:
  • the CDR1 of the heavy chain V-segment comprises the amino acid sequence of
  • the CDR2 of the heavy chain V-segment comprises the amino acid sequence of SEQ ID NO:l l;
  • the CDR1 of the light chain V-segment comprises the amino acid sequence of SEQ ID NO:22;
  • the CDR2 of the light chain V-segment comprises the amino acid sequence of SEQ ID NO:25.
  • the heavy chain V-segment and the light chain V-segment each comprise a complementarity determining region 1 (CDR1) and a complementarity determining region 2 (CDR2); wherein:
  • the CDR1 of the heavy chain V-segment comprises the amino acid sequence of SEQ ID NO:7; ii) the CDR2 of the heavy chain V-segment comprises the amino acid sequence of SEQ ID NO: 10;
  • the CD 1 of the light chain V-segment comprises the amino acid sequence of SEQ ID NO:21;
  • the CDR2 of the light chain V-segment comprises the amino acid sequence of
  • the CDR1 of the heavy chain V-segment comprises SEQ ID NO: 7;
  • the CDR2 of the heavy chain V-segment comprises SEQ ID NO: 10;
  • the heavy chain CDR3 comprises the amino acid sequence selected from the group consisting of SEQ ID NO: 12 and SEQ ID NO: 13;
  • the CDR1 of the light chain V-segment comprises SEQ ID NO:21;
  • the CDR2 of the light chain V-segment comprises SEQ ID NO:24; and vi) the light chain CDR3 comprises SEQ ID NO:26.
  • the heavy chain variable region has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity to the variable region of SEQ ID NO:40 and the light chain variable region has at least 90% amino acid sequence identity to the variable region of SEQ ID NO:41.
  • the antibody comprises a heavy chain comprising SEQ ID NO:40 and a light chain comprising SEQ ID NO:41.
  • the heavy chain variable region has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity to the variable region selected from the group consisting of SEQ ID NO:2 and SEQ ID NO:4 and the light chain variable region has at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity to the variable region selected from the group consisting of SEQ ID NO: 16 and SEQ ID NO: 18.
  • the heavy chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO:2 and SEQ ID NO:4 and the light chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO: 16 and SEQ ID NO: 18.
  • the antibody is an IgG. In some embodiments, the antibody is an IgGl. In some embodiments, the antibody has a heavy chain sharing at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to an amino acid selected from the group consisting of SEQ ID NO:3 and SEQ ID NO:5. In some embodiments, the antibody has a light chain sharing at least 85%, 88%, 89%, 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to an amino acid selected from the group consisting of SEQ ID NO: 17 and SEQ ID NO: 19.
  • the antibody is a FAb' fragment. In some embodiments, the antibody is a single chain antibody (scFv). In some embodiments, the antibody comprises human constant regions. In some embodiments, the antibody comprises a human IgGl constant region. In some embodiments, the human IgGl constant region is mutated to have reduced binding affinity for an effector ligand such as Fc receptor (FcR), e.g., Fc gamma Rl, on a cell or the CI component of complement. See, e.g., U.S. Patent No. 5,624,821. In some embodiments, amino acid residues L234 and L235 of the IgGl constant region are substituted to Ala234 and Ala235. The numbering of the residues in the heavy chain constant region is that of the EU index (see, Kabat, et al., (1983) "Sequences of Proteins of Immunological Interest," U.S. Dept. Health and Human Services).
  • the antibody is linked to a carrier protein, for example, albumin.
  • the antibody is PEGylated.
  • the antibody comprises a heavy chain variable region and a light chain variable region, wherein the heavy chain variable region and the light chain variable region each comprise the following three complementarity determining regions (CDRs): CDR1, CDR2 and CDR3; wherein:
  • the CDR1 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:8;
  • the CDR2 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:l l ;
  • the CDR3 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 14;
  • the CDR1 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:22;
  • the CDR2 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:25; vi) the CDR3 of the light chain variable region comprises the amino acid sequence of SEQ ID O:26.
  • the CDRl of the heavy chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO:6 and SEQ ID NO:7;
  • the CDR2 of the heavy chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO:9 and SEQ ID NO: 10;
  • the CDR3 of the heavy chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO:12 and SEQ ID NO: 13;
  • the CDRl of the light chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO:20 and SEQ ID NO:21;
  • the CDR2 of the light chain variable region comprises the amino acid sequence selected from the group consisting of SEQ ID NO:23 and SEQ ID NO:24;
  • the CDR3 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:26.
  • the antibody comprises a heavy chain variable region and a light chain variable region, wherein the heavy chain variable region and the light chain variable region each comprise the following three complementarity determining regions (CDRs): CDRl, CDR2 and CDR3; wherein:
  • the CDRl of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:6;
  • the CDR2 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:9;
  • the CDR3 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 13;
  • the CDRl of the light chain variable region comprises the amino acid sequence of SEQ ID NO:20;
  • the CDR2 of the light chain variable region comprises the amino acid sequence of SEQ ID O:23;
  • the CDR3 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:26.
  • the heavy chain variable region and the light chain variable region each comprise the following three complementarity determining regions (CDRs): CDRl, CDR2 and CDR3; wherein: i) the CDR1 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:7;
  • the CDR2 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:10;
  • the CDR3 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 12;
  • the CDR1 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:21;
  • the CDR2 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:24;
  • the CDR3 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:26.
  • the antibody comprises a heavy chain variable region and a light chain variable region, wherein the heavy chain variable region and the light chain variable region each comprise the following three complementarity determining regions (CDRs): CDR1, CDR2 and CDR3; wherein:
  • the CDR1 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:7;
  • the CDR2 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10;
  • the CDR3 of the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 13 ;
  • the CDR1 of the light chain variable region comprises the amino acid sequence of SEQ ID O:21;
  • the CDR2 of the light chain variable region comprises the amino acid sequence of SEQ ID NO:24;
  • the CDR3 of the light chain variable region comprises the amino acid sequence of SEQ ID O:26.
  • the formulation of the present inventions can further comprise at least one pharmaceutically acceptable carrier or excipient.
  • the excipient can be a buffer system from the group consisting of citrate, arginine, histidine, sodium succinate, and sodium and/or potassium phosphate buffer systems.
  • histidine or arginine or both histidine and arginine can be used.
  • histidine is present at a concentration of 1-50 mM.
  • histidine is present at a concentration of 5- 15 mM before a lyophilisation step and at a concentration of 15-45 mM after reconstitution into a liquid formulation.
  • histidine is present at a concentration of 10 mM before a lyophilisation step and at a concentration of 30 mM after reconstitution into a liquid formulation.
  • arginine is present at a concentration of 15-60 mM.
  • arginine is present at a concentration of 1-20 mM, or 14-20 mM before a lyophilisation step and at a concentration of 42-60 mM after reconstitution into a liquid formulation.
  • arginine is present at a concentration of 17 mM before a lyophilisation step and at a concentration of 51 mM after reconstitution into a liquid formulation.
  • the formulation of the invention may preferably further comprise a stabilizer.
  • Stabilizers according to the present invention include sucrose, trehalose, mannitol, sorbitol and arginine hydrochloride.
  • the excipient or stabilizer is sucrose.
  • sucrose is present at a concentration of 40-90 mM or 70-90 mM before a lyophilisation step and at a concentration of 120 -270mM or 210-270 mM after reconstitution into a liquid formulation.
  • the formulation of the invention may optionally further comprise one or more excipients selected from a group comprising bulking agent, salt, surfactant and preservative.
  • a bulking agent is a compound which adds mass to a pharmaceutical formulation and contributes to the physical structure of the formulation in lyophilized form. Suitable bulking agents according to the present invention include mannitol, glycine, polyethylene glycol and sorbitol.
  • the pH is between 5.0 and 7.0 or between 5.0 and 6.0. In some embodiments, the pH is between 5.2 and 5.8 or between 5.3 and 5.7. In some embodiments, the pH is 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0. In some embodiments, the antibody is present in an amount of 20-300 mg/ml, 30- 200 mg/ml, 90-200 mg/ml, or 90-180 mg/ml. In some embodiments, the antibody is present in an amount of 30-50 mg/ml before a lyophilisation step and at a concentration of 99.9-150 mM after reconstitution into a liquid formulation.
  • a surfactant can reduce aggregation of the reconstituted protein and/or reduce the formation of particulates in the reconstituted formulation.
  • the amount of surfactant added is such that it reduces aggregation of the reconstituted protein and minimizes the formation of particulates after reconstitution.
  • Suitable surfactants according to the present invention include polysorbates (e.g. polysorbates 20 or 80); poloxamers (e.g.
  • poloxamer 188 Triton; sodium dodecyl sulfate (SDS); sodium laurel sulfate; 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
  • the surfactant may be selected from the group consisting of polysorbate 20 and polysorbate 80.
  • the concentration of the surfactant used for the formulation according to the present invention is from about 0.001-0.5%, or from about 0.005-0.10%, or 0.01 to 0.10%, weight by volume of the formulation.
  • the surfactant can be added to the pre-lyophilized formulation, the lyophilized formulation and/or the reconstituted formulation as desired.
  • preservatives may be used in formulations of invention.
  • Suitable preservatives for use in the formulation of the invention include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds), and benzethonium chloride.
  • Other types of preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol.
  • compositions of the invention may be included in the formulation of the invention provided that they do not adversely affect the desired characteristics of the formulation.
  • Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include additional buffering agents; preservatives; co-solvents; antioxidants including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium.
  • a stable reconstituted formulation is a formulation wherein the antibody therein essentially retains its physical and chemical stability and integrity upon storage for a period of time from the reconstitution to the use, typically a few hours and up to several days.
  • the invention provides a method for preparing a formulation comprising the steps of: (a) lyophilizing the formulation comprising the antibodies, and a buffer system; and (b) reconstituting the lyophilized mixture of step (a) in a reconstitution medium such that the reconstituted formulation is stable.
  • the reconstitution time is below 10 mins.
  • the formulation of step (a) may further comprise a stabilizer and one or more excipients selected from a group comprising bulking agent, salt, surfactant and preservative as hereinabove described.
  • a stabilizer selected from a group comprising bulking agent, salt, surfactant and preservative as hereinabove described.
  • As reconstitution media several diluted organic acids or water, i.e. sterile water, bacteriostatic water for injection (BWFT) or may be used.
  • the reconstitution medium may be selected from water, i.e. sterile water, bacteriostatic water for injection
  • the formulation of the invention comprising anti PCSK9 antibodies is administered preferably by intravenous route, but also by subcutaneous or intramuscular injection route.
  • the formulation may be injected using a syringe.
  • the formulation comprising LGT209 is administered using autoinjector, normal syringe that may be prefilled, optionally in a sterile package, optionally syringes with safety devices.
  • Micro- needle and coated patches with reservoirs are also envisaged as suitable administration devices.
  • the invention provides methods of reducing LDL-C, non-HDL-C and/or total cholesterol in an individual in need thereof, the method comprising administering a formulation according to the invention comprising an antibody or antigen binding molecule against PCSK9 in a therapeutically effective amount to the individual as described herein.
  • the use of a formulation is provided for administration to an individual having hypercholesterolemia or a high baseline LDL-C level.
  • the individual is hyporesponsive or resistant to statin therapy.
  • the individual is intolerant to statin therapy.
