WO2010029513A2 - Pcsk9 antagonists - Google Patents
Pcsk9 antagonists Download PDFInfo
- Publication number
- WO2010029513A2 WO2010029513A2 PCT/IB2009/053990 IB2009053990W WO2010029513A2 WO 2010029513 A2 WO2010029513 A2 WO 2010029513A2 IB 2009053990 W IB2009053990 W IB 2009053990W WO 2010029513 A2 WO2010029513 A2 WO 2010029513A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- pcsk9
- amino acid
- antibodies
- seq
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/40—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/3955—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/06—Antihyperlipidemics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2299/00—Coordinates from 3D structures of peptides, e.g. proteins or enzymes
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
Definitions
- the present invention relates to antibodies, e.g., full length antibodies or antigen- binding portions thereof, peptides, and aptamers that antagonize the activity of extracellular proprotein convertase subtilisin kexin type 9 (PCSK9), including its interaction with the low density lipoprotein (LDL) receptor (LDLR). More specifically, the invention relates to compositions comprising antagonist PCSK9 antibodies, peptides, and/or aptamers and methods of using these antibodies and/or peptides and/or aptamers as a medicament.
- PCSK9 proprotein convertase subtilisin kexin type 9
- LDLR low density lipoprotein receptor
- the antagonist PCSK9 antibodies, peptides, and aptamers can be used therapeutically to lower LDL-cholesterol levels in blood, and can be used in the prevention and/or treatment of cholesterol and lipoprotein metabolism disorders, including familial hypercholesterolemia, atherogenic dyslipidemia, atherosclerosis, and, more generally, cardiovascular disease (CVD).
- CVD cardiovascular disease
- Atherosclerosis is a disease of the arteries and is responsible for coronary heart disease associated with many deaths in industrialized countries.
- Several risk factors for coronary heart disease have now been identified: dyslipidemias, hypertension, diabetes, smoking, poor diet, inactivity and stress.
- VLDL very low density lipoprotein
- LDL hypercholesterolemia
- PCSK9 also known as NARC-1 , was identified as a protein with a genetic mutation in some forms of familial hypercholesterolemia. PCSK9 is synthesized as a zymogen that undergoes autocatalytic processing at the motif LVFAQ in the endoplasmic reticulum. Population studies have shown that some PCSK9 mutations are "gain-of-function" and are found in individuals with autosomal dominant hypercholesterolemia, while other "loss-of-function” (LOF) mutations are linked with reduced plasma cholesterol.
- LEF Loss-of-function
- PCSK9 is secreted into the plasma predominantly by hepatocytes. Genetic modulation of PCSK9 in mice confirmed the ability of PCSK9 to regulate blood lipids, and suggested that it acts to down-regulate hepatic LDLR protein levels.
- PCSK9 down-regulates LDLR protein
- PCSK9 may act both within the hepatocyte and as a secreted ligand for LDLR.
- extracellular PCSK9 binds to cell surface LDLR and promotes LDLR degradation at an intracellular site.
- PCSK9 could interact with the LDLR when the two proteins are translated within the endoplasmic reticulum (ER) and traffic through endosomal compartments towards the cell membrane.
- This invention relates to antagonist antibodies, peptides, and aptamers that selectively interact with and inhibit PCSK9 function. It is demonstrated for the first time that certain PCSK9 antagonists are effective in vivo to lower blood cholesterol.
- the invention provides an isolated antagonist of PCSK9 which comprises an antibody, a peptide, or an aptamer, which interacts with PCSK9 and when administered to a subject lowers the LDL-cholesterol level in blood of said subject.
- the antagonist can be an antibody, for example, a monoclonal antibody or human, humanized, or chimeric antibody.
- the invention provides an isolated anti-PCSK9 antibody which specifically binds to PCSK9 and which is a full antagonist of the PCSK9-mediated effect on LDLR levels when measured in vitro using the LDLR down regulation assay in Huh7 cells disclosed herein.
- the invention provides an isolated antibody which antagonizes the extracellular interaction of PCSK9 with the LDLR, as measured by PCSK9 binding to the LDLR in vitro, and, when administered to a subject, lowers the LDL-cholesterol level in blood of said subject.
- the antibody recognizes an epitope on human PCSK9 that overlaps with more than about 75% of the surface on PCSK9 that interacts with the EGF-like domain of the LDLR as described in Kwon et al., 2008, PNAS, 105:1820-1825.
- the invention provides an antibody that recognizes a first epitope of PCSK9 that overlaps with a second epitope that is recognized by a monoclonal antibody selected from the group consisting of 5A10, which is produced by a hybridoma cell line deposited with the American Type Culture Collection and assigned accession number PTA-8986; 4A5, which is produced by a hybridoma cell line deposited with the American Type Culture Collection and assigned accession number PTA-8985; 6F6, which is produced by a hybridoma cell line deposited with the American Type Culture Collection and assigned accession number PTA-8984, and 7D4, which is produced by a hybridoma cell line deposited with the American Type Culture Collection and assigned accession number PTA-8983.
- 5A10 which is produced by a hybridoma cell line deposited with the American Type Culture Collection and assigned accession number PTA-8986
- 4A5 which is produced by a hybridoma cell line deposited with the American Type Culture Collection and assigned accession number PTA-8985
- the invention provides an antibody to human PCSK9, wherein the antibody recognizes an epitope on human PCSK9 comprising amino acid residues 153-155, 194, 195, 197, 237-239, 367, 369, 374-379 and 381 of the PCSK9 amino acid sequence of SEQ ID NO: 53.
- the antibody epitope on human PCSK9 does not comprise one or more of amino acid residues 71 , 72, 150-152, 187- 192, 198-202, 212, 214-217, 220-226, 243, 255-258, 317, 318, 347-351 , 372, 373, 380, 382, and 383.
- the invention provides an antibody which specifically binds PCSK9 comprising a VH complementary determining region one (CDR1 ) having the amino acid sequence shown in SEQ ID NO:8 (SYYMH), a VH CDR2 having the amino acid sequence shown in SEQ ID NO:9 (EISPFGGRTNYNEKFKS), and/or VH CDR3 having the amino acid sequence shown in SEQ ID NO:10 (ERPLYASDL), or a variant thereof having one or more conservative amino acid substitutions in said sequences of CDR1 , CDR2, and/or CDR3, wherein the variant retains essentially the same binding specificity as the CDR defined by said sequences.
- the variant comprises up to about ten amino acid substitutions and, more preferably, up to about four amino acid substitutions.
- the invention is further directed to an antibody comprising a VL CDR1 having the amino acid sequence shown in SEQ ID NO:11 (RASQGISSALA), a CDR2 having the amino acid sequence shown in SEQ ID NO:12 (SASYRYT), and/or CDR3 having the amino acid sequence shown in SEQ ID NO:13 (QQRYSLWRT), or a variant thereof having one or more conservative amino acid substitutions in said sequences of CDR1 , CDR2, and/or CDR3, wherein the variant retains essentially the same binding specificity as the CDR1 defined by said sequences.
- the variant comprises up to about ten amino acid substitutions and, more preferably, up to about four amino acid substitutions.
- the invention provides an antibody comprising specific VL CDR1 , CDR2, and/or CDR3 sequences, or a variant thereof having one or more conservative amino acid substitutions in CDR1 , CDR2, and/or CDR3 and further comprising a VH complementary determining region CDR1 having the amino acid sequence shown in SEQ ID NO:59, 60, or 8, a VH CDR2 having the amino acid sequence shown in SEQ ID NO:61 or 9, and/or VH CDR3 having the amino acid sequence shown in SEQ ID NO:10, or a variant thereof having one or more conservative amino acid substitutions in said sequences of CDR1 , CDR2, and/or CDR3, wherein the variant retains essentially the same binding specificity as the CDR1 , CDR2, and/or CDR3 defined by said sequences.
- the variant comprises up to about twenty amino acid substitutions and, more preferably, up to about eight amino acid substitutions.
- the antibody of the invention has a variable heavy chain sequence comprising or consisting of SEQ ID NO: 54 and a variable light chain sequence comprising or consisting of SEQ ID NO: 53.
- the invention also provides a humanized antibody comprising polypeptides selected from the groups consisting of SEQ ID NO:14, SEQ ID NO:15, or both SEQ ID NO:14 and SEQ ID NO:15, or a variant thereof having one or more conservative amino acid substitutions in said sequences, wherein the variant retains essentially the same binding specificity as the antibody defined by said sequence(s). It also includes an antibody lacking a terminal lysine on the heavy chain, as this is normally lost in a proportion of antibodies during manufacture.
- the variant comprises up to about twenty amino acid substitutions and more preferably, up to about eight amino acid substitutions.
- the antibody further comprises an immunologically inert constant region, and/or the antibody has an isotype that is selected from the group consisting of lgG2, IgG 4 , lgG2 ⁇ a, IgG 4 Ab, IgG 4 Ac, IgG 4 S228P, lgG 4 ⁇ b S228P and lgG 4 ⁇ c S228P.
- the constant region is aglycosylated Fc.
- the invention provides a method for reducing a level of LDL, LDL-cholesterol, or total cholesterol in blood, serum, or plasma of a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an antagonist of the invention.
- the invention provides a therapeutically effective amount of an antagonist of the invention for use in reducing a level of LDL, LDL-cholesterol, or total cholesterol in blood, serum, or plasma of a subject in need thereof.
- the invention further provides the use of a therapeutically effective amount of an antagonist of the invention in the manufacture of a medicament for reducing a level of LDL, LDL- cholesterol, or total cholesterol in blood, serum, or plasma of a subject in need thereof.
- the invention provides a method of preparing an antibody which specifically binds PCSK9, which comprises: a) providing a PCSK9- negative host animal; b) immunizing said PCSK9-negative host animal with PCSK9; and c) obtaining an antibody.
- the invention also comprises a method for reducing the level of LDL in blood of a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the antibody prepared according to the invention.
- the subject can be further treated by administering a statin.
- the subject is a human subject.
- the antibody is administered in a formulation as a sterile aqueous solution having a pH that ranges from about 5.0 to about 6.5 and comprising from about 1 mg/ml to about 200 mg/ml of antibody, from about 1 millimolar to about 100 millimolar of histidine buffer, from about 0.01 mg/ml to about 10 mg/ml of polysorbate 80, from about 100 millimolar to about 400 millimolar of trehalose, and from about 0.01 millimolar to about 1.0 millimolar of disodium EDTA dihydrate.
- a sterile aqueous solution having a pH that ranges from about 5.0 to about 6.5 and comprising from about 1 mg/ml to about 200 mg/ml of antibody, from about 1 millimolar to about 100 millimolar of histidine buffer, from about 0.01 mg/ml to about 10 mg/ml of polysorbate 80, from about 100 millimolar to about 400 millimolar of trehalose, and from about 0.01 millimolar to about 1.0 millim
- the invention provides a therapeutically effective amount of the antibody prepared according to the invention for use in reducing the level of LDL in blood of a subject in need thereof.
- the invention further provides the use of a therapeutically effective amount of the antibody prepared according to the invention in the manufacture of a medicament for reducing the level of LDL in blood of a subject in need thereof.
- the therapeutically effective amount can optionally be combined with a therapeutically effective amount of a statin.
- the invention provides a hybridoma cell line that produces a PCSK9-specific antibody or an antigen-binding portion thereof, wherein the hybridoma cell line is selected from the group consisting of: 4A5 having an ATCC Accession No. of PTA-8985; 5A10 having an ATCC Accession No. of PTA-8986; 6F6 having an ATCC Accession No. of PTA-8984; and 7D4 having an ATCC Accession No. of PTA-8983.
- the invention provides cell line that recombinant ⁇ produces an antibody which specifically binds to PCSK9 and comprises a heavy chain variable region (VH) complementary determining region one (CDR1 ) having the amino acid sequence shown in SEQ ID NO:8, 59, or 60, a VH CDR2 having the amino acid sequence shown in SEQ ID NO:9 or 61 , and/or VH CDR3 having the amino acid sequence shown in SEQ ID NO:10, or a variant thereof having one or more conservative amino acid substitutions in CDR1 , CDR2, and/or CDR3, and/or comprises a light chain variable region (VL) CDR1 having the amino acid sequence shown in SEQ
- VL CDR3 having the amino acid sequence shown in SEQ ID NO:13, or a variant thereof having one or more conservative amino acid substitutions in CDR1 , CDR2, and/or
- the cell line recombinantly produces an antibody comprising SEQ ID NO: 53 and/or 54, and, more preferably, SEQ ID NO: 14 and/or 15.
- FIG 1 illustrates the effect of anti-PCSK9 antagonistic monoclonal antibodies
- 6F6, 7D4.4 is a subclone of 7D4, 4A5.G3 is a subclone of 4A5, and 5A10.B8 is a subclone of 5A10.
- Figure 2 illustrates the dose-response of anti-PCSK9 antagonist monoclonal antibodies 6F6.G10.3, 7D4.4, 4A5.G3, 5A10.B8, negative control antibody 42H7, and PBS to block the binding of recombinant biotinylated human PCSK9 (A) and mouse PCSK9 (B) to immobilized recombinant LDLR extracellular domain in vitro.
- Figure 3 illustrates the dose-response of anti-PCSK9 monoclonal antagonist antibodies 6F6.G10.3, 7D4.4, 4A5.G3 and 5A10.B8 to block binding of recombinant biotinylated human PCSK9 (30 nM) to Europium labeled recombinant LDLR extracellular domain (10 nM) in solution at neutral pH in vitro.
- Figure 4 illustrates comparative epitope binding of anti-PCSK9 antibodies.
- Figure 5 illustrates Western blots of binding of anti-PCSK9 antibodies to serum PCSK9 from different species.
- Figure 6 illustrates the effect of anti-PCSK9 monoclonal antibody 7D4 on blood cholesterol levels in mice.
- Figure 7 illustrates (A) the effect of a partial antagonist polyclonal anti-PCSK9 mAb CRN6 on LDLR down regulation and (B) the lack of effect on cholesterol levels in mice.
- Figure 8 illustrates the time course of the cholesterol lowering effect obtained using anti-PCSK9 antagonist antibody 7D4 in mice.
- Figure 9 illustrates the dose dependence of the anti-PCSK9 antagonist mAb 7D4 on the reduction of serum total cholesterol, HDL and LDL in mice.
- Figure 10 illustrates the dose dependence of the cholesterol lowering effect of anti-PCSK9 antagonist antibody 5A10 in mice.
- Figure 11 illustrates the dose dependence of the cholesterol lowering effect of anti-PCSK9 antagonist antibodies (A) 4A5 and (B) 6F6 in mice.
- Figure 12 depicts Western blots of anti-PCSK9 antagonist antibodies effect on liver LDLR levels.
- Figure 13 illustrates the lack of effect of anti-PCSK9 antagonist antibody 4A5 in an LDLR-/- mouse model.
- Figure 14 illustrates the effect on total serum cholesterol of multiple administrations of anti-PCSK9 antagonist antibodies in mice over a longer time course than seen with a single dose.
- Figure 15 illustrate the time course of the effects of anti-PCSK9 antagonistic antibody 7D4 on lipid parameters in a cynomolgus monkey model.
- Figure 16 illustrates the dose- and time- response of anti-PCSK9 antagonistic antibody 7D4 on serum cholesterol levels in the cynomolgus monkey.
- Figure 17 illustrates a comparison of anti-PCSK9 antagonistic antibodies 4A5,
- Figure 18 illustrates the time course of the effect of anti-PCSK9 antagonist antibody 7D4 on plasma cholesterol levels of cynomolgus monkeys fed a 33.4% kcal fat diet supplemented with 0.1 % cholesterol.
- Figure 19 illustrates the effect of L1 L3 (humanized anti-PCSK9 monoclonal antibody) on down regulation of LDLR in Huh7 cells.
- Figure 20 illustrates the dose-response of L1 L3 humanized antibody, the mouse precursor 5A10, and negative control antibody 42H7 on blocking the binding of recombinant biotinylated human PCSK9 (A and B) and mouse PCSK9 (C and D) to immobilized recombinant LDLR extracellular domain in vitro at pH 7.5 (A and C) and pH 5.3 (B and D).
- Figure 21 illustrates the effect on serum cholesterol of treatment of mice with 10 mg/kg L1 L3.
- Figure 22 illustrates the effect of administration of 5A10 antibody or L1 L3 to cynomolgus monkeys and measurement of changes in serum HDL (A) and serum LDL (B) as a function of time.
- Figure 23A depicts the crystal structure of the PCSK9 (light gray surface representation) bound to the L1 L3 antibody (black cartoon representation).
- Figure 23B depicts the crystal structure of the PCSK9 (light gray surface representation) bound to the EGF-like domain of the LDLR (black cartoon representation) (Kwon et al., PNAS,
- Figure 23C shows the surface area representation of PCSK9 with the L1 L3 epitope shown in dark gray.
- Figure 23D shows the surface area representation of PCSK9 with the LDLR EGF-like domain epitope shown in dark gray.
- Figures 24 A-G depict the substitutions made in the CDRs of antibody 5A10 in the course of affinity maturation and optimization and to achieve particular properties.
- PCSK9 binding associated with antibodies having these CDR substitutions is also represented.
- the number following each sequence is the SEQ ID NO designated for each sequence.
- the present invention relates to antibodies, peptides, and aptamers that antagonize the function of extracellular PCSK9 including its interaction with the LDLR. More specifically, the invention relates to methods of making antagonist PCSK9 antibodies, peptides, and aptamers, compositions comprising these antibodies, peptides, and/or aptamers, and methods of using these antibodies, peptides, and/or aptamers as a medicament.
- the antagonist PCSK9 antibodies and peptides can be used to lower blood LDL-cholesterol levels, and can be used in the prevention and/or treatment of cholesterol and lipoprotein metabolism disorders, including familial hypercholesterolemia, atherogenic dyslipidemia, atherosclerosis, and, more generally, CVD.
- an "antibody” is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
- a target such as a carbohydrate, polynucleotide, lipid, polypeptide, etc.
- the term encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab') 2 , Fv), single chain (ScFv) and domain antibodies), and fusion proteins comprising an antibody portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site.
- An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
- immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGI , lgG2, lgG3, lgG4, IgAI and lgA2.
- the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
- the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
- monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
- the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
- the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, 1975, Nature 256:495, or may be made by recombinant DNA methods such as described in U.S. Pat. No. 4,816,567.
- the monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et al., 1990, Nature 348:552-554, for example.
- humanized antibody refers to forms of non-human (e.g., murine) antibodies that are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin.
- humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
- CDR complementary determining region
- Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
- the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
- the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
- the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
- human antibody means an antibody having an amino acid sequence corresponding to that of an antibody that can be produced by a human and/or which has been made using any of the techniques for making human antibodies known to those skilled in the art or disclosed herein. This definition of a human antibody includes antibodies comprising at least one human heavy chain polypeptide or at least one human light chain polypeptide.
- Human antibodies can be produced using various techniques known in the art.
- the human antibody is selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., 1996, Nature Biotechnology, 14:309-314; Sheets et al., 1998, Proc. Natl. Acad. Sci. (USA) 95:6157-6162; Hoogenboom and Winter, 1991 , J. MoI. Biol., 227:381 ; Marks et al., 1991 , J. MoI. Biol., 222:581 ).
- Human antibodies can also be made by immunization of animals into which human immunoglobulin loci have been transgenically introduced in place of the endogenous loci, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated.
- This approach is described in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661 ,016.
- the human antibody may be prepared by immortalizing human B lymphocytes that produce an antibody directed against a target antigen (such B lymphocytes may be recovered from an individual or may have been immunized in vitro/ See, e.g., Cole et al.
- variable region of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
- variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) that contain hypervahable regions.
- the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies.
- a CDR may refer to CDRs defined by either approach or by a combination of both approaches.
- PCSK9 refers to any form of PCSK9 and variants thereof that retain at least part of the activity of PCSK9. Unless indicated differently, such as by specific reference to human PCSK9, PCSK9 includes all mammalian species of native sequence PCSK9, e.g., human, canine, feline, equine, and bovine. One exemplary human PCSK9 is found as Uniprot Accession Number Q8NBP7 (SEQ ID NO:16).
- PCSK9 antagonist refers to an antibody, peptide, or aptamer that is able to inhibit PCSK9 biological activity and/or downstream pathway(s) mediated by PCSK9 signaling, including PCSK9-mediated down-regulation of the LDLR, and PCSK9-mediated decrease in LDL blood clearance.
- a PCSK9 antagonist antibody encompasses antibodies that block, antagonize, suppress or reduce (to any degree including significantly) PCSK9 biological activity, including downstream pathways mediated by PCSK9 signaling, such as LDLR interaction and/or elicitation of a cellular response to PCSK9.
- PCSK9 antagonist antibody encompasses all the previously identified terms, titles, and functional states and characteristics whereby the PCSK9 itself, a PCSK9 biological activity (including but not limited to its ability to mediate any aspect of interaction with the LDLR, down regulation of LDLR, and decreased blood LDL clearance), or the consequences of the biological activity, are substantially nullified, decreased, or neutralized in any meaningful degree.
- a PCSK9 antagonist antibody binds PCSK9 and prevents interaction with the LDLR. Examples of PCSK9 antagonist antibodies are provided herein.
- a "full antagonist” is an antagonist which, at an effective concentration, essentially completely blocks a measurable effect of PCSK9.
- a partial antagonist is meant an antagonist that is capable of partially blocking a measurable effect, but that, even at a highest concentration is not a full antagonist.
- essentially completely is meant at least about 80%, preferably, at least about 90%, more preferably, at least about 95%, and most preferably, at least about 98% or 99% of the measurable effect is blocked.
- measurable effects include down regulation of LDLR by a PCSK9 antagonist as assayed in Huh7 cells in vitro, in vivo decrease in blood (or plasma) levels of total cholesterol, and in vivo decrease in LDL levels in blood (or plasma).
- the term "clinically meaningful” means at least a 15% reduction in blood LDL-cholesterol levels in humans or at least a 15% reduction in total blood cholesterol in mice. It is clear that measurements in plasma or serum can serve as surrogates for measurement of levels in blood.
- PCSK9 antagonist peptide or “PCSK9 antagonist aptamer” includes any conventional peptide or polypeptide or aptamer that blocks, antagonizes, suppresses or reduces (to any degree including significantly) PCSK9 biological activity, including downstream pathways mediated by PCSK9 signaling, such as LDLR interaction and/or elicitation of a cellular response to PCSK9.
- PCSK9 antagonist peptides or polypeptides include Fc fusions comprising the LDLR and soluble portions of the LDLR, or mutants thereof with higher affinity to PCSK9.
- polypeptide oligopeptide
- peptide protein
- the terms “polypeptide”, “oligopeptide”, “peptide” and “protein” are used interchangeably herein to refer to chains of amino acids of any length, preferably, relatively short (e.g., 10-100 amino acids).
- the chain may be linear or branched, it may comprise modified amino acids, and/or may be interrupted by non-amino acids.
- the terms also encompass an amino acid chain that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
- polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
- polypeptides can occur as single chains or associated chains.
- polynucleotide or “nucleic acid,” as used interchangeably herein, refer to chains of nucleotides of any length, and include DNA and RNA.
- the nucleotides can be deoxyhbonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a chain by DNA or RNA polymerase.
- a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the chain.
- the sequence of nucleotides may be interrupted by non-nucleotide components.
- a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
- Other types of modifications include, for example, "caps", substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metal
- any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports.
- the 5' and 3' terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms.
- Other hydroxyls may also be dehvatized to standard protecting groups.
- Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2'-O-methyl-, 2'-O-allyl, 2'-fluoro- or 2'-azido-hbose, carbocyclic sugar analogs, alpha- or beta-anomeric sugars, epimehc sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and abasic nucleoside analogs such as methyl riboside.
- One or more phosphodiester linkages may be replaced by alternative linking groups.
- linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S("thioate"), P(S)S ("dithioate"), (O)NR 2 ("amidate"), P(O)R, P(O)OR', CO or CH 2 ("formacetal"), in which each R or R' is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (-O-) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
- PCSK9 antagonist aptamer which comprises a nucleic acid or protein sequence, is, for example, selected from a large pool of random sequences and specifically binds PCSK9.
- the nucleic acid of the aptamer is double-stranded DNA or single-strand RNA.
- Nucleic acid aptamers can include modified bases or functional groups, including but not limited to 2'-fluorine nucleotides and 2'-O-methyl nucleotides.
- Aptamers can include hydrophilic polymers, for example, polyethylene glycol. Aptamers may be made by methods known in the art and selected for PCSK9 antagonist activity by routine modification of the methods disclosed in the Examples.
- an antibody, peptide, or aptamer "interacts with" PCSK9 when the equilibrium dissociation constant is equal to or less than 20 nM, preferably less than about 6 nM, more preferably less than about 1 nM, most preferably less than about 0.2 nM, as measured by the methods disclosed herein in Example 2.
- An epitope that "preferentially binds” or “specifically binds” (used interchangeably herein) to an antibody or a polypeptide is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art.
- a molecule is said to exhibit "specific binding” or “preferential binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances.
- An antibody “specifically binds” or “preferentially binds” to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances.
- an antibody that specifically or preferentially binds to a PCSK9 epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other PCSK9 epitopes or non- PCSK9 epitopes.
- an antibody or moiety or epitope that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target.
- binding does not necessarily require (although it can include) exclusive binding.
- reference to binding means preferential binding.
- substantially pure refers to material which is at least 50% pure (i.e., free from contaminants), more preferably, at least 90% pure, more preferably, at least 95% pure, yet more preferably, at least 98% pure, and most preferably, at least 99% pure.
- a "host cell” includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts.
- Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation.
- a host cell includes cells transfected in vivo with a polynucleotide(s) of this invention.
- the term "Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain.
- the "Fc region” may be a native sequence Fc region or a variant Fc region.
- the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl- terminus thereof.
- the numbering of the residues in the Fc region is that of the EU index as in Kabat. Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991.
- the Fc region of an immunoglobulin generally comprises two constant domains, CH2 and CH3.
- Fc receptor and “FcR” describe a receptor that binds to the Fc region of an antibody.
- the preferred FcR is a native sequence human FcR.
- a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
- Fc ⁇ RII receptors include Fc ⁇ RIIA (an "activating receptor") and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
- FcRs are reviewed in Ravetch and Kinet, 1991 , Ann. Rev. Immunol., 9:457-92; Capel et al., 1994, Immunomethods, 4:25-34; and de Haas et al., 1995, J. Lab. Clin. Med., 126:330-41.
- FcR also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., 1976 J. Immunol., 117:587; and Kim et al., 1994, J. Immunol., 24:249).
- Compet means that a first antibody, or an antigen-binding portion thereof, binds to an epitope in a manner sufficiently similar to the binding of a second antibody, or an antigen-binding portion thereof, such that the result of binding of the first antibody with its cognate epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody.
- the alternative, where the binding of the second antibody to its epitope is also detectably decreased in the presence of the first antibody can, but need not be the case. That is, a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope.
- each antibody detectably inhibits the binding of the other antibody with its cognate epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to "cross-compete" with each other for binding of their respective epitope(s).
- Both competing and cross-competing antibodies are encompassed by the present invention. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate, based upon the teachings provided herein, that such competing and/or cross-competing antibodies are encompassed and can be useful for the methods disclosed herein.
- an antibody with an epitope that "overlaps" with another (second) epitope or with a surface on PCSK9 that interacts with the EGF-like domain of the LDLR is meant the sharing of space in terms of the PCSK9 residues that are interacted with.
- the percent overlap of the claimed antibody's PCSK9 epitope with the surface of PCSK9 which interacts with the EGF-like domain of the LDLR the surface area of PCSK9 buried when in complex with the LDLR is calculated on a per-residue basis. The buried area is also calculated for these residues in the PCSK9:antibody complex.
- a "functional Fc region” possesses at least one effector function of a native sequence Fc region.
- effector functions include C1 q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity; phagocytosis; down-regulation of cell surface receptors (e.g., B cell receptor), etc.
- Such effector functions generally require the Fc region to be combined with a binding domain (e.g., an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
- a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
- a “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region.
- the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g., from about one to about ten amino acid substitutions, and preferably, from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
- the variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably, at least about 90% sequence identity therewith, more preferably, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% sequence identity therewith.
- beneficial or desired clinical results include, but are not limited to, one or more of the following: enhancement of LDL clearance and reducing incidence or amelioration of aberrant cholesterol and/or lipoprotein levels resulting from metabolic and/or eating disorders, or including familial hypercholesterolemia, atherogenic dyslipidemia, atherosclerosis, and, more generally, cardiovascular disease (CVD).
- CVD cardiovascular disease
- Reducing incidence means any of reducing severity (which can include reducing need for and/or amount of (e.g., exposure to) other drugs and/or therapies generally used for this condition.
- a "method of reducing incidence” reflects administering the PCSK9 antagonist antibody, peptide, or aptamer based on a reasonable expectation that such administration may likely cause such a reduction in incidence in that particular individual.
- “Ameliorating” means a lessening or improvement of one or more symptoms as compared to not administering a PCSK9 antagonist antibody, peptide, or aptamer. “Ameliorating” also includes shortening or reduction in duration of a symptom.
- an "effective dosage” or “effective amount” of drug, compound, or pharmaceutical composition is an amount sufficient to effect any one or more beneficial or desired results.
- beneficial or desired results include eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
- beneficial or desired results include clinical results such as reducing hypercholesterolemia or one or more symptoms of dyslipidemia, atherosclerosis, CVD, or coronary heart disease, decreasing the dose of other medications required to treat the disease, enhancing the effect of another medication, and/or delaying the progression of the disease of patients.
- an effective dosage can be administered in one or more administrations.
- an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
- an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
- an "effective dosage" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
- an “individual” or a “subject” is a mammal, more preferably, a human. Mammals also include, but are not limited to, farm animals, sport animals, pets, primates, horses, dogs, cats, mice and rats.
- "vector” means a construct, which is capable of delivering, and, preferably, expressing, one or more gene(s) or sequence(s) of interest in a host cell.
- vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells.
- expression control sequence means a nucleic acid sequence that directs transcription of a nucleic acid.
- An expression control sequence can be a promoter, such as a constitutive or an inducible promoter, or an enhancer.
- the expression control sequence is operably linked to the nucleic acid sequence to be transcribed.
- pharmaceutically acceptable carrier or “pharmaceutical acceptable excipient” includes any material which, when combined with an active ingredient, allows the ingredient to retain biological activity and is non-reactive with the subject's immune system.
- examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents.
- Preferred diluents for aerosol or parenteral administration are phosphate buffered saline (PBS) or normal (0.9%) saline.
- Compositions comprising such carriers are formulated by well known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, PA, 1990; and Remington, The Science and Practice of Pharmacy, 20th Ed., Mack Publishing, 2000).
- k on refers to the rate constant for association of an antibody to an antigen. Specifically, the rate constants (k on and k Off ) and equilibrium dissociation constants are measured using Fab antibody fragments (i.e., univalent) and PCSK9.
- k O ff refers to the rate constant for dissociation of an antibody from the antibody/antigen complex.
- K 0 refers to the equilibrium dissociation constant of an antibody-antigen interaction.
- the invention provides a method for treating or preventing hypercholesterolemia, and/or at least one symptom of dyslipidemia, atherosclerosis,
- CVD or coronary heart disease in an individual comprising administering to the individual an effective amount of a PCSK9 antagonist antibody or peptide or aptamer that antagonizes circulating PCSK9.
- the invention provides an effective amount of a PCSK9 antagonist antibody, peptide, or aptamer that antagonizes circulating PCSK9 for use in treating or preventing hypercholesterolemia, and/or at least one symptom of dyslipidemia, atherosclerosis, CVD or coronary heart disease, in an individual.
- the invention further provides the use of an effective amount of a PCSK9 antagonist antibody, peptide, or aptamer that antagonizes extracellular or circulating PCSK9 in the manufacture of a medicament for treating or preventing hypercholesterolemia, and/or at least one symptom of dyslipidemia, atherosclerosis, CVD or coronary heart disease, in an individual.
- blood cholesterol and/or blood LDL is at least about 10% or 15% lower than before administration. More preferably, blood cholesterol and/or blood LDL is at least about 20% lower than before administration of the antibody. Yet more preferably, blood cholesterol and/or blood LDL is at least 30% lower than before administration of the antibody.
- blood cholesterol and/or blood LDL is at least 40% lower than before administration of the antibody. More advantageously, blood cholesterol and/or blood LDL is at least 50% lower than before administration of the antibody.
- blood cholesterol and/or blood LDL is at least 60% lower than before administration of the antibody. Most preferably, blood cholesterol and/or blood LDL is at least 70% lower than before administration of the antibody.
- PCSK9 antagonist antibodies, peptides, and aptamers also include compositions comprising one or more additional agents. These compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
- suitable excipients such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
- the present invention can be used alone or in combination with other conventional methods of treatment.
- the PCSK9 antagonist antibody, peptide, or aptamer can be administered to an individual via any suitable route. It should be apparent to a person skilled in the art that the examples described herein are not intended to be limiting but to be illustrative of the techniques available.
- the PCSK9 antagonist antibody, peptide, or aptamer is administered to an individual in accord with known methods, such as intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, transdermal, subcutaneous, intra-articular, sublingually, intrasynovial, via insufflation, intrathecal, oral, inhalation or topical routes.
- Administration can be systemic, e.g., intravenous administration, or localized.
- nebulizers for liquid formulations including jet nebulizers and ultrasonic nebulizers are useful for administration.
- Liquid formulations can be directly nebulized and lyophilized powder can be nebulized after reconstitution.
- PCSK9 antagonist antibody, peptide, or aptamer can be aerosolized using a fluorocarbon formulation and a metered dose inhaler, or inhaled as a lyophilized and milled powder.
- a PCSK9 antagonist antibody, peptide, or aptamer is administered via site-specific or targeted local delivery techniques.
- site-specific or targeted local delivery techniques include various implantable depot sources of the PCSK9 antagonist antibody, peptide, or aptamer or local delivery catheters, such as infusion catheters, indwelling catheters, or needle catheters, synthetic grafts, adventitial wraps, shunts and stents or other implantable devices, site specific carriers, direct injection, or direct application. See, e.g., PCT Publ. No. WO 00/53211 and U.S. Patent No. 5,981 ,568.
- PCSK9 antagonist antibody, peptide, or aptamer may be used for administration.
- the PCSK9 antagonist antibody, peptide, or aptamer may be administered neat.
- PCSK9 antagonist antibody, peptide, or aptamer and a pharmaceutically acceptable excipient may be in various formulations.
- Pharmaceutically acceptable excipients are known in the art, and are relatively inert substances that facilitate administration of a pharmacologically effective substance. For example, an excipient can give form or consistency, or act as a diluent.
- Suitable excipients include but are not limited to stabilizing agents, wetting and emulsifying agents, salts for varying osmolahty, encapsulating agents, buffers, and skin penetration enhancers. Excipients as well as formulations for parenteral and nonparenteral drug delivery are set forth in Remington, The Science and Practice of Pharmacy, 20th Ed., Mack Publishing (2000).
- agents can be combined with pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like.
- pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like.
- the particular dosage regimen i.e., dose, timing and repetition, will depend on the particular individual and that individual's medical history.
- PCSK9 antibodies can also be administered via inhalation, as described herein.
- an initial candidate dosage can be about 2 mg/kg.
- a typical daily dosage might range from about any of about 3 ⁇ g/kg to 30 ⁇ g/kg to 300 ⁇ g/kg to 3 mg/kg, to 30 mg/kg, to 100 mg/kg or more, depending on the factors mentioned above.
- dosage of about 1 mg/kg, about 2.5 mg/kg, about 5 mg/kg, about 10 mg/kg, and about 25 mg/kg may be used.