  • the individual has a baseline LDL-C level of at least about 70 mg/dL, for example, at least about 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 mg/dL, or higher.
  • the individual has familial hypercholesterolemia.
  • the individual has triglyceridemia.
  • the individual has a gain-of-function PCSK9 gene mutation.
  • the individual has drug-induced dyslipidemia.
  • total cholesterol is reduced with LDL-C.
  • the formulations according to the invention can be used as a medicament in patients eligible for apheresis. The definitions of apheresis can be found later in this description.
  • the use or the methods further comprise co-administering a therapeutically effective amount of a second agent effective in reducing LDL-C to the individual.
  • the second agent is a statin.
  • the statin can be selected from the group consisting of atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin.
  • the second agent is selected from the group consisting of fibrates, niacin and analogs thereof, cholesterol absorption inhibitors, bile acid sequestrants, thyroid hormone mimetics, a microsomal triglyceride transfer protein (MTP) inhibitor, a diacylglycerol acyltransferase (DGAT) inhibitor, an inhibitory nucleic acid targeting PCSK9 and an inhibitory nucleic acid targeting apoBlOO.
  • MTP microsomal triglyceride transfer protein
  • DGAT diacylglycerol acyltransferase
  • the antibody is administered subcutaneously.
  • small volumes of pharmaceutical formulation for subcutaneous injection usually 1.0 mL-1.2 mL at a maximum.
  • the subcutaneous administration requires high
  • concentration antibody formulations e.g., 50 mg/ml-150 mg ml or more.
  • a liquid pharmaceutical antibody formulation should exhibit a variety of pre-defined characteristics.
  • One of the major concerns in liquid drug products is stability, as proteins tend to form soluble and insoluble aggregates during manufacturing and storage.
  • various chemical reactions can occur in solution (deamidation, oxidation, clipping, isomerization etc.) leading to an increase in degradation product levels and/or loss of bioactivity.
  • a liquid antibody formulation should exhibit a shelf life of more than 18 months. Most preferred a liquid antibody formulation should exhibit a shelf life of more than 24 months.
  • formulation of the invention comprising an antibody, as active ingredient and a buffer system, a stabilizer and a surfactant.
  • the formulation of the invention is liquid but is also suitable to be lyophilized and subsequently be reconstituted to a liquid formulation with a lower, same or higher antibody concentration.
  • a reconstituted formulation is a formulation which has been prepared from a lyophilisate, such that the antibody of the formulation is dispersed in the reconstituted formulation
  • Another aspect provides a method of producing a lyophilized pharmaceutical formulation according to the invention , comprising at least about 10 mg/mL of said antibody post reconstitution, by contacting a) said antibody with b) at least one pharmaceutically acceptable excipient.
  • a method for producing a liquid pharmaceutical formulation comprising at least about 10 mg/mL of said antibody post reconstitution, by contacting a) said antibody with b) at least one pharmaceutically acceptable excipient.
  • Example 5 in connection to figures 1 to 4 describes one way to produce an antibody formulation according to the invention.
  • the antibodies and antigen-binding molecules for use in the formulations of the present invention specifically bind to proprotein convertase subtilisin kexin type 9a
  • PCSK9 The present anti-PCSK9 antibodies and antigen-binding molecules bind to the C-terminus of PCSK9 and have the unexpected property of interfering with PCSK9-mediated degradation of the low density lipoprotein receptor (LDL-R) without interfering with binding of PCSK9 to the LDL-R.
  • the anti-PCSK9 antibodies and antigen binding molecules bind to an epitope within residues 680-692 of PCSK9, for example, an epitope within the amino acid sequence RSRHLAQASQELQ (SEQ ID NO:49), located at the C- terminal end of PCSK9.
  • the antibodies and antigen-binding molecules of the invention bind to PCSK9 while bound on a cell rather than only to circulating PCSK9, they have a comparatively longer in vivo half-life in a patient, e.g., at least about 7 days or longer, and in some embodiments, provide lipid-lowering effects for at least 2 weeks after administration.
  • the anti-PCSK9 antibodies and antigen binding molecules are antagonists of PCSK9 in that they prevent, reduce and/or inhibit PCSK9-mediated degradation of the LDL- R, thereby facilitating increased uptake of low density lipoprotein cholesterol (LDL-C).
  • LDL-C low density lipoprotein cholesterol
  • the anti-PCSK9 antibodies and antigen binding molecules find use in treating subjects suffering from, e.g., dyslipidemia, hypercholesterolemia, triglyceridemia and other PCSK9-mediated disease conditions.
  • Anti-PCSK9 antibody fragments can be produced by any means known in the art, including but not limited to, recombinant expression, chemical synthesis, and enzymatic digestion of antibody tetramers, whereas full-length monoclonal antibodies can be obtained by, e.g., hybridoma or recombinant production.
  • Recombinant expression can be from any appropriate host cells known in the art, for example, mammalian host cells, bacterial host cells, yeast host cells, insect host cells, etc.
  • the constant regions of the anti- PCSK9 antibodies can be any type or subtype, as appropriate, and can be selected to be from the species of the subject to be treated by the present methods (e.g., human, non-human primate or other mammal, for example, agricultural mammal (e.g., equine, ovine, bovine, porcine, camelid), domestic mammal (e.g., canine, feline) or rodent (e.g., rat, mouse, hamster, rabbit).
  • the anti-PCSK9 antibodies are humanized or HumaneeredTM.
  • the constant region isotype is IgG, for example, IgGl.
  • the human IgGl constant region is mutated to have reduced binding affinity for an effector ligand such as Fc receptor (FcR), e.g., Fc gamma Rl, on a cell or the CI component of complement.
  • FcR effector ligand
  • Antibodies containing such mutations mediate reduced or no antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • amino acid residues L234 and L235 of the IgGl constant region are substituted to Ala234 and Ala235.
  • the numbering of the residues in the heavy chain constant region is that of the EU index (see, Kabat, et al., (1983) "Sequences of Proteins of Immunological Interest," U.S. Dept. Health and Human Services). See also, e.g., Woodle, et al, Transplantation (1999) 68(5):608-616; Xu, et al, Cell Immunol (2000) 200(1): 16-26; and Hezareh, et al, J Virol 75(24): 12161-8.
  • Anti-PCSK9 antibodies or antigen-binding molecules of the invention also include single domain antigen-binding units which have a camelid scaffold.
  • Animals in the camelid family include camels, llamas, and alpacas.
  • Camelids produce functional antibodies devoid of light chains.
  • the heavy chain variable (VH) domain folds autonomously and functions independently as an antigen-binding unit. Its binding surface involves only three CDRs as compared to the six CDRs in classical antigen-binding molecules (Fabs) or single chain variable fragments (scFvs).
  • Fabs classical antigen-binding molecules
  • scFvs single chain variable fragments
  • Camelid scaffold-based anti-PCSK9 molecules with binding specificities of the anti-PCSK9 antibodies exemplified herein can be produced using methods well known in the art, e.g., Dumoulin et al., Nature Struct. Biol. 11:500-515, 2002; Ghahroudi et al, FEBS Letters 414:521-526, 1997; and Bond et al, J Mol Biol. 332:643-55, 2003.
  • the improved anti-PC SK9 antibodies of the invention are engineered human antibodies with V-region sequences having substantial amino acid sequence identity to human germline V-region sequences while retaining the specificity and affinity of a reference antibody. See, U.S. Patent Publication No. 2005/0255552 and U.S. Patent Publication No. 2006/0134098, both of which are hereby incorporated herein by reference.
  • the process of improvement identifies minimal sequence information required to determine antigen-binding specificity from the variable region of a reference antibody, and transfers that information to a library of human partial V-region gene sequences to generate an epitope-focused library of human antibody V-regions.
  • a microbial-based secretion system can be used to express members of the library as antibody Fab fragments and the library is screened for antigen- binding Fabs, for example, using a colony-lift binding assay. See, e.g., U.S. Patent
  • Positive clones can be further characterized to identify those with the highest affinity.
  • the resultant engineered human Fabs retain the binding specificity of the parent, reference anti-PC SK9 antibody, typically have equivalent or higher affinity for antigen in comparison to the parent antibody, and have V-regions with a high degree of sequence identity compared with human germ-line antibody V-regions.
  • the minimum binding specificity determinant (BSD) required to generate the epitope-focused library is typically represented by a sequence within the heavy chain CDR3 ("CDRH3") and a sequence within the light chain of CDR3 ("CDRL3").
  • the BSD can comprise a portion or the entire length of a CDR3.
  • the BSD can be comprised of contiguous or non-contiguous amino acid residues.
  • the epitope-focused library is constructed from human V-segment sequences linked to the unique CDR3-FR4 region from the reference antibody containing the BSD and human germ-line J-segment sequences ⁇ see, U.S. Patent Publication No. 2005/0255552).
  • the human V-segment libraries can be generated by sequential cassette replacement in which only part of the reference antibody V-segment is initially replaced by a library of human sequences.
  • the identified human "cassettes" supporting binding in the context of residual reference antibody amino acid sequences are then recombined in a second library screen to generate completely human V-segments ⁇ see, U.S. Patent Publication No. 2006/0134098).
  • paired heavy and light chain CDR3 segments, CDR3-FR4 segments, or J-segments, containing specificity determinants from the reference antibody are used to constrain the binding specificity so that antigen-binders obtained from the library retain the epitope-specificity of the reference antibody. Additional maturational changes can be introduced in the CDR3 regions of each chain during the library construction in order to identify antibodies with optimal binding kinetics.
  • the resulting engineered human antibodies have V-segment sequences derived from the human germ-line libraries, retain the short BSD sequence from within the CDR3 regions and have human germ-line framework 4 (FR4) regions.
  • the anti-PCSK9 antibodies contain a minimum binding sequence determinant (BSD) within the CDR3 of the heavy and light chains derived from the originating or reference monoclonal antibody.
  • BSD binding sequence determinant
  • the remaining sequences of the heavy chain and light chain variable regions (CDR and FR), e.g., V-segment and J-segment, are from corresponding human germline and affinity matured amino acid sequences.
  • the V- segments can be selected from a human V-segment library. Further sequence refinement can be accomplished by affinity maturation.
  • the heavy and light chains of the anti-PC SK9 antibodies contain a human V-segment from the corresponding human germline sequence (FR1-CDR1- FR2-CDR2-FR3), e.g., selected from a human V-segment library, and a CDR3-FR4 sequence segment from the originating monoclonal antibody.
  • the CDR3-FR4 sequence segment can be further refined by replacing sequence segments with corresponding human germline sequences and/or by affinity maturation.
  • the FR4 and/or the CDR3 sequence surrounding the BSD can be replaced with the corresponding human germline sequence, while the BSD from the CDR3 of the originating monoclonal antibody is retained.
  • the corresponding human germline sequence for the heavy chain V-segment is Vhl-02.
  • the corresponding human germline sequence for the heavy chain J-segment is JH4.
  • the heavy chain J- segment comprises the human germline JH4 partial sequence WGQGTLVTVSS (SEQ ID NO:50).
  • the full-length J-segment from human germline JH4 is YFDY WGQGTL VTV S S (SEQ ID NO:51).
  • the variable region genes are referenced in accordance with the standard nomenclature for immunoglobulin variable region genes. Current immunoglobulin gene information is available through the worldwide web, for example, on the ImMunoGeneTics (IMGT), V-base and PubMed databases. See also, Lefranc, Exp Clin Immunogenet.
  • the corresponding human germline sequence for the light chain V-segment is VK3 L6.
  • the corresponding human germline sequence for the light chain J-segment is Jk2.
  • the light chain J- segment comprises the human germline Jk2 partial sequence FGQGTKLEIK (SEQ ID NO:52).
  • the full-length J-segment from human germline Jk2 is YTFGQGTKLEIK (SEQ ID NO:53).
  • the heavy chain V-segment has at least 85%, 89%, 90%, 93%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence QVQLVQSGAEVKKPGASVKVSCKASGYTFS(D T)MYMSWVRQAPGQGLEWMGRID PAN(A/E/G)HTNY(A/D)(P/Q)KFQ(A/G)RVTMTRDTSISTAYMELSRLTSDDTAVYYCA R (SEQ ID NO:28).
  • the heavy chain V-segment has at least 85%, 89%, 90%, 93%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence
  • the light chain V-segment has at least 85%, 89%, 90%, 93%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence
  • the heavy chain V-segment has at least 85%, 89%, 90%, 93%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence QIVLTQSPATLSVSPGERATLSCRASQSVSYMHWYQQKPGQAPRLLIYGVFRRATGIP DRFSGSGSGTDFTLTIGRLEPEDFAVYYC (SEQ ID NO:29). In some embodiments, the heavy chain V-segment has at least 85%, 89%, 90%, 93%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence
  • the heavy chain CDR3 comprises the amino acid sequence
  • the light chain CDR3 variable region comprises the amino acid sequence LQWSSDPPT (SEQ ID NO:26).
  • the heavy chain CDR3 comprises the amino acid sequence selected from the group consisting of SEQ ID NO:12 and SEQ ID NO:13;
  • the light chain CDR3 comprises the amino acid sequence of SEQ ID NO: 1
  • the antibodies of the invention comprise a heavy chain variable region comprising a CDR1 comprising an amino acid sequence (D/T)MYMS (SEQ ID NO:8); a CDR2 comprising an amino acid sequence
  • RIDPAN(A/E/G)HTNY(A/D)(P/Q)KFQ(A/G) (SEQ ID NO:l 1); and a CDR3 comprising an amino acid sequence of SYYYY(A/N)MD(A/F/S/ Y) (SEQ ID NO: 14).
  • the antibodies of the invention comprise a light chain variable region comprising a CDR1 comprising an amino acid sequence
  • R/SAS(Q/S)SVSYMH (SEQ ID NO:22); a CDR2 comprising an amino acid sequence (G/L)(T/V)F(N/R)(L/R)A(S/T) (SEQ ID NO:25); and a CDR3 comprising an amino acid sequence of LQWSSDPPT (SEQ ID NO:26).
  • the heavy chain variable region comprises a FR1 comprising the amino acid sequence of SEQ ID NO:32; a FR2 comprising the amino acid sequence of SEQ ID NO:33; a FR3 comprising the amino acid sequence of SEQ ID NO:34; and a FR4 comprising the amino acid sequence of SEQ ID NO:35.
  • the identified amino acid sequences may have one or more substituted amino acids (e.g., from affinity maturation) or one or two conservatively substituted amino acids.
  • the light chain variable region comprises a FR1 comprising an amino acid sequence of SEQ ID NO:36; a FR2 comprising the amino acid sequence of SEQ ID NO:37; a FR3 comprising the amino acid sequence of SEQ ID NO:38; and a FR4 comprising the amino acid sequence of SEQ ID NO:39.
  • the identified amino acid sequences may have one or more substituted amino acids (e.g., from affinity maturation) or one or two conservatively substituted amino acids.
  • variable regions of the anti-PCSK9 antibodies of the present invention generally will have an overall variable region (e.g., FR1-CDR1-FR2- CDR2-FR3-CDR3-FR4) amino acid sequence identity of at least about 85%, for example, at least about 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% to the corresponding human germline variable region amino acid sequence.
  • overall variable region e.g., FR1-CDR1-FR2- CDR2-FR3-CDR3-FR4 amino acid sequence identity of at least about 85%, for example, at least about 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% to the corresponding human germline variable region amino acid sequence.
  • the heavy chain of the anti-PCSK9 antibodies can have at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the human germline variable region Vhl-02.
  • the light chain of the anti-PCSK9 antibodies can have at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the human germline variable region VK3 L6.
  • only amino acids within the framework regions are added, deleted or substituted.
  • the sequence identity comparison excludes the CD3.
  • the anti-PC SK9 antibodies of the invention comprise a heavy chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:40 and comprise a light chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:41 (i.e., consensus sequences).
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain variable region having at least 85%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:l and comprise a light chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:15 (i.e., mouse LFU720).
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:2 and comprise a light chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:16 (i.e., LGT-209).
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:2 and comprise a light chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO: 18 (i.e., LGT-210).
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:4 and comprise a light chain variable region having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:16 (i.e., LGT-211).
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain polypeptide having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:3 and comprise a light chain polypeptide having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO: 17 ⁇ i.e., LGT-209).
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain polypeptide having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:3 and comprise a light chain polypeptide having at least 85%, 89%, 90%, 91%, 92%,
  • the anti-PCSK9 antibodies of the invention comprise a heavy chain polypeptide having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:5 and comprise a light chain polypeptide having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO: 17 ⁇ i.e., LGT-21 1).
  • the anti-PCSK9 antibodies of the present invention generally will bind PCSK9 with an equilibrium dissociation constant (K D ) of less than about 10 "8 M or 10 "9 M, for example, less than about 10 "10 M or 10 ⁇ N M, in some embodiments less than about lO "12 M or 10 "13 M.
  • K D equilibrium dissociation constant
  • the anti-PCSK9 antibodies optionally can be multimerized and used according to the methods of this invention.
  • the anti- PCSK9 antibodies can be a full-length tetrameric antibody ⁇ i.e., having two light chains and two heavy chains), a single chain antibody ⁇ e.g., a scFv), or a molecule comprising antibody fragments that form one or more antigen-binding sites and confer PCSK9-binding specificity, e.g., comprising heavy and light chain variable regions (for instance, Fab' or other similar fragments).
  • the invention further provides polynucleotides encoding the antibodies described herein, e.g., polynucleotides encoding heavy or light chain variable regions or segments comprising the complementarity determining regions as described herein.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:42, SEQ ID NO:43 and SEQ ID NO:54.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:55.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:42.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:45 (i.e., LGT-209).
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:42.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:44 (i.e., LGT-210).
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:43.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:45 '.e., LGT-211).
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:54.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:55 (i.e., mouse LFU720).
  • Antagonist antibodies can be identified by generating anti-PC SK9 antibodies and then testing each antibody for the ability to reduce or inhibit PCSK9 mediated events, e.g., binding to the LDLR, promoting the degradation of the LDLR.
  • the assays can be carried out in vitro or in vivo.
  • Preferred antibodies bind to PCSK9, do not prevent PCSK9 from binding to LDLR, and reduce or inhibit PCSK9-mediated degradation of LDLR.
  • the binding of the antibodies or antigen binding molecules to PCSK9 can be determined using any method known in the art, including without limitation, ELISA, Biacore and Western Blot.
  • PCSK9-mediated degradation of LDLR also can be measured using any method known in the art.
  • the ability of the anti-PC SK9 antibody or antigen binding molecule to inhibit LDLR degradation is determined using an infusion mouse model.
  • Anti-PCSK9 antibodies or antigen binding molecules are infused intravenously (e.g., 3 ⁇ g/hour) into a mouse and the levels of LDLR in liver membrane preparations is determined in comparison to the levels of LDLR in liver membrane preparations from a mouse that has received intravenous infusions of a control antibody (e.g., that binds to an unrelated antigen).
  • Mice that have received antagonist anti-PCSK9 antibodies will have detectably higher levels of LDLR, e.g., at least 10%, 20%, 50%, 80%, 100% higher, in comparison to mice that have received the control antibody.
  • Anti-PC SK9 antagonist antibodies also can be tested for their therapeutic efficacy in reducing plasma levels of LCL-C, non-HDL-C and/or total cholesterol.
  • Anti-PCSK9 antibodies or antigen binding molecules are infused intravenously (e.g., 3 g/hour) into a mammal (e.g., mouse, rat, non-human primate, human) and the plasma levels of LCL-C, non- HDL-C and/or total cholesterol is determined in comparison to the plasma levels of LCL-C, non-HDL-C and/or total cholesterol from the same mammal before treatment or from a mammal that has received intravenous infusions of a control antibody (e.g., that binds to an unrelated antigen).
  • a control antibody e.g., that binds to an unrelated antigen
  • the mammal that has received antagonist anti-PCSK9 antibodies will have detectably lower plasma levels of LCL-C, non-HDL-C and/or total cholesterol, e.g., at least 10%, 20%, 50%, 80%, 100% lower, in comparison to the mammal before treatment or the mammal that has received the control antibody.
  • the pharmacological compositions comprise a mixture of the anti-PCSK9 antibody or antigen binding molecule and a second pharmacological agent.
  • the compositions may comprise a anti-PCSK9 antibody or antigen-binding molecule of the invention and an agent known to be beneficial for reducing cholesterol, including LDL-C, non-HDL-C and total cholesterol and/or raising HDL-C.
  • Exemplary second agents for inclusion in mixtures with the present anti-PCSK9 antagonist antibody or antigen binding molecule include without limitation an HMG-CoA reductase inhibitor (i.e., a statin), fibrates (e.g., clofibrate, gemfibrozil, fenofibrate, ciprofibrate, bezafibrate), niacin and analogs thereof, cholesterol absorption inhibitors, bile acid sequestrants (e.g., cholestyramine, colestipol, colesvelam), an ileal bile acid transport (IB AT) inhibitor, a thyroid hormone mimetic (e.g., compound KB2115), a microsomal triglyceride transfer protein (MTP) inhibitor, a dual peroxisome proliferator-activated receptor (PPAR) alpha and gamma agonist, an acyl CoArdiacylglycerol acyltransferase (DGAT) inhibitor,
  • Lipid- lowering agents are known in the art, and described, e.g., in Goodman and Gilman's The Pharmacological Basis of Therapeutics, 11th Ed., Brunton, Lazo and Parker, Eds., McGraw- Hill (2006); 2009 Physicians ' Desk Reference (PDR), for example, in the 63rd (2008) Eds., Thomson PDR.
  • the anti-PC SK9 antibodies or antigen binding molecules of the invention are provided as a mixture with a statin.
  • exemplary statins include without limitation, atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin.
  • the anti-PCSK9 antibodies or antigen binding molecules of the invention are provided as a mixture with a pharmacological agent that induces hypercholesterolemia or triglyceridemia.
  • the second pharmacological agent may be a protease inhibitor, for example, Saquinavir, Ritonavir, Indinavir, Nelfinavir, Amprenavir, Lopinavir, Atazanavir, Fosamprenavir, Tipranavir, Darunavir, abacavir- lamivudine-zidovudine (Trizivir).
  • the second pharmacological agent is Tacrolimus.
  • the anti-PCSK9 antagonist antibodies and antigen binding molecules of the invention find use in treating any disease condition mediated by the activity or over-activity ofPCSK9.
  • the individual may have familial or genetically transmitted homozygous or heterozygous hypercholesterolemia in which a functional LDL-R is present. Genetic mutations associated with and/or causative of familial or genetically inherited hypercholesterolemia are summarized, e.g., in Burnett and Hooper, Clin Biochem Rev (2008) 29(1):11-26.
  • the individual may also have other disease conditions or engage in behaviors that contribute to or increase the risk of developing dyslipidemia or hypercholesterolemia.
  • the individual may be obese, or suffer from diabetes or metabolic syndrome.
  • the individual may be a smoker, lead a sedentary lifestyle, or have a diet high in cholesterol.
  • Targeting PCSK9 is useful for the reduction, reversal, inhibition or prevention of dyslipidemia, hypercholesterolemia and postprandial triglyceridemia. See, e.g., Le May, et ah, Arterioscler Thromb Vase Biol (2009) 29(5):684-90; Seidah, Expert Opin Ther Targets (2009) 13(l):19-28; and Poirier, et al, J Biol Chem (2009) PMID 19635789.
  • administration of the present anti-PCSK9 antagonist antibodies and antigen binding molecules finds use in reducing, reversing, inhibiting and preventing, dyslipidemia, hypercholesterolemia and postprandial triglyceridemia in an individual in need thereof.
  • the present anti-PC SK9 antagonist antibodies and antigen binding molecules find use in reducing or lowering low density lipoprotein cholesterol (LDL-C) in an individual in need thereof.
  • the individual may have persistently elevated levels of LDL-C.
  • the individual has LDL-C plasma levels consistently above 70 mg/dL, for example above 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 mg/dL, or higher.
  • the present anti-PCSK9 antagonist antibodies and antigen binding molecules also find use in reducing or lowering non-high density lipoprotein cholesterol (non-HDL-C) or total cholesterol in an individual in need thereof.
  • the individual may already be taking another pharmacological agent to lower cholesterol, and be resistant or intolerant to this agent.
  • the individual may already be under a therapeutic regimen of a statin, which may have proven inefficacious in this individual in lowering LDL-C, non-HDL-C or total cholesterol to acceptable levels.
  • the individual may also be intolerant to the administration of a statin.
  • statins Despite the proven effectiveness of statins and certain other lipid lowering medicines, there remains a substantial need for further LDL-C lowering. Over 1/3 of patients treated and compliant with statin therapy are not at goal (Keevil, JG et al., 2007) and approximately 1 in 15 are intolerant (Davidson, MH and Robinson, JG, 2007). Failure to achieve target LDL-C levels with statins occur in several scenarios such as statin resistance, attenuated response to increasing statin dose (typical of all statins), and statin-induced side effects leading to drug intolerance.
  • cardiovascular risk group is those patients with established CHD and LDL-C >130 mg/dl despite maximum tolerated statin therapy. Given the observed linear relationship between LDL-C and adverse cardiovascular events, these patients may be expected have an event rate as high as 15-30% (see Figure 2-11) (LaRosa, JC et al., 2005).
  • Combined administration of the present anti-PCSK9 antagonist antibodies and antigen binding molecules with a second agent useful in lowering LDL-C or non-HDL-C and/or raising HDL-C will improve the efficaciousness and tolerance of the second agent, for example, by allowing lower doses of the second agent to be administered.
  • the individual has a gain-of-function mutation in the PCSK9 gene, for example, that results in an aberrant increase in the degradation of the LDLR.
  • a gain-of-function mutation in the PCSK9 gene for example, that results in an aberrant increase in the degradation of the LDLR.
  • Abifadel et al first described that PCSK9 mutations could result in clinical ADH (Abifadel, M et al., 2003). Since that time, a number of reports have followed showing that other GOF mutations in the PCSK9 gene can lead to severe clinical dyslipidemia (Abifadel, M et al., 2008; Naoumova, RP et al., 2005; Timms, KM et al, 2004). Some of these patients have a substantially increased risk of premature CAD.
  • the individual is receiving a pharmacological agent the induces dyslipidemia or hypercholesterolemia, i.e., the individual has drug-induced dyslipidemia or hypercholesterolemia.
  • the individual may be receiving a therapeutic regime of protease inhibitors, e.g., for the treatment of an HIV infection.
  • Another pharmacological agent known to cause elevated levels of plasma triglycerides is Tacrolimus, an pharmacological agent known to cause elevated levels of plasma triglycerides.
  • Cyclosporin has been shown to increase LDL significantly. See, e.g., Ballantyne, et al. (1996) 78(5):532-5.
  • Second-generation antipsychotics ⁇ e.g., aripiprazole, clozapine, olanzapine, quetiapine, risperidone, and ziprasidone
  • aripiprazole clozapine
  • olanzapine quetiapine
  • risperidone ziprasidone
  • ziprasidone ziprasidone
  • the patients are eligible for apheresis.
  • LDL apheresis is an established technology indicated for the treatment of severe
  • hypercholesterolemia involves the use of an extracorporeal mechanism that filters the blood using one of several different proprietary technologies (Julius, U et al., 2008) to achieve an acute procedural goal of lowering plasma LDL-C by 60-80% (Moriarty, PM, 2006; Thompson, GR et al., 2010).
  • the procedure removes apoB-containing lipoproteins without disturbing plasma levels of albumin, electrolytes, immunoglobulins, or hemoglobin. It typically requires several hours for completion and involves phlebotomy and the administration of IV heparin to ensure that blood does not clot in the extracorporeal system.
  • Patients on treatment with LDL apheresis are a heterogeneous group including patients with homozygous familial hypercholesterolemia (HoFH), heterozygous familial hypercholesterolemia (HeFH), and those with severe hypercholesterolemia not meeting known definitions of familial hypercholesterolemia (FH).
  • HoFH homozygous familial hypercholesterolemia
  • HeFH heterozygous familial hypercholesterolemia
  • FH familial hypercholesterolemia
  • FH familial hypercholesterolemia
  • Individuals afflicted with FH are among the most severely affected patients.
  • Those with HoFH have profound hypercholesterolemia, typically >500 mg/dl, and predictably develop premature atherosclerosis (Marais, AD et al., 2004).
  • the accelerated disease process can lead to aortic fibrosis and stenosis, ostial coronary artery stenosis, and clinical symptoms as early as 2-3 years of age (Marais, AD et al., 2004; Naoumova, RP et al., 2004).
  • a small number of observational and randomized trials have suggested clinical benefit from apheresis treatment in the FH population (Mehta, PK et al., 2009; Moriarty, PM, 2006; Thompson, GR and Group, H-ULAW, 2008; Ziajka, P, 2005).
  • Apheresis eligible patients are defined as those who despite 6 months of medical and dietary therapy meet the following FDA defined criteria (Szczepiorkowski, ZM et al., 2007; Watts, G and S, H, 2009) referring to types of FH patients: a. Functional homozygotes with an LDL-C >500 mg/dl
  • apheresis costs are similar to renal hemodialysis. Recent estimates from the UK, Australia, and the US give similar figures.
  • the cost per apheresis session can range between $1500-$2300 USD, and the annual costs can range from $35,000-$75,000 USD, depending on country, reimbursement, and frequency of treatment (Novartis, 2010; Thompson, GR and Group, H-ULAW, 2008; Watts, G and S, H, 2009).
  • Pcsk9 antibodies mechanism of action would be predicted to have maximal effect in patients with a fully functional LDL-R, a therapy is offered to those patients meeting apheresis eligibility criteria b through d as stated above.
  • a physician or veterinarian can start doses of the formulations of the invention comprising the antibodies against PCSK9 at levels lower than that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • effective doses of the formulations of the present invention vary depending upon many different factors, including the specific disease or condition to be treated, means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic.
  • Treatment dosages need to be titrated to optimize safety and efficacy.
  • the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 10 mg/kg, of the host body weight.
  • dosages can be 0.3, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mg/kg body weight or within the range of 0.1-12, 0.3-10 or 1-10 mg/kg body weight.
  • Dosing can be daily, weekly, bi-weekly, monthly, or more or less often, as needed or desired.
  • An exemplary treatment regime entails
  • the antibody can be administered in single or divided doses.
  • Antibody is usually administered on multiple occasions. Intervals between single dosages can be weekly, biweekly, monthly or yearly, as needed or desired. Intervals can also be irregular as indicated by measuring blood levels of anti-PC SK9 antibody in the patient. In some methods, dosage is adjusted to achieve a plasma antibody concentration of 1-1000 g/ml and in some methods 25-300 ⁇ ⁇ 1. Alternatively, antibody can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, humanized antibodies show longer half life than that of chimeric antibodies and nonhuman antibodies.
  • the dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic.
  • a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives.
  • a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • the anti-PCSK9 antibody or antigen binding agent is administered when plasma LDL-C levels in the patient rise above a predetermined threshold level, for example, at least about 70, mg/dL, for example, at least about 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 mg/dL, or higher.
  • a predetermined threshold level for example, at least about 70, mg/dL, for example, at least about 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 mg/dL, or higher.
  • the PCSK9 antibody antagonist can be used in combination with agents known to be beneficial for reducing cholesterol, including LDL-C, non-HDL-C and total cholesterol and/or raising HDL-C.
  • Active agents can be administered together in a mixture with the anti-PC SK9 antagonist antibody or each agent can be administered separately.
  • the antibody agent and the other active agent can, but need not, be administered concurrently.
  • Exemplary second agents for use in co-administration with the present anti-PCSK9 antagonist antibody or antigen binding molecule include without limitation an HMG-CoA reductase inhibitor (i.e., a statin), fibrates (e.g., clofibrate, gemfibrozil, fenofibrate, ciprofibrate, bezaflbrate), niacin and analogs thereof, cholesterol absorption inhibitors, bile acid sequestrants (e.g., cholestyramine, colestipol, colesvelam), an ileal bile acid transport (IBAT) inhibitor, a thyroid hormone mimetic (e.g., compound KB2115), a microsomal triglyceride transfer protein (MTP) inhibitor, a dual peroxisome proliferator-activated receptor (PPAR) alpha and gamma agonist, an acyl CoArdiacylglycerol acyltransferase (DGAT) inhibitor
  • the anti-PCSK9 antibodies or antigen binding molecules of the invention are co-administered with a statin.
  • exemplary statins include without limitation, atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin.
  • the anti-PCSK9 antibodies or antigen binding molecules of the invention are co-administered with a pharmacological agent that induces hypercholesterolemia or triglyceridemia.
  • the second pharmacological agent may be a protease inhibitor, for example, Saquinavir, Ritonavir, Indinavir, Nelfmavir, Amprenavir, Lopinavir, Atazanavir, Fosamprenavir, Tipranavir, Darunavir, abacavir- lamivudine-zidovudine (Trizivir).
  • the second pharmacological agent is Tacrolimus.
  • the anti-PCSK9 antibodies or antigen binding molecules of the invention are co-administered with an inhibitory nucleic acid (e.g., an siRNA, an miRNA, an antisense sequence, a ribozyme) that specifically targets PCSK9 or apoBlOO.
  • an inhibitory nucleic acid e.g., an siRNA, an miRNA, an antisense sequence, a ribozyme
  • kits of the present invention comprises an anti-PCSK9 antagonist antibody or antigen binding molecule of the invention, as described herein.
  • the anti-PCSK9 antibodies or antigen binding molecules can be provided in uniform or varying dosages.
  • kits comprise one or more second pharmacological agents, as described herein.
  • the second pharmacological agent can be provided in the same formulation or in separate formulations from the anti-PC SK9 antibodies or antigen binding molecules.
  • the dosages of the first and second agents can be independently uniform or varying.
  • kits comprise the PCSK9 antibody antagonist an one or more agents known to be beneficial for reducing cholesterol, including LDL-C, non-HDL-C and total cholesterol and/or raising HDL-C.
  • Exemplary second agents for inclusion in the kits with the present anti-PCSK9 antagonist antibody or antigen binding molecule include without limitation an HDVlG-CoA reductase inhibitor (i.e., a statin), fibrates (e.g., clofibrate, gemfibrozil, fenofibrate, ciprofibrate, bezafibrate), niacin and analogs thereof, cholesterol absorption inhibitors, bile acid sequestrants (e.g., cholestyramine, colestipol, colesvelam), an ileal bile acid transport (IB AT) inhibitor, a thyroid hormone mimetic (e.g.
  • MTP microsomal triglyceride transfer protein
  • PPAR peroxisome proliferator-activated receptor alpha and gamma agonist
  • an acyl CoA:diacylglycerol acyltransferase (DGAT) inhibitor an acyl CoA holesterol acyltransferase (ACAT) inhibitor
  • DGAT acyl CoA holesterol acyltransferase
  • ACAT Niemann Pick Cl-like 1
  • NPC1-L1 Niemann Pick Cl-like 1
  • ATC1-L1 Niemann Pick Cl-like 1
  • ezetimibe an agonist of ATP Binding Cassette (ABC) proteins G5 or G8
  • CETP cholesterol ester transfer protein
  • CETP cholesterol ester transfer protein
  • the anti-PCSK9 antibodies or antigen binding molecules of the invention provided in kits with a statin.
  • exemplary statins include without limitation, atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin.
  • the anti-PC SK9 antibodies or antigen binding molecules of the invention are provided in kits with a pharmacological agent that induces
  • the second pharmacological agent may be a protease inhibitor, for example, Saquinavir, Ritonavir, Indinavir, Nelfinavir, Amprenavir, Lopinavir, Atazanavir, Fosamprenavir, Tipranavir, Darunavir, abacavir- lamivudine-zidovudine (Trizivir).
  • the second pharmacological agent is Tacrolimus.
  • a dosage form comprising a liquid pharmaceutical composition according to the invention, wherein the antibody formulation contained in a prefilled syringe.
  • concentration of the antibody is usually about 100, 150 or 200 mg/ml but all concentrations mentioned earlier in this description can be used for the prefilled syringe.
  • the prefilled syringe can also be adopted to be inserted into an autoinjector device.
  • the present invention provides an autoinjector comprising an prefilled syringe comprising the formulation according to the invention.
  • Use of an autoinjector will facilitate administration of the antibody formulation into the patient. It also makes it easier for the patients to self-inject the antibody formulation without medical assistance.
  • Use of an autoinjector is often more advantageous in subcutaneous administration of formulations of therapeutic antibodies.
  • a dosage form comprising a lyophilized pharmaceutical composition according to any of the wherein the concentration of the antibody is about 100, 150 or 200 mg/ml after reconstitution in solution. All concentrations mentioned earlier in this description can also be used for the dosage form containing. lyophilized pharmaceutical composition for reconstitution into a liquid formulation before administration to a patient.
  • Example 1 Generation and Identification of the Pcsk9 antagonist NVPLFU720