- the treatment is sustained until a desired suppression of symptoms occurs or until sufficient therapeutic levels are achieved, for example, to reduce blood LDL levels.
- An exemplary dosing regimen comprises administering an initial dose of about 2 mg/kg, followed by a weekly maintenance dose of about 1 mg/kg of the PCSK9 antibody, or followed by a maintenance dose of about 1 mg/kg every other week.
- other dosage regimens may be useful, depending on the pattern of pharmacokinetic decay that the practitioner wishes to achieve. For example, in some embodiments, dosing from one to four times a week is contemplated. In other embodiments dosing once a month or once every other month or every three months is contemplated. The progress of this therapy is easily monitored by conventional techniques and assays.
- the dosing regimen (including the PCSK9 antagonist(s) used) can vary over time.
- the appropriate dosage of a PCSK9 antagonist antibody, peptide, or aptamer will depend on the PCSK9 antagonist antibody, peptide, or aptamer (or compositions thereof) employed, the type and severity of symptoms to be treated, whether the agent is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the agent, the patient's blood PCSK9 levels, the patient's synthesis and clearance rate for PCSK9, the patient's clearance rate for the administered agent, and the discretion of the attending physician.
- the clinician will administer a PCSK9 antagonist antibody, peptide, or aptamer until a dosage is reached that achieves the desired result. Dose and/or frequency can vary over course of treatment.
- Empirical considerations such as the half-life, generally will contribute to the determination of the dosage.
- antibodies that are compatible with the human immune system such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's immune system.
- Frequency of administration may be determined and adjusted over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of symptoms, e.g., hypercholesterolemia.
- sustained continuous release formulations of PCSK9 antagonist antibodies may be appropriate.
- formulations and devices for achieving sustained release are known in the art.
- dosages for an antagonist antibody, peptide, or aptamer may be determined empirically in individuals who have been given one or more administration(s) of an antagonist antibody, peptide, or aptamer. Individuals are given incremental dosages of a PCSK9 antagonist antibody, peptide, or aptamer. To assess efficacy, an indicator of the disease can be followed.
- Administration of a PCSK9 antagonist antibody, peptide, or aptamer in accordance with the method in the present invention can be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
- the administration of a PCSK9 antagonist antibody, peptide, or aptamer may be essentially continuous over a preselected period of time or may be in a series of spaced doses.
- more than one antagonist antibody, peptide, or aptamer may be present. At least one, at least two, at least three, at least four, at least five different, or more antagonist antibodies and/or peptides can be present.
- PCSK9 antagonist antibodies or peptides may have complementary activities that do not adversely affect each other.
- a PCSK9 antagonist antibody, peptide, or aptamer can also be used in conjunction with other PCSK9 antagonists or PCSK9 receptor antagonists.
- one or more of the following PCSK9 antagonists may be used: an antisense molecule directed to a PCSK9 (including an anti-sense molecule directed to a nucleic acid encoding PCSK9), a PCSK9 inhibitory compound, and a PCSK9 structural analog.
- a PCSK9 antagonist antibody, peptide, or aptamer can also be used in conjunction with other agents that serve to enhance and/or complement the effectiveness of the agents.
- Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and may comprise buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine,
- Liposomes containing the PCSK9 antagonist antibody, peptide, or aptamer are prepared by methods known in the art, such as described in Epstein, et al., 1985, Proc. Natl. Acad. Sci. USA 82:3688; Hwang, et al., 1980, Proc. Natl Acad. Sci. USA 77:4030; and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556.
- Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG- PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
- PEG- PE PEG-derivatized phosphatidylethanolamine
- the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroennulsions.
- colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
- Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or 'poly(vinylalcohol)), polylactides (U.S. Pat. No.
- copolymers of L-glutamic acid and 7 ethyl-L-glutamate copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT TM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3-hydroxybutyhc acid.
- LUPRON DEPOT TM injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate
- sucrose acetate isobutyrate sucrose acetate isobutyrate
- poly-D-(-)-3-hydroxybutyhc acid poly-D-(-)-3-hydroxybutyhc acid.
- compositions to be used for in vivo administration must be sterile. This is readily accomplished by, for example, filtration through sterile filtration membranes.
- Therapeutic PCSK9 antagonist antibody, peptide, or aptamer compositions are generally placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
- Suitable emulsions may be prepared using commercially available fat emulsions, such as IntralipidTM, LiposynTM, InfonutrolTM, LipofundinTM and LipiphysanTM.
- the active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g., egg phospholipids, soybean phospholipids or soybean lecithin) and water.
- an oil e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil
- a phospholipid e.g., egg phospholipids, soybean phospholipids or soybean lecithin
- other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emul
- Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%.
- the fat emulsion can comprise fat droplets between 0.1 and 1.0 ⁇ m, particularly 0.1 and 0.5 ⁇ m, and have a pH in the range of 5.5 to 8.0.
- the emulsion compositions can be those prepared by mixing a PCSK9 antagonist antibody, peptide, or aptamer with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water).
- Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
- the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above.
- compositions are administered by the oral or nasal respiratory route for local or systemic effect.
- Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulised by use of gases. Nebulised solutions may be breathed directly from the nebulising device or the nebulising device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
- PCSK9 antagonist antibody refers to any peptide or nucleic acid molecule that blocks, suppresses or reduces (including significantly reduces) PCSK9 biological activity, including downstream pathways mediated by PCSK9 signaling, such as elicitation of a cellular response to PCSK9.
- a PCSK9 antagonist antibody, peptide, or aptamer should exhibit any one or more of the following characteristics: (a) bind to PCSK9; (b) block PCSK9 interaction with the LDLR; (c) block or decrease PCSK9-mediated down-regulation of the LDLR; (d) inhibit the PCSK9-mediated decrease in LDL blood clearance, (e) increase LDL clearance in media by cultured hepatocytes, (f) increase blood LDL clearance by the liver in vivo, (g) sensitize to statins, and (h) block PCSK9 interaction with other yet to be identified factors.
- the antibody, peptide, or aptamer preferably reacts with PCSK9 in a manner that inhibits PCSK9 signaling function and LDLR interaction.
- the PCSK9 antagonist antibody specifically recognizes primate PCSK9.
- the PCSK9 antagonist antibody binds primate and rodent PCSK9.
- the antibodies useful in the present invention can encompass monoclonal antibodies, polyclonal antibodies, antibody fragments (e.g., Fab, Fab', F(ab')2, Fv, Fc, etc.), chimeric antibodies, bispecific antibodies, heteroconjugate antibodies, single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion (e.g., a domain antibody), human antibodies, humanized antibodies, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies.
- the antibodies may be murine, rat, human, or any other origin (including chimeric or humanized antibodies).
- the PCSK9 antagonist antibody is a monoclonal antibody.
- the PCSK9 antagonist antibody can also be humanized. In other embodiments, the antibody is human.
- the antibody comprises a modified constant region, such as a constant region that is immunologically inert, that is, having a reduced potential for provoking an immune response.
- the constant region is modified as described in Eur. J. Immunol., 1999, 29:2613-2624; PCT Publ. No. WO99/58572; and/or UK Patent Application No. 9809951.8.
- the Fc can be human lgG2 or human IgG 4 .
- the Fc can be human IgG 2 containing the mutation A330P331 to S330S331 (IgG 2 Aa), in which the amino acid residues are numbered with reference to the wild type lgG2 sequence.
- the antibody comprises a constant region of IgG 4 comprising the following mutations (Armour et al., 2003, Molecular Immunology 40 585-593): E233F234L235 to P233V234A235 (IgG 4 Ac), in which the numbering is with reference to wild type lgG4.
- the Fc is human IgG 4 E233F234L235 to P233V234A235 with deletion G236 (IgG 4 Ab)-
- the Fc is any human IgG 4 Fc (IgG 4 , IgG 4Ab or lgG 4 ⁇ c ) containing hinge stabilizing mutation S228 to P228 (Aalberse et al., 2002, Immunology 105, 9-19).
- the Fc can be aglycosylated Fc.
- the constant region is aglycosylated by mutating the oligosaccharide attachment residue (such as Asn297) and/or flanking residues that are part of the glycosylation recognition sequence in the constant region.
- the constant region is aglycosylated for N-linked glycosylation enzymatically.
- the constant region may be aglycosylated for N-linked glycosylation enzymatically or by expression in a glycosylation deficient host cell.
- the binding affinity (K 0 ) of a PCSK9 antagonist antibody to PCSK9 can be about 0.002 to about 200 nM.
- the binding affinity is any of about 200 nM, about 100 nM, about 50 nM, about 10 nM, about 1 nM, about 500 pM, about 100 pM, about 60 pM, about 50 pM, about 20 pM, about 15 pM, about 10 pM, about 5 pM, or about 2 pM.
- the binding affinity is less than any of about 250 nM, about 200 nM, about 100 nM, about 50 nM, about 10 nM, about 1 nM, about 500 pM, about 100 pM, about 50 pM, about 20 pM, about 10 pM, about 5 pM, or about 2 pM.
- One way of determining binding affinity of antibodies to PCSK9 is by measuring binding affinity of monofunctional Fab fragments of the antibody.
- an antibody for example, IgG
- an antibody can be cleaved with papain or expressed recombinantly.
- the affinity of a PCSK9 Fab fragment of an antibody can be determined by surface plasmon resonance (Biacore3000TM surface plasmon resonance (SPR) system, Biacore, INC, Piscataway NJ) equipped with pre- immobilized streptavidin sensor chips (SA) using HBS-EP running buffer (0.01 M HEPES, pH 7.4, 0.15 NaCI, 3 mM EDTA, 0.005% v/v Surfactant P20).
- Biotinylated human PCSK9 (or any other PCSK9) can be diluted into HBS-EP buffer to a concentration of less than 0.5 ⁇ g/mL and injected across the individual chip channels using variable contact times, to achieve two ranges of antigen density, either 50-200 response units (RU) for detailed kinetic studies or 800-1 ,000 RU for screening assays. Regeneration studies have shown that 25 mM NaOH in 25% v/v ethanol effectively removes the bound Fab while keeping the activity of PCSK9 on the chip for over 200 injections.
- serial dilutions (spanning concentrations of 0.1 -10x estimated K 0 ) of purified Fab samples are injected for 1 min at 100 ⁇ L/minute and dissociation times of up to 2 hours are allowed.
- concentrations of the Fab proteins are determined by ELISA and/or SDS-PAGE electrophoresis using a Fab of known concentration (as determined by amino acid analysis) as a standard.
- Kinetic association rates (k on ) and dissociation rates (k Off ) are obtained simultaneously by fitting the data globally to a 1 :1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B., 1994. Methods Enzymology 6.
- the route and schedule of immunization of the host animal are generally in keeping with established and conventional techniques for antibody stimulation and production, as further described herein.
- General techniques for production of human and mouse antibodies are known in the art and/or are described herein.
- a currently preferred method of making the antibodies comprises the immunization of PCSK9 " knockout (PCSK9 -/-) animals as disclosed herein.
- any mammalian subject including humans or antibody producing cells therefrom can be manipulated to serve as the basis for production of mammalian, including human, hybhdoma cell lines.
- the host animal is inoculated intraperitoneally, intramuscularly, orally, subcutaneously, intraplantar, and/or intradermally with an amount of immunogen, including as described herein.
- Hybhdomas can be prepared from the lymphocytes and immortalized myeloma cells using the general somatic cell hybridization technique of Kohler, B. and Milstein, C, 1975, Nature 256:495-497 or as modified by Buck, D. W., et al., 1982, In Vitro, 18:377-381.
- Available myeloma lines including but not limited to X63-Ag8.653 and those from the SaIk Institute, Cell Distribution Center, San Diego, Calif., USA, may be used in the hybridization.
- the technique involves fusing myeloma cells and lymphoid cells using a fusogen such as polyethylene glycol, or by electrical means well known to those skilled in the art.
- the cells are separated from the fusion medium and grown in a selective growth medium, such as hypoxanthine-aminopterin- thymidine (HAT) medium, to eliminate unhybhdized parent cells.
- a selective growth medium such as hypoxanthine-aminopterin- thymidine (HAT) medium
- HAT hypoxanthine-aminopterin- thymidine
- Any of the media described herein, supplemented with or without serum, can be used for cultuhng hybridomas that secrete monoclonal antibodies.
- EBV immortalized B cells may be used to produce the PCSK9 monoclonal antibodies of the subject invention.
- hybridomas are expanded and subcloned, if desired, and supernatants are assayed for anti-immunogen activity by conventional immunoassay procedures (e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay).
- immunoassay procedures e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay.
- Hybridomas that may be used as a source of antibodies encompass all derivatives, progeny cells of the parent hybridomas that produce monoclonal antibodies specific for PCSK9, or a portion thereof. Hybridomas that produce such antibodies may be grown in vitro or in vivo using known procedures.
- the monoclonal antibodies may be isolated from the culture media or body fluids, by conventional immunoglobulin purification procedures such as ammonium sulfate precipitation, gel electrophoresis, dialysis, chromatography, and ultrafiltration, if desired.
- Undesired activity, if present, can be removed, for example, by running the preparation over adsorbents made of the immunogen attached to a solid phase and eluting or releasing the desired antibodies off the immunogen.
- a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or
- the PCSK9 antagonist antibody (monoclonal or polyclonal) of interest may be sequenced and the polynucleotide sequence may then be cloned into a vector for expression or propagation.
- the sequence encoding the antibody of interest may be maintained in vector in a host cell and the host cell can then be expanded and frozen for future use.
- Production of recombinant monoclonal antibodies in cell culture can be carried out through cloning of antibody genes from B cells by means known in the art. See, e.g., Tiller et al., 2008, J. Immunol. Methods 329, 112; US Patent No. 7,314,622.
- the polynucleotide sequence may be used for genetic manipulation to "humanize” the antibody or to improve the affinity, or other characteristics of the antibody.
- the constant region may be engineered to more nearly resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. It may be desirable to genetically manipulate the antibody sequence to obtain greater affinity to PCSK9 and greater efficacy in inhibiting PCSK9. It will be apparent to one of skill in the art that one or more polynucleotide changes can be made to the PCSK9 antagonist antibody and still maintain its binding ability to PCSK9.
- a number of "humanized" antibody molecules comprising an antigen-binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent or modified rodent V regions and their associated CDRs fused to human constant domains. See, for example, Winter et al., 1991 , Nature 349:293-299; Lobuglio et al., 1989, Proc. Nat. Acad. Sci. USA 86:4220-4224; Shaw et al., 1987, J Immunol. 138:4534-4538; and Brown et al., 1987, Cancer Res. 47:3577-3583.
- rodent CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody constant domain See, for example, Riechmann et al.,1988, Nature 332:323-327; Verhoeyen et al., 1988, Science 239:1534-1536; and Jones et al., 1986, Nature 321 :522-525.
- Another reference describes rodent CDRs supported by recombinantly engineered rodent framework regions. See, for example, European Patent Publ. No. 0519596. These "humanized" molecules are designed to minimize unwanted immunological response toward rodent anti-human antibody molecules which limits the duration and effectiveness of therapeutic applications of those moieties in human recipients.
- the antibody constant region can be engineered such that it is immunologically inert (e.g., does not trigger complement lysis). See, e.g., PCT Publ. No. WO99/58572; UK Patent Application No. 9809951.8.
- Other methods of humanizing antibodies that may also be utilized are disclosed by Daugherty et al., 1991 , Nucl. Acids Res. 19:2471-2476 and in U.S. Patent Nos. 6,180,377; 6,054,297; 5,997,867; 5,866,692; 6,210,671 ; and 6,350,861 ; and in PCT Publ. No. WO 01/27160.
- Fully human antibodies may be obtained by using commercially available mice that have been engineered to express specific human immunoglobulin proteins.
- Transgenic animals that are designed to produce a more desirable or more robust immune response may also be used for generation of humanized or human antibodies. Examples of such technology are XenomouseTM from Abgenix, Inc. (Fremont, CA), HuMAb-Mouse® and TC MouseTM from Medarex, Inc. (Princeton, NJ), and the Veloclmmune® mouse from Regeneron Pharmaceuticals, Inc. (Tarrytown, NY).
- antibodies may be made recombinantly and expressed using any method known in the art.
- antibodies may be made recombinantly by phage display technology. See, for example, U.S. Patent Nos. 5,565,332; 5,580,717; 5,733,743; and 6,265,150; and Winter et al., 1994, Annu. Rev. Immunol. 12:433-455.
- the phage display technology (McCafferty et al., 1990, Nature 348:552-553) can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
- antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle.
- a filamentous bacteriophage such as M13 or fd
- the filamentous particle contains a single-stranded DNA copy of the phage genome
- selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties.
- the phage mimics some of the properties of the B cell.
- Phage display can be performed in a variety of formats; see, e.g., Johnson, Kevin S. and Chiswell, David J., 1993, Current Opinion in Structural Biology 3:564-571.
- V-gene segments can be used for phage display.
- Clackson et al., 1991 , Nature 352:624-628 isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice.
- a repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Mark et al., 1991 , J. MoI. Biol. 222:581 -597, or Griffith et al., 1993, EMBO J. 12:725-734.
- Antibodies may be made recombinantly by first isolating the antibodies and antibody producing cells from host animals, obtaining the gene sequence, and using the gene sequence to express the antibody recombinantly in host cells (e.g., CHO cells). Another method which may be employed is to express the antibody sequence in plants (e.g., tobacco) or transgenic milk. Methods for expressing antibodies recombinantly in plants or milk have been disclosed. See, for example, Peeters, 2001 , et al. Vaccine 19:2756; Lonberg, N. and D. Huszar, 1995, Int. Rev. Immunol 13:65; and Pollock, et al., 1999, J Immunol Methods 231 :147.
- Immunoassays and flow cytometry sorting techniques such as fluorescence activated cell sorting (FACS) can also be employed to isolate antibodies that are specific for PCSK9.
- the antibodies can be bound to many different carriers.