  • a stable expression cell line secreting human Pcsk9 protein was generated by transfection of HEK293 FreestyleTM cells (Invitrogen, Carlsbad, Ca). Briefly, the cells cultivated in FreestyleTM medium (Invitrogen) plus 10 % fetal calf serum in adherent mode on BioCoat flasks (Becton Dickinson) were transfected using Lipofectamine 2000TM transfection reagent and a recombinant plasmid featuring the mellittin signal sequence, the mature Pcsk9 cDNA (aa 31 -692) and a hise (SEQ ID NO:57) tag at the C-terminus of the sequence (cloned by E.Hampton, GNF, NPL 010051).
  • HPLC-ESI MS analysis of full length protein reveals a mass of 58176.0 Da which is according the expected mass from mellitin-hsPcsk9 aa31-692-His with all Cysteine residues oxidized. Part of sample is additionally N-glycosylated.
  • the contaminating protein of approx 13 kD mass resembles, most likely, the free pro-domain of the protein.
  • the corresponding homologues of Pcsk9 from mouse and cynomolgus monkey were produced in large-scale transient expression approaches using again HEK293 Freestyle cells cultivated in serum-free suspension in
  • Pcsk9 was diluted 1 : 1 with Freunds Complete Adjuvant prior to immunization of Bcl-2 transgenic mice (C57BL/6-Tgn (bcl-2) 22 wehi strain). Mice were immunized using a procedure that calls for Repetitive Immunization at Multiple Sites (RIMMS). Briefly, mice were injected with 1-3 ⁇ g of antigen at 8 specific sites proximal to peripheral lymph nodes (PLN). This procedure was repeated 6 times over a 12-day period. On Day 12, a test bleed was collected and the serum antibody titer was analyzed by ELISA. Pooled PLN were removed from high titer mice on Day 15.
  • RIMMS Repetitive Immunization at Multiple Sites
  • lymphocytes were washed twice with plain DMEM and then dissociated by passage through a .22 micron screen (Falcon #352350). The resulting lymphocytes were washed 2 additional times prior to fusion.
  • NSO/Bcl-2 myeloma cells were mixed with lymphocytes at a ration of 2.5 lymphocytes to 1 NSO cell. The cell mixture was centrifuged and 1 mL of PEG 1500 was subsequently added drop wise to the cell pellet for 1 min. After 30 seconds, 1 mL of DMEM was slowly added, and 1 min later, 19 mL of DMEM was added for 5 min.
  • Fused cells were pelleted, resuspended at a density of 2 x 10s cells/mL in HAT media (DMEM + 20 % FBS, Pen/Strep/Glu, lx NEAA, lx HAT, 0.5x HFCS), and placed at 37 oC for one h. The cells were then plated in 384-well plates at 60 / well. [0186] Ten days after fusion, hybridoma plates were screened for the presence of Pcsk9 specific antibodies. For the ELISA screen, Maxisorp 384-well plates (Nunc #464718) were coated with 50 uL of Pcsk9 (diluted to 15 ng/well in PBS) and incubated overnight at 4 oC.
  • the remaining protein was aspirated and wells were blocked with 1 % BSA in PBS. After 30 min incubation at room temperature, the wells were washed four times with PBS + 0.05 % Tween (PBST). 15 ⁇ _. of hybridoma supernatant was transferred to the ELISA plates. 15 ⁇ , of mouse serum, taken at the time of PLN removal, was diluted 1 : 1000 in PBS and added as a positive control. 50 of secondary antibody (goat anti mouse IgG - HRP (Jackson Immuno Research #115-035-071), diluted 1:5000 in PBS) was added to all wells on the ELISA plates. After incubation at room temperature for 1 h, the plates were washed eight times with PBST.
  • PBST PBS + 0.05 % Tween
  • TMB 25 ⁇ ⁇ of TMB (KPL #50-76-05) was added and after 30 min incubation at room temperature; the plates were read at an absorbance of 605 nm.CellS from positive wells were expanded into 24- well plates in HT media (DMEM + 20 % FBS, Pen/Strep/Glu, lx NEAA, lx HT, 0.5x HFCS).
  • Binding studies of Pcsk9 to the LFU720 (2 ⁇ g/mL) were carried out by capturing the mouse antibody with a rabbit anti-mouse Fey antibody (Biacore #BR-1005-14) that was previously immobilized onto a Series S CM-5 Biacore sensor chip (certified) (Biacore #BR- 1005-30). Covalent binding of the Fey capture antibody was done with the 'Amine Coupling Kit' (Biacore #BR-1000-50).
  • the capture antibody (rabbit-anti-mouse) was attached to the EDCactivated dextran surface with a 50 ⁇ g/mL anti-Fey antibody solution in 10 mM sodium acetate, pH 5 (Biacore #BR-1003-51) at a flow rate of 10 uL/min.
  • a range of concentrations of Pcsk9 from 0.5 ⁇ to 7.8 nM (2 fold serial dilution) were flowed over the captured LFU720 chip in PBS plus 100 mM NaCl, 0.005 % P20 (Biacore #BR-1000-54).
  • the resulting sensorgrams were analyzed using the Biacore S51 Evaluation Software. Data from all concentrations was fitted globally to a 1 : 1 Langmuir model.
  • TR-FRET assay was performed in 384-well white, shallow plates (Perkin Elmer, 6008280).
  • hPcsk9-AF (10.7 nM) was incubated with serial dilutions of unlabeled hPcsk9 protein, EGF-A peptide, or VP-LFU720-AX-1 antibody for 30 minutes at room temperature in 15 uL of assay buffer (20 mM HEPES, pH 7.2, 150 mM NaCl, 1 mM CaCh, 0.1% v/v Tween 20, and 0.1% w/v BSA).
  • the percentage inhibition was calculated with the following formula: 100 - [(normalized value of treated sample/averaged normalized value of untreated samples) x 100].
  • HepG2 cells were trypsinized and seeded at 6 x 10 4 cells per well in 100 ⁇ , of culture medium in flat bottomed 96-well plates (Corning, 3595) which were pre-coated with 1% v/v collagen), then incubated at 37°C in 5% CCte for 24 hours.
  • cells were treated with 100 ⁇ , of serum-free medium containing either hPcsk9 protein, EGF-A peptide and/or NVP-LFU720-AX-1 antibody. After treatment, the medium was discarded, and the cells were washed with 100 ⁇ , of PBS. To harvest the cells, 100 uL of Versine
  • the rabbit anti-hLDL-R IgG 583 exhibited approximately a 7-fold window for detection of hLDL-R on the surface of HepG2 cells as compared to normal rabbit IgG.
  • an experiment was performed using hLDL-R protein as a competitor for binding of this IgG.
  • a dose-dependent decrease in the average medium fluorescence for the anti-hLDL-R IgG 583 towards HepG2 cells was observed with increasing concentrations of hLDL-R protein.
  • anti-hLDL-R IgG 583 specifically recognizes the LDL-R on the surface of HepG2 cells as measured by FACS.
  • Future work used directly labeled anti-hLDL-R-583- Alexa 647 IgG for the FACS quantification of LDL-R on the surface of HepG2 cells.
  • HepG2 cells were trypsinized and seeded at 6 x 10 4 cells per well in 100 of culture medium in flat bottomed 96-well plates (Corning, 3595, which were pre-coated with 1% v/v collagen), then incubated at 37°C in 5% C0 2 for 24 hours. Unless otherwise stated, cells were treated with 100 uL of serum-free medium containing either hPcsk9 protein, EGF- A peptide and/or VP-LFU720-AX-1 antibody.
  • each well received 20 L of 30 Mg/mL 3,3'-dioctadecylindocarbocyanine-labeled low-density lipoprotein (Dil-LDL) (Intracell, RP-077-175) in serum-free medium and incubated at 37°C in 5% C0 2 for 2 hours.
  • the medium was removed by flicking the plates, and the cells were washed with 100 L of phosphate buffered saline (PBS without calcium or magnesium, Invitrogen, 14190-144).
  • PBS phosphate buffered saline
  • trypsin-EDTA was added to each well and incubated for 5 minutes at 37°C in 5% C0 2 .
  • FACS buffer PBS containing 5% FBS, 2 mM EDTA, and 0.2% sodium azide
  • PBS containing 5% FBS, 2 mM EDTA, and 0.2% sodium azide
  • the medium was discarded by flicking the plate, and the cells were fixed by addition of 50 uL of 1% paraformaldehyde (Electron Microscopy Sciences, 15710) in PBS per well.
  • Viable cells were gated and analyzed using a BD LSR II flow cytometer and FACSDIVA software (Becton Dickinson). The median value of Dil-LDL fluorescence was measured at excitation 488 nm and emission 575 nm, and 5000 cells were analyzed.
  • B-cells were harvested from the primary lymph nodes of animals immunized with Pcsk9 protein. Hybridoma were generated using standard PEG-mediated fusion. The resulting fusion was assayed by ELISA, and positive binders to human Pcsk9 were identified and expanded to generate supematants. There were multiple ELISA positive clones identified that were subsequently triaged by functional assays. The clone that was identified as the lead candidate is LFU720.
  • LFU720 multiple other clones were identified that failed to block the interaction of Pcsk9 and LDLr (as measured by FRET) yet when bound to Pcsk9 were able to block Pcsk9's ability to mediate LDLr degradation (as measured by LDLc uptake), including clones 21D6, 5A4-C1, and 13F1. Clone 21D6 also showed the ability to block Pcsk9 degradation of the LDLr in vivo.
  • LFU720 specificity was examined by evaluating binding in ELISA to a series of other proteins. The binding of LFU720 to three other HIS-tagged proteins was compared to binding to Pcsk9-HIS. This demonstrated that the binding to Pcsk9 is specific, and that the antibody was not binding to the HIS tag.
  • TR-FRET assay was used for determining if the anti-hPcsk9 antibody NVP- LFU720 could disrupt the interaction between hPcsk9-AF and hLDL-R-Eu labeled proteins.
  • Unlabeled hPcsk9 protein or EGF-A peptide were evaluated to demonstrate the assay could detect the disruption of the TR-FRET signal generated by interaction of hLDL-R-Eu and hPcsk9-AF labeled proteins.
  • Increasing concentrations of unlabeled hPcsk9 competed with hPcsk9-AF for binding to hLDL-R-Eu, which resulted in a decrease of the TR-FRET signal.
  • the EGF-A peptide disrupted the interaction between hLDL-R-Eu and hPcsk9-AF with an ICsoof 2.5 uM.
  • NVP-LFU720 poorly disrupted the TR-FRET signal between hPcsk9-Eu and hLDL-R-AF with an IC 5 o greater than 1000 nM, and exhibited a U-shaped response at low antibody concentrations.
  • NVP-LFU720 prevented Pcsk9-mediated LDL-R degradation on HepG2 cells treated with exogenous hPcsk9 and led to increased Dil-LDL- uptake with an ECso of 99 nM.
  • Example 2 Creation of PCSK9 Antagonist Antibodies NVP-LGT209. NVP-LGT210 and NVP-LGT211 [0201] This example describes the generation of human antibodies NVP-LGT209,
  • NVP-LGT210 and NVP LGT211 by engineering the murine monoclonal PCSK9 antagonist antibody NVP-LFU720 to have greater sequence homology to a human germline antibody.
  • NVP-LGT209, NVP-LGT210 and NVP LGT211 retain the epitope specificity, affinity, and cynomolgus macaque PCSK9 cross-reactivity of the parent murine antibody, NVP-LFU720.
  • NVP-LGT209, NVP-LGT210 and NVP LGT211 have much higher homology to the human germline sequence than the original murine antibody and should therefore be better tolerated by the human immune system.
  • Mouse monoclonal antibody LFU720 was HumaneeredTM to bring its protein sequence closer to a human germline sequence and decrease its immunogenicity.
  • Antibody HumaneeringTM generates engineered human antibodies with V-region sequences that have high homology to a human germline sequence while still retaining the specificity and affinity of the parent or reference antibody (U.S. Patent Publ. 2005/0255552 and 2006/0134098).
  • the process first identifies the minimum antigen binding specificity determinants (BSDs) in the heavy and light chain variable regions of a reference Fab (typically sequences within the heavy chain CDR3 and the light chain CDR3). As these heavy and light chain BSDs are maintained in all libraries constructed during the HumaneeringTM process, each library is epitope-focused, and the final, fully HumaneeredTM antibodies retain the epitope specificity of the original mouse antibody.
  • BSDs minimum antigen binding specificity determinants
  • full-chain libraries in which an entire light or heavy chain variable region is replaced with a library of human sequences
  • cassette libraries in which a portion of the heavy or light chain variable region of the mouse Fab is replaced with a library of human sequences
  • a bacterial secretion system is used to express members of the library as antibody Fab fragments, and the library is screened for Fabs that bind antigen using a colony-lift binding assay (CLBA). Positive clones are further characterized to identify those with the highest affinity.
  • Identified human cassettes supporting binding in the context of residual murine sequences are then combined in a final library screen to generate completely human V-regions.
  • the resulting HumaneeredTM Fabs have V-segment sequences derived from human libraries, retain the short BSD sequences identified within the CDR3 regions, and have human germline Framework 4 regions. These Fabs are converted to full IgGs by cloning the variable regions of the heavy and light chains into IgG expression vectors. Fully
  • HumaneeredTM antibodies generated in this process retain the binding specificity of the parent, murine antibody, typically have equivalent or higher affinity for antigen than the parent antibody, and have V-regions with a high degree of sequence identity compared with human germline antibody genes at the protein level.
  • V-region DNA from murine monoclonal NVP-LFU720 was amplified by RT- PCR from RNA isolated from the hybridoma cell line using standard methods.
  • Primers successfully used for PCR amplification of the heavy chain variable region from hybridoma cDNA were V H 14 (5 ' -CTTCCTGATGGC AGTGGTT-3 ' ; SEQ ID NO:58) (Chardes T, et al 1999, FEBS Letters 452(3):386-94) and HCconstant (5'-
  • the amplified heavy and light chain variable regions were sequenced. PCR was then used to amplify the V-genes and to incorporate restriction enzyme sites for cloning into KaloBios vectors: Vh into KB 1292 -His (modified version of KB 1292 that encodes a C-terminal flexible linker and 6- His (SEQ ID NO:57) tag of amino acid sequence AAGASHHHHHH (SEQ ID NO:62) on CHI) at Ncol (5') and Nhel (3'); Vk into KB1296 at Ncol (5') and BsiWl (3').