- Carriers can be active and/or inert. Examples of well-known carriers include polypropylene, polystyrene, polyethylene, dextran, nylon, amylases, glass, natural and modified celluloses, polyacrylamides, agaroses and magnetite.
- the nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding antibodies, or will be able to ascertain such, using routine experimentation.
- the carrier comprises a moiety that targets the myocardium.
- DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies).
- the hybhdoma cells serve as a preferred source of such DNA.
- the DNA may be placed into expression vectors (such as expression vectors disclosed in PCT Publ. No. WO 87/04462), which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
- expression vectors such as expression vectors disclosed in PCT Publ. No. WO 87/04462
- host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or mye
- the DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences, Morrison et al., 1984, Proc. Nat. Acad. Sci. 81 :6851 , or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
- “chimeric” or “hybrid” antibodies are prepared that have the binding specificity of a PCSK9 monoclonal antibody herein.
- PCSK9 antagonist antibodies and polypeptides derived from antibodies can be identified or characterized using methods known in the art, whereby reduction, amelioration, or neutralization of PCSK9 biological activity is detected and/or measured.
- a PCSK9 antagonist antibody or polypeptide is identified by incubating a candidate agent with PCSK9 and monitoring binding and/or attendant reduction or neutralization of a biological activity of PCSK9.
- the binding assay may be performed with purified PCSK9 polypeptide(s), or with cells naturally expressing, or transfected to express, PCSK9 polypeptide(s).
- the binding assay is a competitive binding assay, where the ability of a candidate antibody to compete with a known PCSK9 antagonist for PCSK9 binding is evaluated.
- the assay may be performed in various formats, including the ELISA format.
- a PCSK9 antagonist antibody is identified by incubating a candidate agent with PCSK9 and monitoring binding and attendant inhibition of LDLR expression and/or blood cholesterol clearance.
- PCSK9 antagonist antibody Following initial identification, the activity of a candidate PCSK9 antagonist antibody can be further confirmed and refined by bioassays that are known to test the targeted biological activities. Alternatively, bioassays can be used to screen candidates directly. Some of the methods for identifying and characterizing PCSK9 antagonist antibodies, peptides, or aptamers are described in detail in the Examples.
- PCSK9 antagonist antibodies may be characterized using methods well known in the art. For example, one method is to identify the epitope to which it binds, or "epitope mapping.” There are many methods known in the art for mapping and characterizing the location of epitopes on proteins, including solving the crystal structure of an antibody-antigen complex, competition assays, gene fragment expression assays, and synthetic peptide-based assays, as described, for example, in Chapter 11 of Harlow and Lane, Using Antibodies, a Laboratory Manual, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999). In an additional example, epitope mapping can be used to determine the sequence to which a PCSK9 antagonist antibody binds.
- Epitope mapping is commercially available from various sources, for example, Pepscan Systems (Edelhertweg 15, 8219 PH Lelystad, The Netherlands).
- the epitope can be a linear epitope, i.e., contained in a single stretch of amino acids, or a conformational epitope formed by a three-dimensional interaction of amino acids that may not necessarily be contained in a single stretch.
- Peptides of varying lengths e.g., at least 4-6 amino acids long
- the epitope to which the PCSK9 antagonist antibody binds can be determined in a systematic screening by using overlapping peptides derived from the PCSK9 sequence and determining binding by the PCSK9 antagonist antibody.
- the open reading frame encoding PCSK9 is fragmented either randomly or by specific genetic constructions and the reactivity of the expressed fragments of PCSK9 with the antibody to be tested is determined.
- the gene fragments may, for example, be produced by PCR and then transcribed and translated into protein in vitro, in the presence of radioactive amino acids. The binding of the antibody to the radioactively labeled PCSK9 fragments is then determined by immunoprecipitation and gel electrophoresis.
- Certain epitopes can also be identified by using large libraries of random peptide sequences displayed on the surface of phage particles (phage libraries). Alternatively, a defined library of overlapping peptide fragments can be tested for binding to the test antibody in simple binding assays. In an additional example, mutagenesis of an antigen binding domain, domain swapping experiments and alanine scanning mutagenesis can be performed to identify residues required, sufficient, and/or necessary for epitope binding.
- domain swapping experiments can be performed using a mutant PCSK9 in which various fragments of the PCSK9 polypeptide have been replaced (swapped) with sequences from PCSK9 from another species, or a closely related, but antigenically distinct protein (such as another member of the proprotein convertase family).
- sequences from PCSK9 from another species or a closely related, but antigenically distinct protein (such as another member of the proprotein convertase family).
- PCSK9 antagonist antibody Yet another method which can be used to characterize a PCSK9 antagonist antibody is to use competition assays with other antibodies known to bind to the same antigen, i.e., various fragments on PCSK9, to determine if the PCSK9 antagonist antibody binds to the same epitope as other antibodies. Competition assays are well known to those of skill in the art.
- the crystal structure of the antibody and antibody:antigen complex can also be used to characterize the antibody.
- the residues are identified by calculating the difference in accessible surface area between the L1 L3:PCSK9 crystal structure and PCSK9 structure alone.
- PCSK9 residues that show buried surface area upon complex formation with L1 L3 antibody are included as a part of the epitope.
- the solvent accessible surface of a protein is defined as the locus of the centre of a probe sphere (representing a solvent molecule of 1.4 A radius) as it rolls over the Van der Waals surface of the protein.
- the solvent accessible surface area is calculated by generating surface points on an extended sphere about each atom (at a distance from the atom centre equal to the sum of the atom and probe radii), and eliminating those that lie within equivalent spheres associated with neighboring atoms as implemented in program AREAIMOL (Briggs, P.J., 2000, CCP4 Newsletter No. 38, CCLRC, Daresbury).
- An expression vector can be used to direct expression of a PCSK9 antagonist antibody.
- One skilled in the art is familiar with administration of expression vectors to obtain expression of an exogenous protein in vivo. See, e.g., U.S. Patent Nos. 6,436,908; 6,413,942; and 6,376,471.
- Administration of expression vectors includes local or systemic administration, including injection, oral administration, particle gun or cathetehzed administration, and topical administration.
- the expression vector is administered directly to the sympathetic trunk or ganglion, or into a coronary artery, atrium, ventrical, or pericardium.
- Targeted delivery of therapeutic compositions containing an expression vector, or subgenomic polynucleotides can also be used.
- Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al., 1993, Trends Biotechnol. 11 :202; Chiou et al., 1994, Gene Therapeutics: Methods And Applications Of Direct Gene Transfer (J.A. Wolff, ed.); Wu et al., 1988, J. Biol. Chem. 263:621 ; Wu et al., 1994, J. Biol. Chem. 269:542; Zenke et al., 1990, Proc. Natl. Acad. Sci.
- compositions containing a polynucleotide are administered in a range of about 100 ng to about 200 mg of DNA for local administration in a gene therapy protocol. Concentration ranges of about 500 ng to about 50 mg, about 1 ⁇ g to about 2 mg, about 5 ⁇ g to about 500 ⁇ g, and about 20 ⁇ g to about 100 ⁇ g of DNA can also be used during a gene therapy protocol.
- the therapeutic polynucleotides and polypeptides can be delivered using gene delivery vehicles.
- the gene delivery vehicle can be of viral or non-viral origin (see generally, Jolly, 1994, Cancer Gene Therapy 1 :51 ; Kimura, 1994, Human Gene Therapy 5:845; Connelly, 1995, Human Gene Therapy 1 :185; and Kaplitt, 1994, Nature Genetics 6:148). Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence can be either constitutive or regulated.
- Viral-based vectors for delivery of a desired polynucleotide and expression in a desired cell are well known in the art.
- Exemplary viral-based vehicles include, but are not limited to, recombinant retroviruses (see, e.g., PCT Publ. Nos. WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; WO 93/11230; WO 93/10218; WO 91/02805; U.S. Patent Nos. 5, 219,740 and 4,777,127; GB Patent No. 2,200,651 ; and EP Patent No.
- alphavirus-based vectors e.g., Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532)
- AAV adeno-associated virus
- Non-viral delivery vehicles and methods can also be employed, including, but not limited to, polycationic condensed DNA linked or unlinked to killed adenovirus alone (see, e.g., Curiel, 1992, Hum. Gene Ther. 3:147); ligand-linked DNA (see, e.g., Wu, J., 1989, Biol. Chem. 264:16985); eukaryotic cell delivery vehicles cells (see, e.g., U.S. Patent No. 5,814,482; PCT Publ. Nos. WO 95/07994; WO 96/17072; WO 95/30763; and WO 97/42338) and nucleic charge neutralization or fusion with cell membranes. Naked DNA can also be employed.
- Exemplary naked DNA introduction methods are described in PCT Publ. No. WO 90/11092 and U.S. Patent No. 5,580,859.
- Liposomes that can act as gene delivery vehicles are described in U.S. Patent No. 5,422,120; PCT Publ. Nos. WO 95/13796; WO 94/23697; WO 91/14445; and EP 0524968. Additional approaches are described in Philip, 1994, MoI. Cell Biol., 14:2411 , and in Woffendin, 1994 Proc. Natl. Acad. Sci. 91 :1581.
- compositions comprising antibodies described herein or made by the methods and having the characteristics described herein.
- compositions comprise one or more antibodies, peptides, or aptamers that antagonize the interaction of PCSK9 with the LDLR, and/or one or more polynucleotides comprising sequences encoding one or more these antibodies or peptides.
- compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
- PCSK9 antagonist antibodies and peptides of the invention are characterized by any (one or more) of the following characteristics: (a) bind to PCSK9; (b) block PCSK9 interaction with the LDLR; (c) decrease PCSK9-mediated down-regulation of the LDLR; and (d) inhibit PCSK9-mediated inhibition of LDL blood clearance.
- PCSK9 antibodies have two or more of these features. More preferably, the antibodies have three or more of the features. Most preferably, the antibodies have all four characteristics.
- the invention provides any of the following, or compositions (including pharmaceutical compositions) comprising any antibody having a partial light chain sequence and a partial heavy chain sequence as found in Table 1.
- the underlined sequences are CDR sequences according to Kabat and in bold according to Chothia.
- the invention also provides CDR portions of antibodies to PCSK9 (including Chothia and Kabat CDRs). Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed “combined CDRs” or “extended CDRs”). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof.
- the invention also provides methods of making any of these antibodies or polypeptides.
- the antibodies of this invention can be made by procedures known in the art.
- the polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis.
- Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available.
- an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Patent Nos. 5,807,715; 4,816,567; and 6,331 ,415.
- a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody 4A5, 5A10, 6F6, 7D4 or L1 L3.
- the sequence encoding the antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use.
- Vectors (including expression vectors) and host cells are further described herein.
- the invention also encompasses scFv of antibodies of this invention.
- Single chain variable region fragments are made by linking light and/or heavy chain variable regions by using a short linking peptide.
- An example of a linking peptide is (GGGGS) 3 (SEQ ID NO:24), which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region.
- Linkers of other sequences have been designed and used. Bird et al., 1988, supra.
- Linkers should be short, flexible polypeptides and preferably comprised of less than about 20 amino acid residues. Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports.
- the single chain variants can be produced either recombinantly or synthetically.
- an automated synthesizer can be used for synthetic production of scFv.
- a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli.
- a suitable host cell either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli.
- Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides.
- the resultant scFv can be isolated using standard protein purification techniques known in the art.
- Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al., 1993, Proc. Natl. Acad Sci. USA 90:6444-6448; Poljak, R. J., et al., 1994, Structure 2:1121 -1123).
- bispecific antibodies monoclonal antibodies that have binding specificities for at least two different antigens
- methods for making bispecific antibodies are known in the art (see, e.g., Suresh et al., 1986, Methods in Enzymology 121 :210).
- the recombinant production of bispecific antibodies was based on the coexpression of two immunoglobulin heavy chain-light chain pairs, with the two heavy chains having different specificities (Millstein and Cuello, 1983, Nature 305, 537-539).
- antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
- the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2 and CH3 regions. It is preferred to have the first heavy chain constant region (CH1 ), containing the site necessary for light chain binding, present in at least one of the fusions.
- DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are cotransfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance.
- the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm.
- This asymmetric structure with an immunoglobulin light chain in only one half of the bispecific molecule, facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations.
- This approach is described in PCT Publ. No. WO 94/04690.
- Heteroconjugate antibodies comprising two covalently joined antibodies, are also within the scope of the invention. Such antibodies have been used to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (PCT Publ. Nos. WO 91/00360 and WO 92/200373; EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents and techniques are well known in the art, and are described in U.S. Patent No. 4,676,980.
- Chimeric or hybrid antibodies also may be prepared in vitro using known methods of synthetic protein chemistry, including those involving cross-linking agents.
- immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond.
- suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyhmidate.
- Humanized antibody comprising one or more CDRs of antibodies 5A10 or 7D4 or one or more CDRs derived from antibodies 5A10 or 7D4 can be made, for example, using any methods know in the art. For example, four general steps may be used to humanize a monoclonal antibody. These are: (1 ) determining the nucleotide and predicted amino acid sequence of the starting antibody light and heavy variable domains; (2) designing the humanized antibody, i.e., deciding which antibody framework region to use during the humanizing process; (3) using the actual humanizing methodologies/techniques; and (4) transfecting and expressing the humanized antibody. See, for example, U.S. Patent Nos. 4,816,567; 5,807,715; 5,866,692; 6,331 ,415; 5,530,101 ; 5,693,761 ; 5,693,762; 5,585,089; and 6,180,370.
- the Fc portion can be modified to avoid interaction with FCY receptor and the complement and immune systems.
- the techniques for preparation of such antibodies are described in WO 99/58572.
- the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. See, for example, U.S. Patent Nos. 5,997,867 and 5,866,692.
- Humanized antibody comprising the light or heavy chain variable regions or one or more CDRs of an antibody or its variants shown in Table 1 , or one or more CDRs derived from the antibody or its variants shown in Table 2 can be made using any methods known in the art.
- Humanized antibodies may be made by any method known in the art.
- the invention encompasses modifications to the antibodies and polypeptides of the invention variants shown in Table 1 , including functionally equivalent antibodies which do not significantly affect their properties and variants which have enhanced or decreased activity and/or affinity.
- the amino acid sequence may be mutated to obtain an antibody with the desired binding affinity to PCSK9.
- Modification of polypeptides is routine practice in the art and need not be described in detail herein. Modification of polypeptides is exemplified in the Examples. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or which mature (enhance) the affinity of the polypeptide for its ligand, or use of chemical analogs.
- Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
- terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to an epitope tag.
- Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody of an enzyme or a polypeptide which increases the half-life of the antibody in the blood circulation.
- Substitution variants have at least one amino acid residue in the antibody molecule removed and a different residue inserted in its place.
- the sites of greatest interest for substitutional mutagenesis include the hypervahable regions, but FR alterations are also contemplated.
- Conservative substitutions are shown in Table 2 under the heading of "conservative substitutions.” If such substitutions result in a change in biological activity, then more substantial changes, denominated “exemplary substitutions" in Table 2, or as further described below in reference to amino acid classes, may be introduced and the products screened.
- Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
- Naturally occurring residues are divided into groups based on common side-chain properties:
- Non-conservative substitutions are made by exchanging a member of one of these classes for another class.
- cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking.
- cysteine bond(s) may be added to the antibody to improve its stability, particularly where the antibody is an antibody fragment such as an Fv fragment.
- Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the variable region. Changes in the variable region can alter binding affinity and/or specificity. In some embodiments, no more than one to five conservative amino acid substitutions are made within a CDR domain. In other embodiments, no more than one to three conservative amino acid substitutions are made within a CDR domain. In still other embodiments, the CDR domain is CDR H3 and/or CDR L3.
- Modifications also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation.
- Antibodies are glycosylated at conserved positions in their constant regions (Jefferis and Lund, 1997, Chem. Immunol. 65:111 -128; Wright and Morrison, 1997, TibTECH 15:26-32).
- the oligosaccharide side chains of the immunoglobulins affect the protein's function (Boyd et al., 1996, MoI. Immunol. 32:1311 -1318; Wittwe and Howard, 1990, Biochem.
- Oligosaccharides may also serve to target a given glycoprotein to certain molecules based upon specific recognition structures. Glycosylation of antibodies has also been reported to affect antibody-dependent cellular cytotoxicity (ADCC).
- CHO cells with tetracycline-regulated expression of ⁇ (1 ,4)-N-acetylglucosaminyltransferase III (GnTIII), a glycosyltransferase catalyzing formation of bisecting GIcNAc, was reported to have improved ADCC activity (Umana et al., 1999, Nature Biotech. 17:176-180).
- N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
- the thpeptide sequences asparagine-X-sehne, asparagine-X-threonine, and asparagine-X-cysteine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
- O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
- glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above- described tripeptide sequences (for N-linked glycosylation sites).
- the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
- the glycosylation pattern of antibodies may also be altered without altering the underlying nucleotide sequence. Glycosylation largely depends on the host cell used to express the antibody.
- glycosylation pattern of the antibodies can be expected (see, e.g., Hse et al., 1997, J. Biol. Chem. 272:9062-9070).
- factors that affect glycosylation during recombinant production of antibodies include growth mode, media formulation, culture density, oxygenation, pH, purification schemes and the like.
- Various methods have been proposed to alter the glycosylation pattern achieved in a particular host organism including introducing or overexpressing certain enzymes involved in oligosaccharide production (U.S. Patent Nos. 5,047,335; 5,510,261 and 5.278,299).
- Glycosylation or certain types of glycosylation, can be enzymatically removed from the glycoprotein, for example, using endoglycosidase H (Endo H), N-glycosidase F, endoglycosidase F1 , endoglycosidase F2, endoglycosidase F3.
- Endo H endoglycosidase H
- N-glycosidase F N-glycosidase F
- endoglycosidase F1 endoglycosidase F2
- endoglycosidase F3 endoglycosidase F3
- the recombinant host cell can be genetically engineered to be defective in processing certain types of polysaccharides.
- Modifications include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay. Modified polypeptides are made using established procedures in the art and can be screened using standard assays known in the art, some of which are described below and in the Examples.
- the antibody comprises a modified constant region, such as a constant region that is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate ADCC, or does not activate microglia; or have reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating ADCC, or activating microglia.
- a modified constant region such as a constant region that is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate ADCC, or does not activate microglia; or have reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating ADCC, or activating microglia.
- Different modifications of the constant region may be used to achieve optimal level and/or combination of effector functions. See, for example, Morgan et al., 1995, Immunology 86:319-324; Lund et al., 1996,
- the constant region is modified as described in Eur. J. Immunol., 1999, 29:2613-2624; PCT Publ. No. WO99/58572; and/or UK Patent Application No. 9809951.8.
- the antibody comprises a human heavy chain lgG2 constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wild type lgG2 sequence). Eur. J. Immunol., 1999, 29:2613-2624.
- the constant region is aglycosylated for N-linked glycosylation.
- the constant region is aglycosylated for N-linked glycosylation by mutating the glycosylated amino acid residue or flanking residues that are part of the N- glycosylation recognition sequence in the constant region.
- N-glycosylation site N297 may be mutated to A, Q, K, or H.
- the constant region is aglycosylated for N-linked glycosylation.
- the constant region may be aglycosylated for N-linked glycosylation enzymatically (such as removing carbohydrate by enzyme PNGase), or by expression in a glycosylation deficient host cell.
- antibody modifications include antibodies that have been modified as described in PCT Publ. No. WO 99/58572. These antibodies comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant domain of a human immunoglobulin heavy chain. These antibodies are capable of binding the target molecule without triggering significant complement dependent lysis, or cell-mediated destruction of the target. In some embodiments, the effector domain is capable of specifically binding FcRn and/or Fc ⁇ Rllb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain C H 2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy.
- affinity matured antibodies can be produced by procedures known in the art (Marks et al., 1992, Bio/Technology, 10:779-783; Barbas et al., 1994, Proc Nat. Acad. Sci, USA 91 :3809- 3813; Schier et al., 1995, Gene, 169:147-155; Yelton et al., 1995, J. Immunol., 155:1994-2004; Jackson et al., 1995, J. Immunol., 154(7):3310-9; Hawkins et al., 1992, J. MoI. Biol., 226:889-896; and PCT Publ. No. WO2004/058184).
- library scanning mutagenesis One way of characterizing a CDR of an antibody and/or altering (such as improving) the binding affinity of a polypeptide, such as an antibody, termed "library scanning mutagenesis".
- library scanning mutagenesis works as follows. One or more amino acid positions in the CDR are replaced with two or more (such as 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, or 20) amino acids using art recognized methods. This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of two or more members (if two or more amino acids are substituted at every position).
- the library also includes a clone comprising the native (unsubstituted) amino acid.
- a small number of clones, e.g., about 20-80 clones (depending on the complexity of the library), from each library are screened for binding affinity to the target polypeptide (or other binding target), and candidates with increased, the same, decreased, or no binding are identified.
- Methods for determining binding affinity are well-known in the art. Binding affinity may be determined using Biacore surface plasmon resonance analysis, which detects differences in binding affinity of about 2-fold or greater. Biacore is particularly useful when the starting antibody already binds with a relatively high affinity, for example a K 0 of about 10 nM or lower. Screening using Biacore surface plasmon resonance is described in the Examples, herein.
- Binding affinity may be determined using Kinexa Biocensor, scintillation proximity assays, ELISA, ORIGEN immunoassay (IGEN), fluorescence quenching, fluorescence transfer, and/or yeast display. Binding affinity may also be screened using a suitable bioassay.
- every amino acid position in a CDR is replaced (in some embodiments, one at a time) with all 20 natural amino acids using art recognized mutagenesis methods (some of which are described herein). This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of 20 members (if all 20 amino acids are substituted at every position).
- the library to be screened comprises substitutions in two or more positions, which may be in the same CDR or in two or more CDRs.
- the library may comprise substitutions in two or more positions in one CDR.
- the library may comprise substitution in two or more positions in two or more CDRs.
- the library may comprise substitution in 3, 4, 5, or more positions, said positions found in two, three, four, five or six CDRs.
- the substitution may be prepared using low redundancy codons. See, e.g., Table 2 of Balint et al., 1993, Gene 137(1 ):109-18).
- the CDR may be CDRH3 and/or CDRL3.
- the CDR may be one or more of CDRL1 , CDRL2, CDRL3, CDRH1 , CDRH2, and/or CDRH3.
- the CDR may be a Kabat CDR, a Chothia CDR, or an extended CDR.
- Candidates with improved binding may be sequenced, thereby identifying a CDR substitution mutant which results in improved affinity (also termed an "improved" substitution).
- Candidates that bind may also be sequenced, thereby identifying a CDR substitution which retains binding.
- candidates each comprising an amino acid substitution at one or more position of one or more CDR
- candidates with improved binding are also useful for the design of a second library containing at least the original and substituted amino acid at each improved CDR position (i.e., amino acid position in the CDR at which a substitution mutant showed improved binding).
- Preparation, screening, and selection of this library is discussed further below.
- Library scanning mutagenesis also provides a means for characterizing a CDR, in so far as the frequency of clones with improved binding, the same binding, decreased binding or no binding also provide information relating to the importance of each amino acid position for the stability of the antibody-antigen complex. For example, if a position of the CDR retains binding when changed to all 20 amino acids, that position is identified as a position that is unlikely to be required for antigen binding. Conversely, if a position of CDR retains binding in only a small percentage of substitutions, that position is identified as a position that is important to CDR function.
- the library scanning mutagenesis methods generate information regarding positions in the CDRs that can be changed to many different amino acids (including all 20 amino acids), and positions in the CDRs which cannot be changed or which can only be changed to a few amino acids.
- Candidates with improved affinity may be combined in a second library, which includes the improved amino acid, the original amino acid, and may further include additional substitutions at that position, depending on the complexity of the library that is desired, or permitted using the desired screening or selection method.
- a second library which includes the improved amino acid, the original amino acid, and may further include additional substitutions at that position, depending on the complexity of the library that is desired, or permitted using the desired screening or selection method.
- and adjacent amino acid position can be randomized to at least two or more amino acids. Randomization of adjacent amino acids may permit additional conformational flexibility in the mutant CDR, which may, in turn, permit or facilitate the introduction of a larger number of improving mutations.
- the library may also comprise substitution at positions that did not show improved affinity in the first round of screening.
- the second library is screened or selected for library members with improved and/or altered binding affinity using any method known in the art, including screening using Biacore surface plasmon resonance analysis, and selection using any method known in the art for selection, including phage display, yeast display, and ribosome display.
- fusion proteins comprising one or more fragments or regions from the antibodies or polypeptides of this invention.
- a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of a variable light chain region shown in SEQ ID NOs: 53, 16, 17, 18, or 19 and/or at least 10 amino acids of a variable heavy chain region shown in SEQ ID NOs: 54, 20, 21 , 22, or 23.
- a fusion polypeptide is provided that comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region.
- the fusion polypeptide comprises a light chain variable region and/or a heavy chain variable region, as shown in any of the sequence pairs selected from among SEQ ID NOs: 53 and 54, 16 and 20, 17 and 21 , 18 and 22, and 19 and 23.
- the fusion polypeptide comprises one or more CDR(s).
- the fusion polypeptide comprises CDR H3 (VH CDR3) and/or CDR L3 (VL CDR3).
- a fusion protein contains one or more antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region.
- heterologous sequences include, but are not limited to a "tag" such as a FLAG tag or a 6His tag.
- tags are well known in the art.
- a fusion polypeptide can be created by methods known in the art, for example, synthetically or recombinantly.
- the fusion proteins of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, although they may also be prepared by other means known in the art, including, for example, chemical synthesis.
- compositions comprising antibodies or polypeptides conjugated (for example, linked) to an agent that facilitate coupling to a solid support (such as biotin or avidin).
- a solid support such as biotin or avidin.
- Conjugation generally refers to linking these components as described herein.
- the linking (which is generally fixing these components in proximate association at least for administration) can be achieved in any number of ways. For example, a direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other.
- a nucleophilic group such as an amino or sulfhydryl group
- a carbonyl-containing group such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.
- An antibody or polypeptide of this invention may be linked to a labeling agent such as a fluorescent molecule, a radioactive molecule or any others labels known in the art.
- Labels are known in the art which generally provide (either directly or indirectly) a signal.
- compositions including pharmaceutical compositions
- kits comprising, as this disclosure makes clear, any or all of the antibodies and/or polypeptides described herein.
- the invention also provides isolated polynucleotides encoding the antibodies and peptides of the invention, and vectors and host cells comprising the polynucleotide.
- the invention provides polynucleotides (or compositions, including pharmaceutical compositions), comprising polynucleotides encoding any of the following: the antibodies 4A5, 5A10, 6F6, 7D4, L1 L3, or any fragment or part thereof having the ability to antagonize PCSK9.
- the invention provides polynucleotides encoding any of the antibodies (including antibody fragments) and polypeptides described herein, such as antibodies and polypeptides having impaired effector function.
- Polynucleotides can be made and expressed by procedures known in the art.
- the invention provides compositions (such as pharmaceutical compositions) comprising any of the polynucleotides of the invention.
- the composition comprises an expression vector comprising a polynucleotide encoding the antibody as described herein.
- the composition comprises an expression vector comprising a polynucleotide encoding any of the antibodies or polypeptides described herein.
- the composition comprises either or both of the polynucleotides shown in SEQ ID NO:25 and SEQ ID NO:26. Expression vectors, and administration of polynucleotide compositions are further described herein.
- the invention provides a method of making any of the polynucleotides described herein.
- Polynucleotides complementary to any such sequences are also encompassed by the present invention.
- Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules.
- RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
- Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence.
- Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein.
- Variants preferably exhibit at least about 70% identity, more preferably, at least about 80% identity, yet more preferably, at least about 90% identity, and most preferably, at least about 95% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
- Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
- a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, 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.
- Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wl), using default parameters.
- This program embodies several alignment schemes described in the following references: Dayhoff, M.O., 1978, A model of evolutionary change in proteins - Matrices for detecting distant relationships.
- Dayhoff, M.O. ed. Atlas of Protein Sequence and Structure (National Biomedical Research Foundation, Washington DC), Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol.
- the "percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (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 bases 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 reference sequence (i.e.
- variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof.
- Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence). Suitable "moderately stringent conditions" include prewashing in a solution of 5 X
- SSC 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65°C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0.1 % SDS.
- highly stringent conditions or “high stringency conditions” are those that: (1 ) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1 % sodium dodecyl sulfate at 50 0 C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1 % bovine serum albumin/0.1 % Ficoll/0.1 % polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42°C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCI, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1 % sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/ml), 0.1 % SDS
- nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides.
- the resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
- the polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
- a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein.
- Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome.
- the polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al., 1989, supra.
- PCR allows reproduction of DNA sequences.
- PCR technology is well known in the art and is described in U.S. Patent Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, MuIMs et al., 1994, eds. (Birkauswer Press, Boston, MA).
- RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., 1989, supra, for example.
- Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector.
- Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Blueschpt (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, pMB9, CoIEI , pCR1 , RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
- plasmids and bacterial viruses e.g., pUC18, pUC19, Blueschpt (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, pMB9, CoIEI , pCR1 , RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
- cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen.
- Expression vectors generally are replicable polynucleo
- Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publ. No. WO 87/04462.
- Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator).
- suitable transcriptional controlling elements such as promoters, enhancers and terminator
- one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
- the vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE- dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus).
- electroporation employing calcium chloride, rubidium chloride, calcium phosphate, DEAE- dextran, or other substances
- microprojectile bombardment e.g., where the vector is an infectious agent such as vaccinia virus.
- infection e.g., where the vector is an infectious agent such as vaccinia virus.
- the choice of introducing vectors or polynucleotides will often depend on features of the host cell.
- the invention also provides host cells comprising any of the polynucleotides described herein. Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest.
- mammalian host cells include but are not limited to COS, HeLa, NSO, and CHO cells. See also PCT Publ. No. WO 87/04462.
- Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe; or K. lactis).
- the host cells express the cDNAs at a level of about 5 fold higher, more preferably, 10 fold higher, even more preferably, 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells.
- Screening the host cells for a specific binding to PCSK9 or a PCSK9 domain is effected by an immunoassay or FACS.
- a cell overexpressing the antibody or protein of interest can be identified.
- compositions used in the methods of the invention comprise an effective amount of a PCSK9 antagonist antibody, a PCSK9 antagonist antibody derived polypeptide, or other PCSK9 antagonists described herein. Examples of such compositions, as well as how to formulate them, are also described in an earlier section and below.
- the composition further comprises a PCSK9 antagonist.
- the composition comprises one or more PCSK9 antagonist antibodies.
- the PCSK9 antagonist antibody recognizes human PCSK9.
- the PCSK9 antagonist antibody is humanized.
- the PCSK9 antagonist antibody comprises a constant region that does not trigger an unwanted or undesirable immune response, such as antibody- mediated lysis or ADCC.
- the PCSK9 antagonist antibody comprises one or more CDR(s) of the antibody (such as one, two, three, four, five, or, in some embodiments, all six CDRs).
- the PCSK9 antagonist antibody is human.
- compositions can comprise more than one PCSK9 antagonist antibody (e.g., a mixture of PCSK9 antagonist antibodies that recognize different epitopes of PCSK9).
- Other exemplary compositions comprise more than one PCSK9 antagonist antibodies that recognize the same epitope(s), or different species of PCSK9 antagonist antibodies that bind to different epitopes of PCSK9.
- composition used in the present invention can further comprise pharmaceutically acceptable carriers, excipients, or stabilizers (Remington: The Science and Practice of Pharmacy 20th Ed., 2000, Lippincott Williams and Wilkins, Ed. K. E. Hoover), in the form of lyophilized formulations or aqueous solutions.
- Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
- the antibody is administered in a formulation as a sterile aqueous solution having a pH that ranges from about 5.0 to about 6.5 and comprising from about 1 mg/ml to about 200 mg/ml of antibody, from about 1 millimolar to about 100 millimolar of histidine buffer, from about 0.01 mg/ml to about 10 mg/ml of polysorbate 80, from about 100 millimolar to about 400 millimolar of trehalose, and from about 0.01 millimolar to about 1.0 millimolar of disodium EDTA dihydrate.
- the PCSK9 antagonist antibody and compositions thereof can also be used in conjunction with other agents that serve to enhance and/or complement the effectiveness of the agents.
- Kits The invention also provides kits for use in the instant methods.
- Kits of the invention include one or more containers comprising a PCSK9 antagonist antibody (such as a humanized antibody) or peptide described herein and instructions for use in accordance with any of the methods of the invention described herein.
- these instructions comprise a description of administration of the PCSK9 antagonist antibody, peptide, or aptamer for the above described therapeutic treatments.
- the antibody is a humanized antibody. In some embodiments, the antibody is human. In other embodiments, the antibody is a monoclonal antibody.
- the instructions relating to the use of a PCSK9 antagonist antibody generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
- the containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
- the kits of this invention are in suitable packaging.
- Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Also contemplated are packages for use in combination with a specific device, such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump.
- a kit may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
- the container may also have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
- At least one active agent in the composition is a PCSK9 antagonist antibody.
- the container e.g., pre-filled syringe or autoinjector
- Kits may optionally provide additional components such as buffers and interpretive information.
- the kit comprises a container and a label or package insert(s) on or associated with the container.
- DNA fragments encoding V H and V L regions can first be obtained using any of the methods described above.
- Various modifications, e.g., mutations, deletions, and/or additions can also be introduced into the DNA sequences using standard methods known to those of skill in the art.
- mutagenesis can be carried out using standard methods, such as PCR-mediated mutagenesis, in which the mutated nucleotides are incorporated into the PCR primers such that the PCR product contains the desired mutations or site-directed mutagenesis.
- substitution for example, that may be made is to change one or more cysteines in the antibody, which may be chemically reactive, to another residue, such as, without limitation, alanine or serine.
- alanine or serine for example, there can be a substitution of a non-canonical cysteine.
- the substitution can be made in a CDR or framework region of a variable domain or in the constant domain of an antibody.
- the cysteine is canonical.
- the antibodies may also be modified, e.g., in the variable domains of the heavy and/or light chains, e.g., to alter a binding property of the antibody.
- a mutation may be made in one or more of the CDR regions to increase or decrease the K 0 of the antibody for PCSK9, to increase or decrease k O ff, or to alter the binding specificity of the antibody.
- Techniques in site-directed mutagenesis are well-known in the art. See, e.g., Sambrook et al. and Ausubel et al., supra.
- a modification or mutation may also be made in a framework region or constant domain to increase the half-life of a PCSK9 antibody. See, e.g., PCT Publ. No.
- a mutation in a framework region or constant domain can also be made to alter the immunogenicity of the antibody, to provide a site for covalent or non-covalent binding to another molecule, or to alter such properties as complement fixation, FcR binding and antibody-dependent cell-mediated cytotoxicity.
- a single antibody may have mutations in any one or more of the CDRs or framework regions of the variable domain or in the constant domain.
- V H and V L sequences In a process known as "germlining", certain amino acids in the V H and V L sequences can be mutated to match those found naturally in germline V H and V L sequences.
- the amino acid sequences of the framework regions in the V H and V L sequences can be mutated to match the germline sequences to reduce the risk of immunogenicity when the antibody is administered.
- Germline DNA sequences for human V H and V L genes are known in the art (see e.g., the "Vbase” human germline sequence database; see also Kabat, E. A., et al. (1991 ) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publ. No. 91-3242; Tomlinson et al., 1992, J. MoI. Biol. 227:776-798; and Cox et al., 1994, Eur. J. Immunol. 24:827-836.
- Another type of amino acid substitution that may be made is to remove potential proteolytic sites in the antibody. Such sites may occur in a CDR or framework region of a variable domain or in the constant domain of an antibody. Substitution of cysteine residues and removal of proteolytic sites may decrease the risk of heterogeneity in the antibody product and thus increase its homogeneity.