  • Fab fragments were expressed by secretion from E. coli using KaloBios expression vectors. Cells were grown in 2xYT medium to an OD600 of -0.6. Expression was induced by adding IPTG to 100 ⁇ and shaking for 4 hours at 33°C. Assembled Fab was obtained from periplasmic fractions by osmotic lysis and purification by affinity chromatography using Ni-NTA columns (HisTrap HP columns; GE Healthcare catalog #17-5247-01) according to standard methods. Fabs were eluted in buffer containing 500 mM imidazole and thoroughly dialyzed against PBS pH 7.4 without calcium and magnesium.
  • the original murine NVP-LFU720 Vh is closest to human germline sequence Vhl-02 in its CDRs, so a mixture of the two KaloBios libraries that contains Vhl subgroup members (KB 1410 and KB 1411) was used in making the full Vh library.
  • a mixture of the two KaloBios human V-segment libraries containing Vk3 subgroup members was used in making the full Vk library.
  • Vh and Vk libraries were then used as templates for the construction of cassette libraries in which only part of the parent murine V-segment is replaced by a library of human sequences.
  • Two types of cassettes were constructed by bridge PCR: front-end cassettes containing human sequences in FR1, CDR1, and the first part of FR2 were amplified from the mixture of Vhl libraries (KB 1410 and KB1411) or the mixture of Vk3 libraries (KB 1423 and KB 1424) described above as a template.
  • Middle cassettes containing human sequences in the last part of FR2, CDR2, and FR3 were amplified using the full human Vh- or Vk-region libraries described above as templates.
  • Vh cassettes had overlapping common sequences in FR2 at amino acid positions 45-49 (Kabat numbering); Vk cassettes had overlapping common sequences in FR2 at amino acid residues 35-39 (Kabat numbering).
  • front-end and middle human cassette libraries were constructed by PCR for human V-heavy 1 and V-kappa 3 isotypes.
  • Each Vh cassette library was cloned into vector KB 1292 -His at Ncol (5') and Kpnl (3'); each Vk cassette library was cloned into vector KB 1296-B (modified version of KaloBios vector KB 1296 which has a silent Hindlll site added in FR4) at Ncol (5') and Hindlll (3').
  • Resultant Vh or Vk plasmid libraries were then combined with the complementary chain from the optimized reference Fab SR038 (e.g., the Vh front-end library was combined with the optimized reference Vk vector) by digestion with BssYill and Clal and subsequent ligation to create libraries of dicistronic vectors expressing full Fabs.
  • Recombinant human or cynomolgus macaque PCSK9-His6 antigen was used for all ELISA assays. Typically, PCSK9-His6 antigen diluted in PBS pH 7.4 was bound to a 96- well microtiter plate at 300 ng/well by overnight incubation at 4°C.
  • the plate was blocked with a solution of 3% BSA in PBS for one hour at 37 °C, and then rinsed once with PBST.
  • Fab-containing induced cell medium or diluted, purified Fab (50 iL) was then added to each well. After a one-hour incubation at 37°C, the plate was rinsed three times with PBST.
  • Anti- human-kappa chain HRP conjugate (Sigma #A7164) diluted 1 :5000 in PBS (50 uL) was added to each well, and the plate was incubated for 45 min at room temperature.
  • the plate was washed three times with PBST, then 100 uL of SureBlue TMB substrate (KPL #52-00- 03) was added to each well and the plate was incubated for -10 min at room temperature. The plate was read at 650 nm in a spectrophotometer.
  • a 384- we 11 plate was coated with a panel of purified human or mouse antigens at 88 ng per well and incubated overnight at 4°C. The plate was blocked and washed as described above, then 22 ⁇ _, of purified mouse or human anti-PCSK9 antibody diluted to 2 ⁇ g/mL in PBS was added to each well. The plate was incubated for 1 hr at 37°C then washed with PBST.
  • Anti-mouse Fc antibody Jackson ImmunoResearch Labs #115-035-071
  • anti-human kappa antibody Sigma #A7164 conjugated to HRP was diluted 1 :5000 in PBS (25 uL) and added to each well. The plate was incubated for 1 hr at room temperature, then washed and developed as described above.
  • Screening of HumaneeredTM libraries of Fab fragments was carried out essentially as described in (U.S. Patent Publ. 2005/0255552 and 2006/0134098) using nitrocellulose filters coated with PCSK9-His6 at 6 ⁇ g/mL.
  • Fabs bound to the antigen-coated filter were detected using an alkaline phosphatase-conjugated anti-human kappa light chain antibody (Sigma #A3813) diluted 1 :5000 in PBST, and blots were developed with DuoLux chemiluminescent substrate for alkaline phosphatase (Vector Laboratories #SK-6605).
  • PCSK9 with C-terminal Avi- (for site-directed biotinylation) and His6 (SEQ ID NO:57) tags was generated by inserting an EcoRI restriction site between the gene encoding PCSK9 and the His6 (SEQ ID NO:57) tag in the pRS5a/PCSK9 plasmid; expresses amino acids 31-692 of PCSK9 Uniprot Accession Q8NBP7 with a C-terminal His6 (SEQ ID NO:57) tag).
  • Oligonucleotides encoding the Avi tag (amino acid sequence:
  • PCSK9-Avi-His6 was performed in the 293 Freestyle Expression System (Invitrogen), and secreted recombinant protein was purified using Ni-NTA resin (QIAGEN). Following purification, PCSK9-Avi-His6 protein was dialyzed against 10 mM Tris pH 8.0, 50 mM NaCl.
  • the protein was biotinylated in vitro with biotin-protein ligase (Avidity) according to the manufacturer's protocol. Upon completion, the reaction was dialyzed against PBS pH 7.2, and biotinylation was verified by Western blot, probing with HRP-conjugated streptavidin.
  • ECL electrochemiluminescence
  • Fully HumaneeredTM VP-LGT209, NVP-LGT210 and NVP-LGT211 antibodies were produced by co-transfection of vectors as follows into 293 Freestyle cells using 293fectin transfection reagent (Invitrogen #51 -0031 ) according to the
  • RT-PCR products from hybridoma cells that express NVP-LFU720 were sequenced, and this sequence was largely (95% or greater) verified at the protein level using a ThermoElectron LTQ-Orbitrap Mass Spectrometer.
  • the heavy and light chain variable regions of LFU720 were then cloned into KaloBios vectors in order to create the reference Fab SR032.
  • the first amino acid in NVP-LFU720 Vk had to be changed from a glutamine (Q) to a glutamic acid (E) to enable cloning into KaloBios vectors for generation of the reference Fab SR032; therefore, the SR032 Vk has glutamic acid at the first Vk position.
  • the Fab SR032 has intact murine V-regions from NVP-LFU720 fused with human constant regions and was purified from E. coli. In a dilution ELISA test of PCSK9-His6 antigen binding, the cloned SR032 reference Fab produced binding curves that were dependent on Fab concentration. In addition to the reference Fab (SR032), an optimized reference Fab, SR038, was constructed. Several framework amino acid residues in SR032 were changed to human germline in SR038.
  • the human germline residues incorporated into the optimized reference Fab SR038 in FR1 and FR3 are those specified by the PCR primers used to amplify the human V- segment repertoire and thus are present in all members of the HumaneeredTM V-region libraries.
  • the optimized reference Fab is constructed to assess whether or not any of the changes to human germline alter the properties of Fab binding. By dilution ELISA using purified Fabs, the affinities of SR032 and SR038 for recombinant PCSK9 antigen were determined to be identical, indicating that the amino acid changes in SR038 are tolerated.
  • Heavy and light chain front-end and middle cassette libraries subgroup-restricted to Vhl or Vk3 were generated and screened by CLBA.
  • front-end cassettes which supported binding to PCSK9 antigen were identified by colony-lift binding assay, but Vh middle cassettes were not.
  • Vk both front-end and middle cassettes were identified by colony lift, and in addition, some full Vk clones were identified (due to contamination of the Vk front-end library with full-chain Vk clones).
  • Many binders from each library reconfirmed in an ELISA assay on Fab-containing cell supernatants, and several Fabs from each library were purified from periplasmic fractions and rank-ordered by affinity using Forte analysis.
  • HumaneeredTM Fabs SR066, #1-2, #3-2, and #4-2 contained an "Asn-Gly" ("NG") amino acid sequence in CDRH2, which came from the original mouse antibody. This sequence is known to potentially undergo deamidation, an undesirable property for production. Therefore, at the same time as efforts were made to improve the affinity of the HumaneeredTM SR066 Fab, two mutagenic libraries were constructed in the context of SR066, in which the "N" or the "G” were substituted with every possible amino acid except the original amino acid (e.g., in the "N removal library,” "N” was substituted with every amino acid except for "N”).
  • Fab-containing cell supernatants from this library were then screened by ELISA to identify clones that no longer had the "NG” sequence but still retained the SR066 level of binding (which is less than that of the optimized reference Fab SR038).
  • the "N removal library” did not yield any Fabs that bound to the same extent as SR066, but the “G removal library” yielded Fabs with sufficient levels of binding that had a number of different substitutions at the "G” position.
  • Fabs that had "NG” to "NE,” “NA,” and “NM” changes were chosen for purification and Forte kinetic analysis and found to have similar kinetics to that of the parent Fab SR066.
  • Vh sequence of SR079 modified version of SR066 that has improved affinity up to CDRH2 was amplified and stitched to the sequence of a "G removal library” clone (containing "NE” in CDRH2) from CDRH2 through most of FR4 by bridge PCR.
  • This Vh sequence was cloned into pRS5a-hIgGl LALA + Kpnl (silent IgGl cloning vector that had been modified to add a Kpnl site in FR4 without affecting the amino acid sequence of the Vh) at Nrul (5') and Kpnl (3') to create pSR074.
  • Vk from FR1 up to partway through FR4 was amplified and cloned into pRS5a-hkappa at Agel and BsiWI to create pSR072. These vectors were verified by sequencing and co-transfected into 293 Freestyle cells.
  • This IgG (referred to as "7472") was purified from antibody-containing cell media. Surprisingly, antibody 7472 had a
  • This light chain was cloned into pRS5a-hkappa as described above to generate pJGlO and pJGOl, and a full IgG was generated by co-transfection of pSR074 (He vector) and pJGlO (Lc vector) or pJGOl (Lc vector). Together, these changes partially rescued the affinity of the HumaneeredTM antibody as determined by Biacore.
  • Table 4 shows alignments of portions of the light chains of the human antibody 7472, the parental mouse antibody LFU720, and the light chain encoded by pJGlO. This portion is identical to the light chain encoded by pJGOl.
  • the light chain vectors were used for expression of the final HumaneeredTM antibodies NVP-LGT209, NVP-LGT210 and NVP- LGT-211.
  • a Fab library was constructed to affinity mature CDR3 of the pSR074 heavy chain.
  • CDR3 residues were substituted singly with every other amino acid, excluding the original residue.
  • This library was screened by CLBA, binders were verified by ELISA, and the resultant Fabs were purified and tested by bio-layer interferometry to determine whether they had improved binding kinetics relative to the parental human Fab.
  • One CDRH3 substitution was identified which did significantly improve affinity: an A to N change (Table 5). This change was subsequently made in the context of the pSR074 heavy chain IgG expression plasmid; this new construct was called pJG04.
  • the final HumaneeredTM antibody NVP-LGT209 (LGT209) was generated by co-transfection of pJG04 (He) and pJGlO and subsequent purification.
  • the final HumaneeredTM antibody NVP- LGT210 (LGT210) was generated by co-transfection of pJG04 (He) and pJGOl and subsequent purification. This change was not introduced into antibody NVP-LGT211 (LGT211).
  • Table 5 shows alignments of heavy chain CDR3 of the human antibody 7472, the parental mouse antibody LFU720, and the heavy chain encoded by pJG04 (heavy chain vector used for expression of the final HumaneeredTM antibodies NVP-LGT209 and NVP- LGT210). The A to N substitution is underlined.
  • the binding affinities of the NVP-LGT209, NVP-LGT210 and NVP-LGT211 antibodies to human PCSK9 were determined to be between 150-190 pM as indicated in Table 6. This suggests high affinity interaction between the antibodies and PCSK9 in solution.
  • LGT209, LGT210 and LGT21 1 were evaluated for specific binding to human and cynomolgus macaque (cyno) Pcsk9.
  • This ELISA assay shows that, like the parental mouse antibody LFU720, the HumaneeredTM antibodies LGT209, LGT210 and LGT211 are able to bind both human and cyno Pcsk9 in a similar manner.
  • TMYMS SEQ ID NO:66
  • only clones that contain the original mouse residues (in bold) retain the binding kinetics of mouse Ab (as determined by biolayer interferometry analysis). Therefore, these residues within the heavy chain CDR1 play a role in binding.
  • the amino acid at position 1 can be A, D, E, N or Q.
  • the amino acid at position 4 can be V, I , L or M.
  • Positions 1-4 as QIVL (SEQ IOD NO:69) (as in the mouse parent LFU720, LGT209 and LGT211 Vk) have improved binding over positions 1-4 as EIVM (SEQ ID NO:67) (as in LGT210 Vk), as determined by Biacore.
  • Example 4 Composition of and process of making a pharmaceutical formulation of a PCSK9 monoclonal antibody
  • DS refers to drug substance (antibody before formulation) and DP to drug product.
  • the drug product will be put in final packaging form.
  • DS before lyophilization
  • DP after reconstitution
  • Example 5 A pharmaceutical formulation of LGT209
  • a pharmaceutical formulation of LGT209 was made according to the process steps of example 4 according to Table 9. The formulation was stable and found suitable for administrations to humans. Table 9 - Composition of DP and PP - formulation of LGT209
  • LGT209 CHO solution 50 mg/mL 50.00 150.0