- Another type of amino acid substitution eliminates asparagine-glycine pairs, which form potential deamidation sites, by altering one or both of the residues.
- the C-terminal lysine of the heavy chain of a PCSK9 antibody of the invention can be cleaved.
- the heavy and light chains of the PCSK9 antibodies may optionally include a signal sequence.
- V H and V L segments of the present invention are obtained, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to convert the variable region genes to full- length antibody chain genes, to Fab fragment genes, or to a scFv gene.
- a V L - or V H -encoding DNA fragment is operatively linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker.
- the term "operatively linked”, as used in this context, is intended to mean that the two DNA fragments are joined such that the amino acid sequences encoded by the two DNA fragments remain in-frame.
- the isolated DNA encoding the V H region can be converted to a full-length heavy chain gene by operatively linking the V H -encoding DNA to another DNA molecule encoding heavy chain constant regions (CH1 , CH2 and CH3).
- the sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat, E. A., et al., 1991 , Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publ. No. 91 -3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
- the heavy chain constant region can be an IgGI , lgG2, lgG3, lgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an IgGI or lgG2 constant region.
- the IgG constant region sequence can be any of the various alleles or allotypes known to occur among different individuals, such as Gm(1 ), Gm(2), Gm(3), and Gm(17). These allotypes represent naturally occurring amino acid substitution in the IgGI constant regions.
- the V H -encoding DNA can be operatively linked to another DNA molecule encoding only the heavy chain CH1 constant region.
- the CH1 heavy chain constant region may be derived from any of the heavy chain genes.
- the isolated DNA encoding the V L region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the V L -encoding DNA to another DNA molecule encoding the light chain constant region, C L .
- the sequences of human light chain constant region genes are known in the art (see e.g., Kabat, E. A., et al., 1991 , Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publ. No. 91 -3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
- the light chain constant region can be a kappa or lambda constant region.
- the kappa constant region may be any of the various alleles known to occur among different individuals, such as lnv(1 ), lnv(2), and lnv(3).
- the lambda constant region may be derived from any of the three lambda genes.
- the V H - and V L -encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (GIy 4 -Ser) 3 , such that the V H and V L sequences can be expressed as a contiguous single-chain protein, with the V L and V H regions joined by the flexible linker (See e.g., Bird et al., 1988, Science 242:423-426; Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al., 1990, Nature 348:552-554.
- a flexible linker e.g., encoding the amino acid sequence (GIy 4 -Ser) 3 , such that the V H and V L sequences can be expressed as a contiguous single-chain protein, with the V L and V H regions joined by the flexible linker
- the single chain antibody may be monovalent, if only a single V H and V L are used, bivalent, if two V H and V L are used, or polyvalent, if more than two V H and V L are used. Bispecific or polyvalent antibodies may be generated that bind specifically to PCSK9 and to another molecule.
- a fusion antibody or immunoadhesin may be made that comprises all or a portion of a PCSK9 antibody of the invention linked to another polypeptide.
- only the variable domains of the PCSK9 antibody are linked to the polypeptide.
- the V H domain of a PCSK9 antibody is linked to a first polypeptide, while the V L domain of a PCSK9 antibody is linked to a second polypeptide that associates with the first polypeptide in a manner such that the V H and V L domains can interact with one another to form an antigen binding site.
- the V H domain is separated from the V L domain by a linker such that the V H and V L domains can interact with one another.
- V H -linker-V L antibody is then linked to the polypeptide of interest.
- fusion antibodies can be created in which two (or more) single-chain antibodies are linked to one another. This is useful if one wants to create a divalent or polyvalent antibody on a single polypeptide chain, or if one wants to create a bispecific antibody.
- other modified antibodies may be prepared using PCSK9 antibody encoding nucleic acid molecules.
- PCSK9 antibody encoding nucleic acid molecules For instance, "Kappa bodies” (III et al., 1997, Protein Eng. 10:949-57), “Minibodies” (Martin et al., 1994, EMBO J. 13:5303-9), “Diabodies” (Holliger et al., 1993, Proc. Natl. Acad. Sci. USA 90:6444-6448), or “Janusins” (Traunecker et al., 1991 , EMBO J. 10:3655-3659 and Traunecker et al., 1992, Int. J. Cancer (Suppl.) 7:51 -52) may be prepared using standard molecular biological techniques following the teachings of the specification.
- Bispecific antibodies or antigen-binding fragments can be produced by a variety of methods including fusion of hybhdomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, 1990, Clin. Exp. Immunol. 79:315-321 , Kostelny et al., 1992, J. Immunol. 148:1547-1553.
- bispecific antibodies may be formed as "diabodies" or "Janusins.”
- the bispecific antibody binds to two different epitopes of PCSK9.
- the modified antibodies described above are prepared using one or more of the variable domains or CDR regions from a human PCSK9 antibody provided herein.
- Hybridomas Representative antibodies (hybhdomas) of the present invention were deposited in the American Type Culture Collection (ATCC) on February 28, 2008, and were assigned the accession numbers in Table 3. Hybridomas were deposited for antibodies 4A5, 5A10, 6F6 and 7D4.
- Example 1 Generating and Screening PCSK9 Antagonist Antibodies General procedures for immunization of animals for generating monoclonal antibodies:
- PCSK9 -/- that is, null or knock-out mice
- antigen was prepared by mixing the recombinant proteins with adjuvant, lmmunogen was given via injection to the scruff of the neck, the foot pads and intrapehtoneally, approximately every 3 days over the course of 11 days, with the last boost administered i.v., without adjuvant.
- mice On Day 13, the mice were euthanized and their spleens were removed. Lymphocytes were immortalized by fusion with an established cell line to make hybhdoma clones using standard hybridoma technology, distributed into 96 well plates. Clones were allowed to grow, then selected by ELISA screening using the immunizing antigen, as below.
- ELISA screening of antibodies Supernatant media from growing hybridoma clones were screened separately for their ability to bind the recombinant human PCSK9 or recombinant mouse PCSK9. The assays were performed with 96-well plates coated overnight with 100 ⁇ l of a 1 ⁇ g/ml solution of one of the antigens. Excess reagents were washed from the wells between each step with PBS containing 0.05% Tween-20. Plates were then blocked with PBS containing 0.5% BSA. Supernatant was added to the plates and incubated at room temperature for 2 hours. Horse radish peroxidase (HRP) conjugated goat-anti mouse Fc was added to bind to the mouse antibodies bound to the antigen.
- HRP horse radish peroxidase
- Tetramethyl benzidine was then added as substrate for HRP to detect the amount of mouse antibody present in the supernatant. The reaction was stopped and the relative amount of antibody was quantified by reading the absorbance at 450 nm. Hybridoma clones that secreted antibodies that are capable of binding to either mouse or human PCSK9 were selected for further analysis.
- PCSK9-mediated LDLR down-regulation in Huh7 cells Hybridoma clones secreting human or mouse PCSK9 binding antibodies were expanded and supernatants were harvested. Total IgGs were purified from approximately 10 ml of the supernatant using protein A beads, dialyzed into PBS buffer, and the final volume reduced to yield solutions with 0.7-1 mg/ml of antibodies. Purified antibodies were then used to test their ability to inhibit the ability of PCSK9 to mediate LDLR down-regulation in Huh7 cells. Huh7 cells were plated and allowed to grow to 80% confluency in RPMI media containing 10% FBS, 4 mM glutamine, and penicillin and streptavidin in 96 well plates.
- the medium was changed to one containing 10% de- lipidated FBS for 8-16 hrs to induce LDLR expression.
- Cells were then incubated for 8- 16 hours with 40 ⁇ l/well of 293 expression media supplemented with 6 ⁇ g/ml of human (preferably) or mouse PCSK9, with or without 70-100 ⁇ g/ml of test antibodies.
- the PCSK9 and antibody containing media were removed at the end of incubation, and cells were lysed with 17 ⁇ l lysis buffer by shaking at 4 C for an hour.
- the lysis buffer consisted of 50 mM glycerol phosphate, 10 mM HEPES pH 7.4, 1 % Triton X-100, 20 mM NaCI, and a cocktail of protease inhibitors (Roche).
- Cell lysates were collected and analyzed for LDLR protein levels via staining of Western blots following SDS polyacrylamide gel electrophoresis. Hybhdoma clones producing antibodies that can partially or fully rescue LDLR level were selected for further analysis.
- LDLR down regulation assay is meant the above assay using Huh7 cells.
- Figure 1 illustrates the effect of anti-PCSK9 antagonistic monoclonal antibodies 7D4.4, 4A5.G3, 6F6.G10.3 and 5A10.B8 on the ability of human and mouse PCSK9 to down regulate LDLR in cultured Huh7 cells.
- 100 nM of mouse or human recombinant PCSK9, and a serial dilution of 25-800 nM of antibodies were used.
- the figures are Western blots showing that the antibodies are in general more effective in blocking the function of human PCSk9 than mouse PCSK9.
- the several antibodies have generally similar affinities for human PCSK9 but vary in their affinity for murine PCSK9.
- affinities of PCSK9 antibodies to PCSK9 were measured on a surface plasmon resonance Biacore 3000 biosensor equipped with a research-grade sensor chip using HBS-EP running buffer (Biacore AB, Uppsala, Sweden - now GE Healthcare).
- Rabbit polyclonal anti-Ms IgGs were amine-coupled at saturating levels onto the chip using a standard N-hydroxysuccinimide/ ethyldimethylaminopropyl carbodiimide (NHS/EDC) chemistry.
- the buffer was switched to HBS-EP + 1 mg/mL BSA + 1 mg/mL CM-dextran.
- PCSK9 IgGs Full-length PCSK9 IgGs were diluted to about 15 ⁇ g/mL and captured for 1 min at 5 ⁇ L/min to give levels of about 500RU per flow cell, leaving one blank to serve as a reference channel. 3.73-302 nM hPCSK9 or 2.54-206 nM mPCSK9 were injected as a 5-membered 3-fold series for 1 min at 100 ⁇ L/min. Dissociation was monitored for 5 min. The chip was regenerated after the last injection of each titration with two 30 sec pulses of 100 mM phosphoric acid. Buffer cycles provided blanks for double-referencing the data, which were then fit globally to a simple binding model using Biaevaluation software v.4.1.
- PCSK9 has been shown to bind LDLR with an affinity of 180 nM under neutral pH (Cunningham et al., 2007, Nat Struct MoI Biol, 14(5):413-9).
- Recombinant mouse or human PCSK9 protein was biotinylated using the Pierce reagents following the manufacture's instructions.
- ELISA plates (Corning Mixisorb) were coated with a solution of 1 ⁇ g/ml recombinant LDLR extracellular domain (R&D Systems) in each well at 4 C overnight, blocked with 2% BSA + PBS for 2 hrs at room temperature, and then washed 5 times with washing buffer (1x PBS + 0.05% Tween-20).
- LDLR-PCSK9 binding can be stabilized by adding 50 ⁇ l of 4% FDH + 4% sucrose + PBS solution and incubate for 5 min.
- Wells were washed 5 times with washing buffer, incubated with 1 :2000 dilution of HRP conjugated Strepavidin (Invitrogen) for 1 hr at RT, washed 5 times with washing buffer.
- TMB substrate was added to the wells, the solution was incubated 20 to 30 min at RT, and the reaction was terminated using 1 M phosphoric acid. Signals were read at 450 nm.
- Figure 2 illustrates the dose-response of anti-PCSK9 antagonist monoclonal antibodies 6F6.G10.3, 7D4.4, 4A5.G3, 5A10.B8, negative control antibody 42H7, and PBS on blocking the binding of recombinant biotinylated human PCSK9 and mouse PCSK9 to immobilized recombinant LDLR extracellular domain in vitro.
- Part A) shows human PCSK9 binding to human LDLR extracellular domain and that 7D4, 4A5, 5A10, and 6F6 are effective in blocking binding, whereas 42H7 and PBS are not.
- Part B) shows mouse PCSK9 binding to human LDLR extracellular domain.
- Figure 3 illustrates the dose-response of anti-PCSK9 monoclonal antagonist antibodies 6F6.G10.3, 7D4.4, 4A5.G3 and 5A10.B8 on blocking binding of recombinant biotinylated human PCSK9 (30 nM) to Europium labeled recombinant LDLR extracellular domain (10 nM) in solution at neutral pH in vitro. This assay measures binding in free solution at neutral pH.
- Example 4 Epitope Mapping/Binding of Antibodies using the Crystal Structure of the L1 L3PCSK9 Complex, Biacore, and Mutagenesis a. Crystal structure of the L1 L3PCSK9 complex. The residues were identified by calculating the difference in accessible surface area between the L1 L3PCSK9 crystal structure and PCSK9 structure alone. PCSK9 residues that show buried surface area upon complex formation with L1 L3 antibody were included as a part of the epitope.
- the solvent accessible surface of a protein was defined as the locus of the centre of a probe sphere (representing a solvent molecule of 1.4 A radius) as it rolls over the Van der Waals surface of the protein.
- the solvent accessible surface area was calculated by generating surface points on an extended sphere about each atom (at a distance from the atom centre equal to the sum of the atom and probe radii), and eliminating those that lie within equivalent spheres associated with neighboring atoms as implemented in program AREAIMOL (Briggs, P.J., 2000, CCP4 Newsletter No. 38, CCLRC, Daresbury).
- AREAIMOL program AREAIMOL
- Figure 23A shows the crystal structure of the PCSK9 (light gray surface representation) bound to the L1 L3 antibody (black cartoon representation).
- the epitope for L1 L3 binding to PCSK9 involves residues 153-155, 194, 197, 237-239, 367, 369, 374-379 and 381 of the PCSK9 amino acid sequence (SEQ ID NO:53).
- the epitope for the LDLR EGF domain binding to PCSK9 involves residues 153-155, 194, 238, 367, 369, 372, 374-375, and 377-381 (Kwon et al., 2008, PNAS 105: 1820-1825).
- IgGs Full- length IgGs were amine-coupled to a CM5 sensor chip (three per chip at about 7000RU final), using a standard EDC/NHS-mediated amine-coupling chemistry. One flow cell was left unmodified to provide a reference channel. Human-PCSK9 (100 nM) was premixed with an array of IgGs (final 500 nM) and these complexes were injected over the chip using 1 min injections at 10 ⁇ L/min. Antibodies that bind to competing epitopes will block the binding of PCSK9 to the antibody immobilized on the chip.
- a classical sandwich approach was used by first injecting human-PCSK9 at 50 nM for 1 min at 10 ⁇ L/min (to tether it via the IgG on the chip) and then binding an array of IgGs (final 500 nM each) for 2 mins each.
- the immobilized IgGs were regenerated with a mild acid (Pierce gentle elution buffer + 1 M NaCI).
- Antibodies directed to known different epitopes were used as controls for positive sandwich formation in this assay.
- Structure-guided mutagenesis to map antibody binding epitopes.
- PCSK9 surface-residue mutants F379A, I369A, R194A, D374Y, D238R, T377R, K222A, R199A, F216A, R218A, R237A, D192R, D367R, R165A, R167A, A443T, A53V, I474V, H449A
- F379A, I369A, R194A, D374Y, D238R, T377R, K222A R199A, F216A, R218A, R237A, D192R, D367R, R165A, R167A, A443T, A53V, I474V, H449A
- the 19 single point mutants were generated from the previously described wild-type DNA construct (Cunningham et al., 2007, supra) using standard DNA techniques.
- the mutant proteins were expressed using transient transfection in HEK293T cells and secreted into the cell media.
- the mutant proteins were purified with the high-throughput AKTA Xpress system (GE Healthcare) by Ni 2+ and size-exclusion chromatography steps, using conditions similar to those described earlier. Protein concentrations were determined using the LabChip instrument (Bio- Rad).
- PCSK9-blocking murine antibodies 4A5, 7D4, 5A10 and 6F6 were expressed with transient transfection in HEK293F cells and purified with a protein G column eluted with 0.1 M Glycine buffer at pH 2.8 and neutralized into 1.0 M Tris at pH 9.0. e.
- the regions of PCSK9 that are contacted by monoclonal antibodies 5A10 and 7D4 (preparation described later herein) were determined by protein tomography (Sidec AB, Sweden).
- the loops at positions 186-200, 371 -379, 176-181 , 278-283, 449-453, 402-406, and 236-245 of PCSK9 were proximal to amino acid residues of the antibody.
- the sequences corresponding to the loops are shown in Table 5, and in a preferred embodiment, the antagonists of the invention bind to one or more of these sequences in PSCK9.
- Mutant binding experiments were performed using Biacore 3000 at 25 0 C with a running buffer of 50 mM Tris-HCI pH 7.5, 150 mM NaCI and 0.02% P20. Mutants were tested at 333 nM or 111 nM concentrations in duplicates, with the ones giving weakened binding compared to the wild-type as the residues involved in mAb binding (listed below).
- RNAeasy Micro kit from QIAGEN.
- cDNA was synthesized using Superscript III RT kit from Invitrogen.
- Variable regions from the PCSK9 antibodies were cloned using the mouse IgG-Primer Sets from Novagen, which consist of degenerate primers for cloning mouse IgG heavy chain genes and the mouse kappa or lambda light chains.
- PCR cycling conditions were the followings: 1 cycle at 92 C for 2 min; two cycles at 94 C for 30 sec, 44 C for 30 sec and 72 C for 2 min; two cycles at 94 C for 30 sec, 46 C for 30 sec and 72 C for 2 min; two cycles at 94 C for 30 sec, 48 C for 30 sec and 72 C for 2 min; two cycles at 94 C for 30 sec, 50 C for 30 sec and 72 C for 2 min; two cycles at 94 C for 30 sec, 52 C for 30 sec and 72 C for 2 min; followed by 35 cycles at 94 C for 30 sec, 54 C for 30 sec and 72 C for 45 sec.
- the resulting PCR products were cloned into Topo-TA cloning vector from Invitrogen and sequenced. The cloned antibody sequences were confirmed by N- terminal sequencing of the first 10 amino acids of the original antibodies produced from ascites.
- Example 6 Generation of Antigens for Immunization
- Recombinant human PCSK9 protein was produced as reported Cunningham et al., 2007, Nat Struct MoI Biol, 14(5):413-9.
- To produce recombinant mouse PCSK9 protein the cDNA of mouse PCSK9 was cloned into mammalian expression vector PRK5 with the addition of a 6-His tag at the C-terminus by methods known in the art, transiently transfected and expressed in HEK293 cells.
- Recombinant protein was purified from conditioned media using a Ni column.
- PCSK9-Specific Antibodies as PCSK9 Antagonists 1. Identification of PCSK9-specific antagonist antibodies a. Identification of PCSK9-blocking antibodies
- Murine antibodies to human and/or mouse PCSK9 were generated by immunizing mice with human-PCSK9 and mouse-PCSK9 synthetic peptides as prepared in Example 6 or recombinant proteins, and screening antibodies by ELISA assay using human and/or mouse PCSK9 recombinant protein as the antigens as described in Example 1 and other standard hybhdoma procedures. Over 500 positive clones were obtained and allowed to grow to confluency in 6 well plates with 10 ml media. Media supernatant were collected and total IgGs in the conditioned media were purified using mAb Select (Pierce). The ability of purified and concentrated mouse IgGs to inhibit mouse and human PCSK9 function was tested in Huh7 cells using the methods described in Example 1.
- Hybridoma clones expressing IgGs that showed some degrees of blocking were expanded and retested. 60 promising clones were subcloned, expanded, and injected into either Balb/c or nude mice to produce ascites. Antibodies purified from ascites fluid were retested for their ability to inhibit the down regulation of LDLR by human or mouse PCSK9 in Huh7 cells. Four hybridoma clones, 4A5, 5A10, 6F6, and 7D4, were identified as being able to completely inhibit human PCSK9 function, and at least partially inhibit mouse PCSK9 function.
- PCSK9 has been shown to be co-localized with LDLR in cellular compartments (Lagace et al., 2006, J Clin Inv, 116(11 ):2995-3005. Recombinant PCSK9 protein also binds to LDLR extracellular domain in vitro (Fisher et al., 2007, JBC, 282(28):20502-12.
- PCSK9 antibodies that partially or completely blocked PCSK9 function on LDLR and representatives of antibodies that do not block. All partial antagonistic antibodies also partially inhibited LDLR extracellular domain binding to PCSK9, except one.
- Antagonistic antibodies that can completely block PCSK9 function, namely 4A5, 5A10, 6F6 and 7D4 also completely inhibited LDLR extracellular domain binding to PCSK9 (Table 5). IC 5 O values of these four antibodies correlates with their binding affinity to PCSK9.
- Figure 4 illustrates the epitope binning of anti-PCSK9 antibodies.
- Part A) shows epitope information of anti-PCSK9 mAbs, determined by binding to synthetic 13-18-mer peptides or epitope binding via Biacore.
- Part B) shows the ability of immobilized antibodies 6F6, 5A10 and 4A5 to bind to human PCSK9 premixed with the mAbs indicated on the y axis by Biacore assay.
- 6G7 Another monoclonal anti-PCSK9 antibody, termed 6G7, binds to recombinant mouse PCSK9 but not human PCSK9. See Table 6. 6G7, 4A5, 5A10, 6F6, and 7D4 mutually exclude each other's binding to mouse PCSK9. Chimera analysis between mouse and human PCSK9 reveals that 6G7 binding to PCSK9 requires the catalytic domain. See Table 6. Thus the binding sites of 4A5, 5A10, 6F6, and 7D4 overlap the catalytic site and/or the epitope bound by 6G7.
- antibodies were incubated with plasma from different species and the resultant complexes were purified and probed by an independent anti PCSK9 antibody on Western blots.
- the antibodies 4A5, 5A10, 6F6, and 7D4 recognized human, cynomolgus monkey, mouse, and rat PCSK9. See Figure 5.
- Antibody 6G7 recognized only murine PCSK9 and an unrelated control antibody 42H7 did not recognize any tested PCSK9. Id.
- the amino acid sequences of the variable domains of PCSK9 antibodies 4A5, 5A10, 6F6, and 7D4 were determined using the method described in Example 5. The sequences indicate that the antibodies are related but different from each other.
- Table 1 shows the amino acid sequences of the variable regions of each antibody.
- Table 7 shows the CDR sequences of the light chains and heavy chains of Table 1 as identified by the Kabat and Chotia methods.
- Anti-PCSK9 IgGs 4A5, 5A10 and 6F6 were amine coupled to the Biacore chip.
- hPCSK9 100 nM was mixed with 500 nM of 4A5, 5A10, 6F6 or 7D4 in various ratios and injected for 1 min at 10 ⁇ l/min.
- the four antibodies mutually blocked one another irrespective of the assay orientation tested, suggesting that they all bind to competing epitopes. In contrast, they are able to form sandwich complexes with other non-fully- blocking antibodies that were mapped to specific regions using synthetic peptides.
- PCSK9 specific antibodies as PCSK9 antagonist in vivo a.
- PCSK9 antagonist antibodies lower serum cholesterol in mice
- PCSK9 antagonist monoclonal antibodies can affect cholesterol levels in vivo by inhibiting the function of extracellular PCSK9
- the effect of 7D4 was tested against mouse PCSK9 in vitro, on serum cholesterol when injected into mice. 6 to 7 week old male C57/bl6 mice were kept on a 12 hr light/dark cycle, bled to collect approximately 70 ⁇ l serum on day -7.
- Antagonist PCSK9 antibody 7D4, and a control isotype matching monoclonal antibody were injected into male 7 week old C57/bl6 mice via i.p. injections on days 0, 1 , 2, and 3. Mice were sacrificed on day 4 without fasting, and serum samples were collected.
- FIG. 7 illustrates that a partial antagonist polyclonal anti-PCSK9 mAb CRN6 does not affect cholesterol levels in mice.
- Figure 7A shows that CRN6 antibody partially blocks PCSK9 mediated down regulation of LDLR in Huh7 cells in vitro.
- Figure 7B shows that administration of CRN6 antibody does not affect serum cholesterol levels in mice.
- Part B shows HDL cholesterol. LDL cholesterol levels were very low.
- Figure 9 illustrates that the anti-PCSK9 antagonist mAb 7D4 dose dependently reduces serum total cholesterol, HDL, and LDL in mice.
- Figure 9A shows total cholesterol levels, which decreased to less than 60% of control after administration of 3 to 30 mg/kg/day.
- Figure 9B shows HDL levels, which decreased to less than 70% after administration of 3 to 30 mg/kg/day.
- Figure 9C shows LDL levels, which decreased to nearly zero at all tested doses of 0.3 mg/kg/day and above.
- Figure 10 illustrates that anti-PCSK9 antagonist antibody 5A10 dose dependently lowers cholesterol levels in mice.
- FIG 11 illustrates that anti-PCSK9 antagonist antibodies 4A5 and 6F6 lower cholesterol levels in mice in a dose-dependent fashion.
- the antibody 4A5 showed a graduated decrease in total serum cholesterol with increasing dose of antibody.
- the antibody 6F6 showed decrease in total serum cholesterol at 10 mg/kg/day.
- Anti-PCSK9 antagonist antibodies 4A5, 5A10, 6F6 and 7D4 increase liver LDLR levels in mice as found by Western blot analysis. See Figure 12.
- mice For 7D4, 8 week old BI6/c57 mice were administered with 10 mg/kg of antibodies on days 0, 1 , 2, and 3 via i. p. bolus injection, animals were sacrificed on day 4, and whole liver lysate of 3 individual animals were analyzed for LDLR and GAPDH protein levels by Western blot. All antibody-treated mice showed high levels of LDLR as compared to the PBS control mice.
- FIG. 13 illustrates that anti-PCSK9 antagonist antibody has no effect in the
- Figure 14 illustrates that multiple treatments of anti-PCSK9 antagonist antibodies in mice can substantially decrease total serum cholesterol.
- Eight week old C57/bl6 mice were administered the indicated doses of antibodies or PBS on days 0, 7, 14 and 21 by i.v. bolus injection.
- Example 8 PCSK9 Antagonist Antibodies Lower Serum LDL in Non-human Primates
- antibody 7D4 was tested in cynomolgus monkeys.
- Four 3-4 year old cynomolgus monkey were injected with vehicle (PBS+0.01 % Tween 20) on day 0, and 10 mg/kg 7D4 on day 7.
- Plasma lipid profiles were analyzed on days 0, 2, 7, 9, 11 , 14, 21 and 28 following overnight fasting.
- Two male and two female cynomolgus monkeys 3-5 years of age in each group were given the indicated dose of 7D4 on day 7 and an equal volume of saline on day 0 by i.v. bolus injection. Plasma samples were taken at indicated time points and plasma LDL levels were measured.
- Figure 17 illustrates a comparison of anti-PCSK9 antibodies 4A5, 5A10, 6F6 and 7D4 on serum cholesterol levels in the cynomolgus monkey.
- Two male and two female cynomolgus monkeys 3-6 years of age in each group were given 1 mg/kg of the indicated antibody on day 0 by i.v. bolus injection.
- Plasma samples were taken at indicated time points, plasma LDL levels were measured and normalized to that on day - 2.
- Figure 18 illustrates the effect of anti-PCSK9 antagonist antibody 7D4 on plasma cholesterol levels of cynomolgus monkeys fed a 33.4% kcal fat diet supplemented with 0.1 % cholesterol.
- Six 3-5 year old cynomolgus monkeys were put on high-fat diet for 16 weeks.
- Three monkeys were treated with 10 mg/kg 7D4 and three with saline on the indicated date. LDL levels of individual monkeys were measured and normalized to that of the treatment day.
- the murine monoclonal antibody 5A10 was humanized and affinity matured to provide the L1 L3 antibody.
- L1 L3 has an affinity for murine PCSK9 of 200 pM and an affinity for human PCSK9 of 100 pM when measured by Biacore.
- L1 L3 completely inhibits the PCSK9-mediated down regulation of LDLR in cultured Huh7 cells when incubated with 100 nM human or murine PCSK9 antibody. See Figure 19.
- Figure 20 illustrates the dose-response of L1 L3, mouse precursor 5A10, and negative control antibody 42H7 to block the binding of recombinant biotinylated human PCSK9 and mouse PCSK9 to immobilized recombinant LDLR extracellular domain in vitro.
- Figure 2OA shows human PCSK9 binding to human LDLR extracellular domain at pH 7.5.
- Figure 2OB shows human PCSK9 binding to human LDLR extracellular domain at pH 5.3.
- Figure 2OC shows mouse PCSK9 binding to human LDLR extracellular domain at pH 7.5.
- Figure 2OD shows mouse PCSK9 binding to human LDLR extracellular domain at pH 5.3.
- Figure 21 shows the effect on serum cholesterol of treatment with 10 mg/kg L1 L3 in mice.
- amino acid sequence of L1 L3 fully humanized heavy chain (SEQ ID NO:15) is shown in Table 8.
- sequence of the variable region is underlined (SEQ ID NO: 54).
- amino acid sequence of L1 L3 fully humanized light chain (SEQ ID NO:14) is shown in Table 9.
- the variable region is underlined (SEQ ID NO: 53).
- Figure 22 shows the effect of intravenous administration of an effective dose (3 mg/kg) of antibody 5A10 (solid circles) or antibody L1 L3 (solid squares) to each of four cynomolgus monkeys at day zero.
- the change in serum HDL ( Figure 22A) and serum LDL ( Figure 22B) was measured from -2 to +28 days. Both antibodies resulted in greater than about 70% decrease in serum LDL levels by about seven days, an effect that substantially persisted for about six more days in the animals administered L1 L3. All the animals showed normal liver and kidney function and near-normal hematocrits.
- L1 L3 dose-dependently reduced LDL-C with a maximum effect observed in the 10 mg/kg group, which maintained a 70% reduction in LDL-C levels until day 21 post- dosing, and fully recovered by day 31.
- HDL-C levels were not affected by L1 L3 treatment in all dose groups.
- L1 L3 in non-human primates with hypercholesterolemia and pharmacodynamic interactions between L1 L3 and HMG-CoA reductase inhibiting statins were investigated.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Epidemiology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Endocrinology (AREA)
- Heart & Thoracic Surgery (AREA)
- Diabetes (AREA)
- Obesity (AREA)
- Cardiology (AREA)
- Hematology (AREA)
- Urology & Nephrology (AREA)
- Vascular Medicine (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicinal Preparation (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
Claims
Priority Applications (15)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN200980145121.2A CN102333542B (en) | 2008-09-12 | 2009-09-11 | PCSK9 antagonist |
CA2736349A CA2736349C (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists |
JP2011526617A JP4898976B2 (en) | 2008-09-12 | 2009-09-11 | PCSK9 antagonist |
NZ591541A NZ591541A (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists |
BRPI0919289-1A BRPI0919289A2 (en) | 2008-09-12 | 2009-09-11 | pcsk9 antagonist. |
KR1020117008305A KR101230675B1 (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists |
MX2011002726A MX2011002726A (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists. |
EP09808999A EP2344194A2 (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists |
RU2011109178/10A RU2528735C2 (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists |
AU2009290438A AU2009290438B2 (en) | 2008-09-12 | 2009-09-11 | PCSK9 antagonists |
IL211519A IL211519A (en) | 2008-09-12 | 2011-03-02 | Pcsk9 antagonists |
ZA2011/01688A ZA201101688B (en) | 2008-09-12 | 2011-03-04 | Pcsk9 antagonists |
HK12102005.5A HK1161136A1 (en) | 2008-09-12 | 2012-02-28 | Pcsk9 antagonists pcsk9 |
IL229059A IL229059A (en) | 2008-09-12 | 2013-10-24 | Pcsk9 antagonists |
PH12014501261A PH12014501261A1 (en) | 2008-09-12 | 2014-06-04 | Pcsk9 antagonists |
Applications Claiming Priority (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US9671608P | 2008-09-12 | 2008-09-12 | |
US61/096,716 | 2008-09-12 | ||
US23216109P | 2009-08-07 | 2009-08-07 | |
US61/232,161 | 2009-08-07 | ||
US23564309P | 2009-08-20 | 2009-08-20 | |
US61/235,643 | 2009-08-20 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2010029513A2 true WO2010029513A2 (en) | 2010-03-18 |
WO2010029513A3 WO2010029513A3 (en) | 2010-07-15 |
Family
ID=42005569
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2009/053990 WO2010029513A2 (en) | 2008-09-12 | 2009-09-11 | Pcsk9 antagonists |
Country Status (20)
Country | Link |
---|---|
US (5) | US8080243B2 (en) |
EP (1) | EP2344194A2 (en) |
JP (4) | JP4898976B2 (en) |
KR (1) | KR101230675B1 (en) |
CN (1) | CN102333542B (en) |
AR (1) | AR073292A1 (en) |
AU (1) | AU2009290438B2 (en) |
BR (1) | BRPI0919289A2 (en) |
CA (1) | CA2736349C (en) |
CO (1) | CO6351752A2 (en) |
HK (1) | HK1161136A1 (en) |
IL (2) | IL211519A (en) |
MX (1) | MX2011002726A (en) |
NZ (1) | NZ591541A (en) |
PE (2) | PE20151952A1 (en) |
PH (1) | PH12014501261A1 (en) |
RU (2) | RU2528735C2 (en) |
TW (2) | TWI445716B (en) |
WO (1) | WO2010029513A2 (en) |
ZA (1) | ZA201101688B (en) |
Cited By (139)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20110129478A1 (en) * | 2008-07-10 | 2011-06-02 | Toray Industries, Inc. | Pharmaceutical composition for treatment and prevention of cancer |
US8030457B2 (en) | 2007-08-23 | 2011-10-04 | Amgen, Inc. | Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9) |
GB2481373A (en) * | 2010-06-21 | 2011-12-28 | Weiming Xu | Treatment of hypercholesterolaemia by ubiquitination of PCSK9 |
US8188234B2 (en) | 2008-02-07 | 2012-05-29 | Merck Sharp & Dohme Corp. | 1D05 PCSK9 antagonists |
US8188233B2 (en) | 2008-02-07 | 2012-05-29 | Merck Sharp & Dohme Corp. | 1B20 PCSK9 antagonists |
WO2012088313A1 (en) * | 2010-12-22 | 2012-06-28 | Genentech, Inc. | Anti-pcsk9 antibodies and methods of use |
EP2493507A2 (en) * | 2009-10-30 | 2012-09-05 | Merck Sharp & Dohme Corp. | Ax213 and ax132 pcsk9 antagonists and variants |
WO2012101252A3 (en) * | 2011-01-28 | 2012-11-15 | Sanofi | Human antibodies to pcsk9 for use in methods of treating particular groups of subjects |
US8344114B2 (en) | 2006-11-07 | 2013-01-01 | Merck Sharp & Dohme Corp. | Antagonists of PCSK9 |