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Obesity (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Dermatology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des formulations pharmaceutiques comprenant des antagonistes d'anticorps dirigés contre la proprotéine convertase subtilisine/kexine de type 9a ("PCSK9").
PCT/EP2012/061045 2011-06-10 2012-06-11 Formulations pharmaceutiques d'antagonistes de pcsk9 WO2012168491A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161495548P 2011-06-10 2011-06-10
US61/495,548 2011-06-10

Publications (1)

Publication Number Publication Date
WO2012168491A1 true WO2012168491A1 (fr) 2012-12-13

Family

ID=46298399

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/061045 WO2012168491A1 (fr) 2011-06-10 2012-06-11 Formulations pharmaceutiques d'antagonistes de pcsk9

Country Status (1)

Country Link
WO (1) WO2012168491A1 (fr)

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
EP2742940A1 (fr) 2012-12-13 2014-06-18 IP Gesellschaft für Management mbH Sels d'éthers aza-bicyclo-di-aryl destinés à être administrés une fois par jour, deux fois par jour ou trois fois par jour
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8859741B2 (en) 2007-08-23 2014-10-14 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8883157B1 (en) 2013-12-17 2014-11-11 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8945560B1 (en) 2014-07-15 2015-02-03 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
US8999341B1 (en) 2014-07-15 2015-04-07 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US9034332B1 (en) 2014-07-15 2015-05-19 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9045548B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9051378B1 (en) 2014-07-15 2015-06-09 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9062105B1 (en) 2014-07-15 2015-06-23 Kymab Limited Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
GB2521355A (en) * 2013-12-17 2015-06-24 Kymab Ltd Human targets I
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
US9150660B1 (en) 2014-07-15 2015-10-06 Kymab Limited Precision Medicine by targeting human NAV1.8 variants for treatment of pain
US9266961B2 (en) 2012-06-15 2016-02-23 Genentech, Inc. Anti-PCSK9 antibodies, formulations, dosing, and methods of use
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2017051273A1 (fr) * 2015-09-22 2017-03-30 Pfizer Inc. Procédé de préparation d'une formulation de protéine thérapeutique et formulation d'anticorps produite par un tel procédé
WO2017118307A1 (fr) 2016-01-05 2017-07-13 江苏恒瑞医药股份有限公司 Anticorps anti-pcsk9, fragment de liaison à l'antigène associé et application médicale associée
WO2017151783A1 (fr) 2016-03-03 2017-09-08 Regeneron Pharmaceuticals, Inc. Procédés de traitement de patients atteints d'hyperlipidémie par administration d'un inhibiteur de pcsk9 en combinaison avec un inhibiteur d'angptl3
WO2017177181A1 (fr) 2016-04-08 2017-10-12 Regeneron Pharmaceuticals, Inc. Méthodes de traitement de l'hyperlipidémie à l'aide d'un inhibiteur d'angptl8 et d'un inhibiteur d'angptl3
WO2017189813A1 (fr) 2016-04-28 2017-11-02 Regeneron Pharmaceuticals, Inc. Méthodes de traitement de patients présentant une hypercholestérolémie familiale
WO2018189705A1 (fr) 2017-04-13 2018-10-18 Cadila Healthcare Limited Vaccin pcsk9 à base de nouveaux peptides
WO2018225041A1 (fr) 2017-06-09 2018-12-13 Sanofi Biotechnology Méthodes de traitement de l'hyperlipidémie chez des patients diabétiques par administration d'un inhibiteur de pcsk9
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2019173530A1 (fr) 2018-03-06 2019-09-12 Sanofi Biotechnology Utilisation d'un inhibiteur de pcsk9 pour réduire le risque cardiovasculaire
WO2020150473A2 (fr) 2019-01-18 2020-07-23 Dogma Therapeutics, Inc. Inhibiteurs de pcsk9 et leurs procédés d'utilisation
US10793643B2 (en) 2015-12-31 2020-10-06 Jiangsu Hengrui Medicine Co., Ltd. PCSK9 antibody, antigen-binding fragment thereof, and medical application thereof
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
JP2020532491A (ja) * 2017-06-30 2020-11-12 江▲蘇▼恒瑞医▲薬▼股▲フン▼有限公司Jiangsu Hengrui Medicine Co., Ltd. Pcsk−9抗体を含む医薬組成物及びその使用
WO2022032137A1 (fr) 2020-08-07 2022-02-10 Regeneron Pharmaceuticals, Inc. Méthodes de traitement de l'hypercholestérolémie réfractaire impliquant un inhibiteur d'angptl3
RU2779389C2 (ru) * 2015-09-22 2022-09-06 Пфайзер Инк. Способ получения терапевтического белкового препарата и препарат на основе антител, полученный по такому способу
US11673967B2 (en) 2011-07-28 2023-06-13 Regeneron Pharmaceuticals, Inc. Stabilized formulations containing anti-PCSK9 antibodies
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11904017B2 (en) 2015-08-18 2024-02-20 Regeneron Pharmaceuticals, Inc. Methods for reducing or eliminating the need for lipoprotein apheresis in patients with hyperlipidemia by administering alirocumab

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US20050255552A1 (en) 2004-01-20 2005-11-17 Kalobios, Inc. Antibody specificity transfer using minimal essential binding determinants
US20060134098A1 (en) 2004-11-16 2006-06-22 Kalobios, Inc. Immunoglobulin variable region cassette exchange
US20070020685A1 (en) 2005-07-22 2007-01-25 Kalobios Pharmaceuticals, Inc. Secretion of antibodies without signal peptides from bacteria
WO2008057459A2 (fr) * 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2009026558A1 (fr) * 2007-08-23 2009-02-26 Amgen Inc. Protéines de liaison à un antigène pour proprotéine convertase subtilisine kexine de type 9 (pcsk9)
WO2009100297A1 (fr) * 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de pcsk9 1d05
WO2010068526A1 (fr) * 2008-12-12 2010-06-17 Merck Sharp & Dohme Corp. Immunodosage de pcsk9
WO2011072263A1 (fr) 2009-12-11 2011-06-16 Irm Llc Antagonistes de pcsk9

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US20050255552A1 (en) 2004-01-20 2005-11-17 Kalobios, Inc. Antibody specificity transfer using minimal essential binding determinants
US20060134098A1 (en) 2004-11-16 2006-06-22 Kalobios, Inc. Immunoglobulin variable region cassette exchange
US20070020685A1 (en) 2005-07-22 2007-01-25 Kalobios Pharmaceuticals, Inc. Secretion of antibodies without signal peptides from bacteria
WO2008057459A2 (fr) * 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2009026558A1 (fr) * 2007-08-23 2009-02-26 Amgen Inc. Protéines de liaison à un antigène pour proprotéine convertase subtilisine kexine de type 9 (pcsk9)
WO2009100297A1 (fr) * 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de pcsk9 1d05
WO2010068526A1 (fr) * 2008-12-12 2010-06-17 Merck Sharp & Dohme Corp. Immunodosage de pcsk9
WO2011072263A1 (fr) 2009-12-11 2011-06-16 Irm Llc Antagonistes de pcsk9

Non-Patent Citations (88)

* Cited by examiner, † Cited by third party
Title
"2009 Physicians' Desk Reference (PDR", 2008
"Art, Science and Technology of Pharmaceutical Compounding", 2008
"Goodman and Gilman's The Pharmacological Basis of Therapeutics", 2006, MCGRAW-HILL
"Remington's Science and Practice of Pharmacy", 2005
ABIFADEL ET AL., HUM MUTAT, vol. 30, no. 4, 2009, pages 520 - 9
ABIFADEL ET AL., JMED GENET, vol. 45, no. 12, 2008, pages 780 - 6
AL-LAZIKANI ET AL., J.MOL.BIOL., vol. 273, 1997, pages 927 - 748
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUC. ACIDS RES., vol. 25, 1977, pages 3389 - 3402
ANN L DAUGHERTY AND RANDALL J MRSNY ED - STEVEN J SHIRE ET AL: "Formulation and Delivery Issues for Monoclonal Antibody Therapeutics", 1 January 2010, CURRENT TRENDS IN MONOCLONAL ANTIBODY DEVELOPMENT AND MANUFACTURING, SPRINGER, US, PAGE(S) 103 - 129, ISBN: 978-0-387-76642-3, XP009133774 *
AUSUBEL ET AL., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, 1995
BATZER ET AL., NUCLEIC ACID RES., vol. 19, 1991, pages 5081
BAXTER; WEBB, NATURE REVIEWS DRUG DISCOVERY, vol. 8, 2009, pages 308 - 320
BAYS; STEIN, EXPERT OPIN PHARMACOTHER, vol. 4, no. 11, 2003, pages 1901 - 38
BIRCH ET AL., JMED CHEM, vol. 52, no. 6, 2009, pages 1558 - 68
BIRCH, JMED CHEM, vol. 52, no. 6, 2009, pages 1558 - 68
BOND ET AL., J MOL BIOL., vol. 332, 2003, pages 643 - 55
BROOKS ET AL., CURR PSYCHIATRY REP, vol. 11, no. 1, 2009, pages 33 - 40
BURNETT; HOOPER, CLIN BIOCHEM REV, vol. 29, no. 1, 2008, pages 11 - 26
BURNETT; HOOPER, CLIN BIOCHEM REV., vol. 29, no. 1, 2008, pages 11 - 26
CHANG ET AL., CURR OPIN DRUG DISCO DEVEL, vol. 5, no. 4, 2002, pages 562 - 70
CHARDES T ET AL., FEBS LETTERS, vol. 452, no. 3, 1999, pages 386 - 94
CHOTHIA ET AL., J. MOL. BIOL., vol. 227, 1992, pages 799 - 817
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877 - 883
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
COLE ET AL.: "Monoclonal Antibodies and Cancer Therapy", 1985, ALAN R. LISS, INC., pages: 77 - 96
CREIGHTON, PROTEINS, 1984
DUMOULIN ET AL., NATURE STRUCT. BIOL., vol. 11, 2002, pages 500 - 515
EXP CLIN IMMUNOGENET., vol. 18, no. 4, 2001, pages 242 - 54
FASANO ET AL., ATHEROSCLEROSIS, vol. 203, no. 1, 2009, pages 166 - 71
GHAHROUDI ET AL., FEBS LETTERS, vol. 414, 1997, pages 521 - 526
GIUDICELLI ET AL., NUCLEIC ACIDS RES., vol. 33, 1 January 2005 (2005-01-01), pages D256 - 61
HARLOW; LANE: "Using Antibodies, A Laboratory Manual", 1998
HENDERSON, J CLIN PSYCHIATRY, vol. 69, no. 2, 2008, pages E04
HENIKOFF; HENIKOFF, PROC. NATL. ACAD. SCI. USA, vol. 89, 1989, pages 10915
HEZAREH ET AL., J VIROL, vol. 75, no. 24, pages 12161 - 8
JOHNSON ET AL., NUCLEIC ACIDS RES., vol. 29, 2001, pages 205 - 206
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1983, U.S. DEPT. HEALTH AND HUMAN SERVICES
KABAT ET AL.: "Sequences of Proteins oflmmunological Interest", 1983, U.S. DEPT. HEALTH AND HUMAN SERVICES
KARLIN; ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5787
KASTELEIN, INT J CLIN PRACT SUPPL, March 2003 (2003-03-01), pages 45 - 50
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497
KOSTELNY ET AL., J. IMMUNOL., vol. 148, 1992, pages 1547 - 1553
KOZBOR ET AL., IMMUNOLOGY TODAY, vol. 4, 1983, pages 72
KUBY: "Immunology", 2000, W.H. FREEMAN & CO.
LE MAY ET AL., ARTERIOSCLER THROMB VASC BIOL, vol. 29, no. 5, 2009, pages 684 - 90
LEE ET AL., JMICROBIOL BIOTECHNOL, vol. 18, no. 11, 2008, pages 1785 - 8
LEFRANC, EXP CLIN IMMUNOGENET., vol. 18, no. 2, 2001, pages 100 - 16
LEFRANC, EXP CLIN IMMUNOGENET., vol. 18, no. 3, 2001, pages 161 - 74
LEFRANC, M.P., NUCLEIC ACIDS RES., vol. 29, 2001, pages 207 - 209
LI ET AL., RECENT PAT DNA GENE SEQ
LI HAI ET AL: "Recent patents on PCSK9: a new target for treating hypercholesterolemia", RECENT PATENTS ON DNA & GENE SEQUENCES,, vol. 3, no. 3, 1 November 2009 (2009-11-01), pages 201 - 212, XP002619049, ISSN: 1872-2156, DOI: 10.2174/187221509789318388 *
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 745
MARKS ET AL., BIOTECHNOLOGY, vol. 10, 1992, pages 779 - 783
MARTIN ET AL., METHODS ENZYMOL., vol. 203, 1991, pages 121 - 153
MARTIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 9268 - 9272
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 554
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
MORRISON, ADV. IMMUNOL., vol. 44, 1988, pages 65 - 92
NEEDLEMAN; WUNSCH, J MOL. BIOL., vol. 48, 1970, pages 443
NI YAN G ET AL: "A PCSK9 C-terminal Domain Binding Fab Inhibits PCSK9 Internalization and Restores LDL-uptake", CIRCULATION, LIPPINCOTT WILLIAMS & WILKINS, US, vol. 120, no. 18, Suppl. 2, 1 November 2009 (2009-11-01), pages S477, XP008121212, ISSN: 0009-7322 *
NI YAN G ET AL: "A proprotein convertase subtilisin-like/kexin type 9 (PCSK9) C-terminal domain antibody antigen-binding fragment inhibits PCSK9 internalization and restores low density lipoprotein uptake", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, INC, BETHESDA, MD, USA, vol. 285, no. 17, 23 April 2010 (2010-04-23), pages 12882 - 12891, XP002619048, ISSN: 1083-351X, [retrieved on 20100219], DOI: 10.1074/JBC.M110.113035 *
OH ET AL., ARCH PHARM RES, vol. 32, no. 1, 2009, pages 43 - 7
OHTSUKA ET AL., J BIOL. CHEM., vol. 260, 1985, pages 2605 - 2608
PADLAN, MOLEC. IMMUN., vol. 28, 1991, pages 489 - 498
PADLAN, MOLEC. IMMUN., vol. 31, no. 3, 1994, pages 169 - 217
PEARSON; LIPMAN, PROC. NAT'L. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PIEHLER ET AL., JLMMUNOL METHODS, vol. 201, 1997, pages 189 - 206
POIRIER ET AL., JBIOL CHEM, 2009
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
RASHID S ET AL., PROC NATL ACAD SCI, vol. 102, 2005, pages 5374 - 5379
REES ET AL.: "Protein Structure Prediction", 1996, OXFORD UNIVERSITY PRESS, pages: 141 - 172
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
ROSSOLINI ET AL., MOL. CELL. PROBES, vol. 8, 1994, pages 91 - 98
RUIZ ET AL., NUCLEIC ACIDS RES., vol. 28, 2000, pages 219 - 221
SEIDAH, EXPERT OPIN THER TARGETS, vol. 13, no. 1, 2009, pages 19 - 28
SMITH; WATERMAN, ADV. APPL. MATH., vol. 2, 1970, pages 482C
SONGSIVILAI; LACHMANN, CLIN. EXP. IMMUNOL., vol. 79, 1990, pages 315 - 321
SUDHOP ET AL., DRUGS, vol. 62, no. 16, 2002, pages 2333 - 47
TENENBAUM ET AL., ADV CARDIOL, vol. 45, 2008, pages 127 - 53
TOMKIN, DIABETES CARE, vol. 31, no. 2, 2008, pages S241 - S248
TOMODA; OMURA, PHARMACOL THER, vol. 115, no. 3, 2007, pages 375 - 89
VAUGHAN; SOLLAZZO, COMB CHEM HIGH THROUGHPUT SCREEN., vol. 4, 2001, pages 417 - 30
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
WANG W ET AL: "ANTIBODY STRUCTURE, INSTABILITY, AND FORMULATION", JOURNAL OF PHARMACEUTICAL SCIENCES, AMERICAN PHARMACEUTICAL ASSOCIATION, WASHINGTON, US, vol. 96, no. 1, 1 January 2007 (2007-01-01), pages 1 - 26, XP009084505, ISSN: 0022-3549, DOI: 10.1002/JPS.20727 *
WOODLE, TRANSPLANTATION, vol. 68, no. 5, 1999, pages 608 - 616
XU ET AL., CELL IMMUNOL, vol. 200, no. 1, 2000, pages 16 - 26

Cited By (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8859741B2 (en) 2007-08-23 2014-10-14 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9056915B2 (en) 2007-08-23 2015-06-16 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9493576B2 (en) 2007-08-23 2016-11-15 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9045547B2 (en) 2007-08-23 2015-06-02 Amgen Inc. Methods of using antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9920134B2 (en) 2007-08-23 2018-03-20 Amgen Inc. Monoclonal antibodies to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8981064B2 (en) 2007-08-23 2015-03-17 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8889834B2 (en) 2007-08-23 2014-11-18 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8871914B2 (en) 2007-08-23 2014-10-28 Amgen, Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8871913B2 (en) 2007-08-23 2014-10-28 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8883983B2 (en) 2007-08-23 2014-11-11 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US11673967B2 (en) 2011-07-28 2023-06-13 Regeneron Pharmaceuticals, Inc. Stabilized formulations containing anti-PCSK9 antibodies
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9266961B2 (en) 2012-06-15 2016-02-23 Genentech, Inc. Anti-PCSK9 antibodies, formulations, dosing, and methods of use
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
EP3251673A1 (fr) 2012-12-13 2017-12-06 IP Gesellschaft für Management mbH Polythérapie comportant un inhibiteur de cdk4/6 et un inhibiteur de pi3k à utiliser dans le traitement du cancer
EP2742940A1 (fr) 2012-12-13 2014-06-18 IP Gesellschaft für Management mbH Sels d'éthers aza-bicyclo-di-aryl destinés à être administrés une fois par jour, deux fois par jour ou trois fois par jour
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US9040052B1 (en) 2013-12-17 2015-05-26 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US11434305B2 (en) 2013-12-17 2022-09-06 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
GB2521355A (en) * 2013-12-17 2015-06-24 Kymab Ltd Human targets I
US8883157B1 (en) 2013-12-17 2014-11-11 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US10618971B2 (en) 2013-12-17 2020-04-14 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US10611849B2 (en) 2013-12-17 2020-04-07 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11773175B2 (en) 2014-03-04 2023-10-03 Kymab Limited Antibodies, uses and methods
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US8945560B1 (en) 2014-07-15 2015-02-03 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
US9303089B2 (en) 2014-07-15 2016-04-05 Kymab Limited Methods of treating anaemia
US9439963B2 (en) 2014-07-15 2016-09-13 Kymab Limited Methods of treating anaemia
US11555066B2 (en) 2014-07-15 2023-01-17 Kymab Limited Precision medicine for cholesterol treatment
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9023359B1 (en) 2014-07-15 2015-05-05 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US9045548B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9187562B1 (en) 2014-07-15 2015-11-17 Kymab Limited Methods for treating anaemia
US9051378B1 (en) 2014-07-15 2015-06-09 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9150660B1 (en) 2014-07-15 2015-10-06 Kymab Limited Precision Medicine by targeting human NAV1.8 variants for treatment of pain
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US9109034B1 (en) 2014-07-15 2015-08-18 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9034331B1 (en) 2014-07-15 2015-05-19 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9428578B2 (en) 2014-07-15 2016-08-30 Kymab Limited Methods of treating anaemia
US8999341B1 (en) 2014-07-15 2015-04-07 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US10711059B2 (en) 2014-07-15 2020-07-14 Kymab Limited Methods for treating neurodegenerative diseases using anti-PD-L1 antibodies
US9062105B1 (en) 2014-07-15 2015-06-23 Kymab Limited Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
US9394568B2 (en) 2014-07-15 2016-07-19 Kymab Limited Methods of treating anaemia
US9068012B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9034332B1 (en) 2014-07-15 2015-05-19 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US10618955B2 (en) 2014-07-15 2020-04-14 Kymab Limited Methods for treating neurodegenerative disease using anti-PD-1 antibodies
US11904017B2 (en) 2015-08-18 2024-02-20 Regeneron Pharmaceuticals, Inc. Methods for reducing or eliminating the need for lipoprotein apheresis in patients with hyperlipidemia by administering alirocumab
AU2016329034B2 (en) * 2015-09-22 2019-05-23 Pfizer Inc. Method of preparing a therapeutic protein formulation and antibody formulation produced by such a method
WO2017051273A1 (fr) * 2015-09-22 2017-03-30 Pfizer Inc. Procédé de préparation d'une formulation de protéine thérapeutique et formulation d'anticorps produite par un tel procédé
RU2779389C2 (ru) * 2015-09-22 2022-09-06 Пфайзер Инк. Способ получения терапевтического белкового препарата и препарат на основе антител, полученный по такому способу
US10793643B2 (en) 2015-12-31 2020-10-06 Jiangsu Hengrui Medicine Co., Ltd. PCSK9 antibody, antigen-binding fragment thereof, and medical application thereof
WO2017118307A1 (fr) 2016-01-05 2017-07-13 江苏恒瑞医药股份有限公司 Anticorps anti-pcsk9, fragment de liaison à l'antigène associé et application médicale associée
WO2017151783A1 (fr) 2016-03-03 2017-09-08 Regeneron Pharmaceuticals, Inc. Procédés de traitement de patients atteints d'hyperlipidémie par administration d'un inhibiteur de pcsk9 en combinaison avec un inhibiteur d'angptl3
WO2017177181A1 (fr) 2016-04-08 2017-10-12 Regeneron Pharmaceuticals, Inc. Méthodes de traitement de l'hyperlipidémie à l'aide d'un inhibiteur d'angptl8 et d'un inhibiteur d'angptl3
WO2017189813A1 (fr) 2016-04-28 2017-11-02 Regeneron Pharmaceuticals, Inc. Méthodes de traitement de patients présentant une hypercholestérolémie familiale
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
WO2018189705A1 (fr) 2017-04-13 2018-10-18 Cadila Healthcare Limited Vaccin pcsk9 à base de nouveaux peptides
WO2018225041A1 (fr) 2017-06-09 2018-12-13 Sanofi Biotechnology Méthodes de traitement de l'hyperlipidémie chez des patients diabétiques par administration d'un inhibiteur de pcsk9
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
RU2782792C2 (ru) * 2017-06-30 2022-11-02 Цзянсу Хэнжуй Медисин Ко., Лтд. Фармацевтическая композиция, содержащая антитела к pcsk-9, и ее применение
EP3647325A4 (fr) * 2017-06-30 2021-03-17 Shanghai Hengrui Pharmaceutical Co., Ltd Composition pharmaceutique comprenant un anticorps pcsk-9 et son utilisation
JP2020532491A (ja) * 2017-06-30 2020-11-12 江▲蘇▼恒瑞医▲薬▼股▲フン▼有限公司Jiangsu Hengrui Medicine Co., Ltd. Pcsk−9抗体を含む医薬組成物及びその使用
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
WO2019173530A1 (fr) 2018-03-06 2019-09-12 Sanofi Biotechnology Utilisation d'un inhibiteur de pcsk9 pour réduire le risque cardiovasculaire
TWI840376B (zh) 2018-06-21 2024-05-01 美商默沙東有限責任公司 Pcsk9拮抗劑化合物
WO2020150473A2 (fr) 2019-01-18 2020-07-23 Dogma Therapeutics, Inc. Inhibiteurs de pcsk9 et leurs procédés d'utilisation
WO2022032137A1 (fr) 2020-08-07 2022-02-10 Regeneron Pharmaceuticals, Inc. Méthodes de traitement de l'hypercholestérolémie réfractaire impliquant un inhibiteur d'angptl3

Similar Documents

Publication Publication Date Title
WO2012168491A1 (fr) Formulations pharmaceutiques d'antagonistes de pcsk9
US8710192B2 (en) PCSK9 antagonists
WO2012170607A2 (fr) Utilisation d'antagonistes de pcsk9
US20130315927A1 (en) Pcsk9 antagonists
JP6114326B2 (ja) 組織因子経路阻害因子に対する抗体
CN107922507B (zh) 抗pcsk9抑制性抗体用来治疗接受脂蛋白单采的高脂血症患者
JP7110199B2 (ja) 抗第XI/XIa因子抗体による処置法
US20190309089A1 (en) Method of treating atherosclerosis
JP7032662B2 (ja) Pcsk9抗体、その抗原結合フラグメント及び医薬用途
WO2022000046A1 (fr) Formulation à haute concentration de protéines de liaison à l'antigène du facteur xii
JP7104108B2 (ja) ヘテロ接合性家族性高コレステロール血症(heFH)を有する患者を処置するための方法
US20230406942A1 (en) Igf1r antibodies
TW202246350A (zh) 血漿激肽釋放素抗體及其用途

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: 12727644

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: 12727644

Country of ref document: EP

Kind code of ref document: A1