WO2013008185A1 (en) | 2011-07-14 | 2013-01-17 | Pfizer Inc. | Treatment with anti-pcsk9 antibodies |
WO2013039958A1 (en) | 2011-09-16 | 2013-03-21 | Eli Lilly And Company | Antibodies to pcsk9 and uses thereof |
WO2013086443A1 (en) | 2011-12-08 | 2013-06-13 | Amgen Inc. | Agonistic human lcat antigen binding proteins and their use in therapy |
WO2013091103A1 (en) * | 2011-12-20 | 2013-06-27 | Adaerata, Limited Partnership | Single domain antibodies as inhibitors of pcsk9 |
WO2013188855A1 (en) * | 2012-06-15 | 2013-12-19 | Genentech, Inc. | Anti-pcsk9 antibodies, formulations, dosing, and methods of use |
WO2014081780A1 (en) | 2012-11-21 | 2014-05-30 | Amgen Inc. | Drug delivery device |
WO2014143770A1 (en) | 2013-03-15 | 2014-09-18 | Amgen Inc. | Body contour adaptable autoinjector device |
WO2014144096A1 (en) | 2013-03-15 | 2014-09-18 | Amgen Inc. | Drug cassette, autoinjector, and autoinjector system |
WO2014150983A2 (en) * | 2013-03-15 | 2014-09-25 | Amgen Inc. | Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9 |
WO2014149699A1 (en) | 2013-03-15 | 2014-09-25 | Eli Lilly And Company | Bifunctional protein |
WO2014149357A1 (en) | 2013-03-22 | 2014-09-25 | Amgen Inc. | Injector and method of assembly |
JP2014527967A (en) * | 2011-09-16 | 2014-10-23 | リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. | Method of reducing lipoprotein (a) levels by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9) |
US8883157B1 (en) | 2013-12-17 | 2014-11-11 | Kymab Limited | Targeting rare human PCSK9 variants for cholesterol treatment |
WO2014197752A1 (en) | 2013-06-07 | 2014-12-11 | Regeneron Pharmaceuticals, Inc. | Methods fo inhibting atherosclerosis by administering an inhibitor of pcsk9 |
KR20150006471A (en) * | 2012-05-08 | 2015-01-16 | 앨더바이오 홀딩스 엘엘씨 | Anti-pcsk9 antibodies and use thereof |
US8980273B1 (en) | 2014-07-15 | 2015-03-17 | 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 |
US8986691B1 (en) | 2014-07-15 | 2015-03-24 | Kymab Limited | Method of treating atopic dermatitis or asthma using antibody to IL4RA |
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 |
EP2862877A1 (en) | 2013-10-18 | 2015-04-22 | Sanofi | Methods for inhibiting atherosclerosis by administering an inhibitor of PCSK9 |
US9017678B1 (en) | 2014-07-15 | 2015-04-28 | Kymab Limited | Method of treating rheumatoid arthritis using antibody to IL6R |
WO2015061389A1 (en) | 2013-10-24 | 2015-04-30 | Amgen Inc. | Drug delivery system with temperature-sensitive control |
WO2015061386A1 (en) | 2013-10-24 | 2015-04-30 | Amgen Inc. | Injector and method of assembly |
US9034332B1 (en) | 2014-07-15 | 2015-05-19 | Kymab Limited | Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment |
US9045548B1 (en) | 2014-07-15 | 2015-06-02 | 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 |
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 |
US9067998B1 (en) | 2014-07-15 | 2015-06-30 | Kymab Limited | Targeting PD-1 variants for treatment of cancer |
WO2015119906A1 (en) | 2014-02-05 | 2015-08-13 | Amgen Inc. | Drug delivery system with electromagnetic field generator |
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 |
DE202014010499U1 (en) | 2013-12-17 | 2015-10-20 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
WO2015171777A1 (en) | 2014-05-07 | 2015-11-12 | Amgen Inc. | Autoinjector with shock reducing elements |
WO2015187799A1 (en) | 2014-06-03 | 2015-12-10 | Amgen Inc. | Systems and methods for remotely processing data collected by a drug delivery device |
EP2975059A1 (en) | 2014-07-15 | 2016-01-20 | Kymab Limited | Antibodies for use in treating conditions related to specific pcsk9 variants in specific patients populations |
WO2016008899A1 (en) | 2014-07-15 | 2016-01-21 | Kymab Limited | Targeting human pcsk9 for cholesterol treatment |
WO2016020799A1 (en) | 2014-08-06 | 2016-02-11 | Rinat Neuroscience Corp. | Methods for reducing ldl-cholesterol |
WO2016020798A1 (en) | 2014-08-06 | 2016-02-11 | Rinat Neuroscience Corp. | Methods for reducing ldl-cholesterol |
WO2016023916A1 (en) | 2014-08-12 | 2016-02-18 | Kymab Limited | Treatment of disease using ligand binding to targets of interest |
WO2016046684A1 (en) | 2014-09-23 | 2016-03-31 | Pfizer Inc. | Treatment with anti-pcsk9 antibodies |
WO2016061220A2 (en) | 2014-10-14 | 2016-04-21 | Amgen Inc. | Drug injection device with visual and audio indicators |
WO2016071701A1 (en) | 2014-11-07 | 2016-05-12 | Kymab Limited | Treatment of disease using ligand binding to targets of interest |
WO2016085820A1 (en) * | 2014-11-24 | 2016-06-02 | Shire Human Genetic Therapies, Inc. | Lysosomal targeting and uses thereof |
WO2016100055A1 (en) | 2014-12-19 | 2016-06-23 | Amgen Inc. | Drug delivery device with live button or user interface field |
WO2016100781A1 (en) | 2014-12-19 | 2016-06-23 | Amgen Inc. | Drug delivery device with proximity sensor |
DE202015009002U1 (en) | 2014-07-15 | 2016-08-18 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
WO2016133947A1 (en) | 2015-02-17 | 2016-08-25 | Amgen Inc. | Drug delivery device with vacuum assisted securement and/or feedback |
WO2016138434A1 (en) | 2015-02-27 | 2016-09-01 | Amgen Inc. | Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement |
US9540449B2 (en) | 2012-08-13 | 2017-01-10 | Regeneron Pharmaceuticals, Inc. | Anti-PCSK9 antibodies with pH-dependent binding characteristics |
US9550837B2 (en) | 2008-12-15 | 2017-01-24 | Regeneron Pharmaceuticals, Inc. | Therapeutic uses of anti-PCSK9 antibodies |
WO2017039786A1 (en) | 2015-09-02 | 2017-03-09 | Amgen Inc. | Syringe assembly adapter for a syringe |
WO2017055966A1 (en) | 2015-10-01 | 2017-04-06 | Pfizer Inc. | Low viscosity antibody compositions |
WO2017100501A1 (en) | 2015-12-09 | 2017-06-15 | Amgen Inc. | Auto-injector with signaling cap |
WO2017120178A1 (en) | 2016-01-06 | 2017-07-13 | Amgen Inc. | Auto-injector with signaling electronics |
US9724411B2 (en) | 2008-12-15 | 2017-08-08 | Regeneron Pharmaceuticals, Inc. | Methods for treating hypercholesterolemia and reducing LDL-C using antibodies to PCSK9 |
WO2017160799A1 (en) | 2016-03-15 | 2017-09-21 | Amgen Inc. | Reducing probability of glass breakage in drug delivery devices |
WO2017189089A1 (en) | 2016-04-29 | 2017-11-02 | Amgen Inc. | Drug delivery device with messaging label |
WO2017192287A1 (en) | 2016-05-02 | 2017-11-09 | Amgen Inc. | Syringe adapter and guide for filling an on-body injector |
WO2017197222A1 (en) | 2016-05-13 | 2017-11-16 | Amgen Inc. | Vial sleeve assembly |
WO2017200989A1 (en) | 2016-05-16 | 2017-11-23 | Amgen Inc. | Data encryption in medical devices with limited computational capability |
WO2017209899A1 (en) | 2016-06-03 | 2017-12-07 | Amgen Inc. | Impact testing apparatuses and methods for drug delivery devices |
WO2018004842A1 (en) | 2016-07-01 | 2018-01-04 | Amgen Inc. | Drug delivery device having minimized risk of component fracture upon impact events |
WO2018034784A1 (en) | 2016-08-17 | 2018-02-22 | Amgen Inc. | Drug delivery device with placement detection |
WO2018054240A1 (en) * | 2016-09-20 | 2018-03-29 | Wuxi Biologics (Shanghai) Co. Ltd. | Novel anti-pcsk9 antibodies |
WO2018081234A1 (en) | 2016-10-25 | 2018-05-03 | Amgen Inc. | On-body injector |
WO2018113781A1 (en) | 2016-12-24 | 2018-06-28 | 信达生物制药(苏州)有限公司 | Anti-pcsk9 antibody and application thereof |
WO2018136398A1 (en) | 2017-01-17 | 2018-07-26 | Amgen Inc. | Injection devices and related methods of use and assembly |
WO2018151890A1 (en) | 2017-02-17 | 2018-08-23 | Amgen Inc. | Drug delivery device with sterile fluid flowpath and related method of assembly |
WO2018152073A1 (en) | 2017-02-17 | 2018-08-23 | Amgen Inc. | Insertion mechanism for drug delivery device |
WO2018164829A1 (en) | 2017-03-07 | 2018-09-13 | Amgen Inc. | Needle insertion by overpressure |
WO2018165143A1 (en) | 2017-03-06 | 2018-09-13 | Amgen Inc. | Drug delivery device with activation prevention feature |
WO2018165499A1 (en) | 2017-03-09 | 2018-09-13 | Amgen Inc. | Insertion mechanism for drug delivery device |
WO2018183039A1 (en) | 2017-03-28 | 2018-10-04 | Amgen Inc. | Plunger rod and syringe assembly system and method |
US10111953B2 (en) | 2013-05-30 | 2018-10-30 | Regeneron Pharmaceuticals, Inc. | Methods for reducing remnant cholesterol and other lipoprotein fractions by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9) |
WO2018226515A1 (en) | 2017-06-08 | 2018-12-13 | Amgen Inc. | Syringe assembly for a drug delivery device and method of assembly |
WO2018226565A1 (en) | 2017-06-08 | 2018-12-13 | Amgen Inc. | Torque driven drug delivery device |
WO2018236619A1 (en) | 2017-06-22 | 2018-12-27 | Amgen Inc. | Device activation impact/shock reduction |
WO2018237225A1 (en) | 2017-06-23 | 2018-12-27 | Amgen Inc. | Electronic drug delivery device comprising a cap activated by a switch assembly |
EP3424949A1 (en) * | 2010-04-13 | 2019-01-09 | Bristol-Myers Squibb Company | Fibronectin based scaffold domain proteins that bind pcsk9 |
WO2019014014A1 (en) | 2017-07-14 | 2019-01-17 | Amgen Inc. | Needle insertion-retraction system having dual torsion spring system |
WO2019018169A1 (en) | 2017-07-21 | 2019-01-24 | Amgen Inc. | Gas permeable sealing member for drug container and methods of assembly |
WO2019022951A1 (en) | 2017-07-25 | 2019-01-31 | Amgen Inc. | Drug delivery device with gear module and related method of assembly |
WO2019022950A1 (en) | 2017-07-25 | 2019-01-31 | Amgen Inc. | Drug delivery device with container access system and related method of assembly |
WO2019032482A2 (en) | 2017-08-09 | 2019-02-14 | Amgen Inc. | Hydraulic-pneumatic pressurized chamber drug delivery system |
WO2019036181A1 (en) | 2017-08-18 | 2019-02-21 | Amgen Inc. | Wearable injector with sterile adhesive patch |
WO2019040548A1 (en) | 2017-08-22 | 2019-02-28 | Amgen Inc. | Needle insertion mechanism for drug delivery device |
WO2019070472A1 (en) | 2017-10-04 | 2019-04-11 | Amgen Inc. | Flow adapter for drug delivery device |
WO2019070552A1 (en) | 2017-10-06 | 2019-04-11 | Amgen Inc. | Drug delivery device with interlock assembly and related method of assembly |
WO2019074579A1 (en) | 2017-10-09 | 2019-04-18 | Amgen Inc. | Drug delivery device with drive assembly and related method of assembly |
WO2019090303A1 (en) | 2017-11-06 | 2019-05-09 | Amgen Inc. | Fill-finish assemblies and related methods |
WO2019090079A1 (en) | 2017-11-03 | 2019-05-09 | Amgen Inc. | System and approaches for sterilizing a drug delivery device |
WO2019089178A1 (en) | 2017-11-06 | 2019-05-09 | Amgen Inc. | Drug delivery device with placement and flow sensing |
WO2019094138A1 (en) | 2017-11-10 | 2019-05-16 | Amgen Inc. | Plungers for drug delivery devices |
WO2019099324A1 (en) | 2017-11-16 | 2019-05-23 | Amgen Inc. | Door latch mechanism for drug delivery device |
WO2019099322A1 (en) | 2017-11-16 | 2019-05-23 | Amgen Inc. | Autoinjector with stall and end point detection |
EP3398968A4 (en) * | 2015-12-31 | 2019-08-07 | Jiangsu Hengrui Medicine Co., Ltd. | Pcsk9 antibody, antigen-binding fragment thereof, and medicinal application thereof |
US10428157B2 (en) | 2013-11-12 | 2019-10-01 | Sanofi Biotechnology | Dosing regimens for use with PCSK9 inhibitors |
US10472425B2 (en) | 2011-07-28 | 2019-11-12 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-PCSK9 antibodies |
WO2019231582A1 (en) | 2018-05-30 | 2019-12-05 | Amgen Inc. | Thermal spring release mechanism for a drug delivery device |
WO2019231618A1 (en) | 2018-06-01 | 2019-12-05 | Amgen Inc. | Modular fluid path assemblies for drug delivery devices |
EP3593839A1 (en) | 2013-03-15 | 2020-01-15 | Amgen Inc. | Drug cassette |
EP3401336A4 (en) * | 2016-01-05 | 2020-01-22 | Jiangsu Hengrui Medicine Co., Ltd. | Pcsk9 antibody, antigen-binding fragment thereof, and medical uses thereof |
US10544232B2 (en) | 2014-07-16 | 2020-01-28 | Sanofi Biotechnology | Methods for treating patients with heterozygous familial hypercholesterolemia (heFH) with an anti-PCSK9 antibody |
WO2020023444A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Delivery devices for administering drugs |
WO2020023220A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation |
WO2020023336A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Hybrid drug delivery devices with grip portion |
WO2020023451A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Delivery devices for administering drugs |
WO2020028009A1 (en) | 2018-07-31 | 2020-02-06 | Amgen Inc. | Fluid path assembly for a drug delivery device |
WO2020068476A1 (en) | 2018-09-28 | 2020-04-02 | Amgen Inc. | Muscle wire escapement activation assembly for a drug delivery device |
WO2020068623A1 (en) | 2018-09-24 | 2020-04-02 | Amgen Inc. | Interventional dosing systems and methods |
WO2020072577A1 (en) | 2018-10-02 | 2020-04-09 | Amgen Inc. | Injection systems for drug delivery with internal force transmission |
WO2020072846A1 (en) | 2018-10-05 | 2020-04-09 | Amgen Inc. | Drug delivery device having dose indicator |
WO2020081479A1 (en) | 2018-10-15 | 2020-04-23 | Amgen Inc. | Drug delivery device having damping mechanism |
WO2020081480A1 (en) | 2018-10-15 | 2020-04-23 | Amgen Inc. | Platform assembly process for drug delivery device |
WO2020091981A1 (en) | 2018-11-01 | 2020-05-07 | Amgen Inc. | Drug delivery devices with partial drug delivery member retraction |
WO2020092056A1 (en) | 2018-11-01 | 2020-05-07 | Amgen Inc. | Drug delivery devices with partial needle retraction |
WO2020091956A1 (en) | 2018-11-01 | 2020-05-07 | Amgen Inc. | Drug delivery devices with partial drug delivery member retraction |
US10688119B2 (en) | 2015-03-20 | 2020-06-23 | Aarhus Universitet | Inhibitors of PCSK9 for treatment of lipoprotein metabolism disorders |
US10772956B2 (en) | 2015-08-18 | 2020-09-15 | Regeneron Pharmaceuticals, Inc. | Methods for reducing or eliminating the need for lipoprotein apheresis in patients with hyperlipidemia by administering alirocumab |
EP3515950A4 (en) * | 2016-09-20 | 2020-10-28 | Wuxi Biologics Ireland Limited. | Novel anti-pcsk9 antibodies |
WO2020219482A1 (en) | 2019-04-24 | 2020-10-29 | Amgen Inc. | Syringe sterilization verification assemblies and methods |
US10822385B2 (en) | 2016-01-13 | 2020-11-03 | Novo Nordisk A/S | EGF(A) analogues with fatty acid substituents |
WO2021041067A2 (en) | 2019-08-23 | 2021-03-04 | Amgen Inc. | Drug delivery device with configurable needle shield engagement components and related methods |
US11130794B2 (en) | 2017-07-19 | 2021-09-28 | Novo Nordisk A/S | Bifunctional compounds |
US11173177B2 (en) | 2016-09-20 | 2021-11-16 | Aarhus Universitet | Compounds for treatment of lipoprotein metabolism disorders |
WO2022246055A1 (en) | 2021-05-21 | 2022-11-24 | Amgen Inc. | Method of optimizing a filling recipe for a drug container |
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 |
Families Citing this family (53)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US8357371B2 (en) | 2008-12-15 | 2013-01-22 | Regeneron Pharmaceuticals, Inc. | Methods for treating hypercholesterolemia using antibodies to PCSK9 |
EP2473605B1 (en) | 2009-09-03 | 2018-04-11 | Pfizer Vaccines LLC | Pcsk9 vaccine |
AR079336A1 (en) * | 2009-12-11 | 2012-01-18 | Irm Llc | ANTAGONISTS OF THE PRO-PROTEIN CONVERTASE-SUBTILISINE / TYPE 9 QUEXINE (PCSK9) |
BR112012028764A2 (en) * | 2010-05-11 | 2017-03-14 | Aveo Pharmaceuticals Inc | anti-pos antifgfr2 |
JO3756B1 (en) | 2010-11-23 | 2021-01-31 | Regeneron Pharma | Human antibodies to the glucagon receptor |
RU2644684C2 (en) | 2012-03-16 | 2018-02-13 | Регенерон Фармасьютикалз, Инк. | Antibodies with built in the light circuit by hystidine and genetically modified excellent from human animals for their obtaining |
MX2014011047A (en) | 2012-03-16 | 2015-04-08 | Regeneron Pharma | Mice that produce antigen-binding proteins with ph-dependent binding characteristics. |
US20140013456A1 (en) | 2012-03-16 | 2014-01-09 | Regeneron Pharmaceuticals, Inc. | Histidine Engineered Light Chain Antibodies and Genetically Modified Non-Human Animals for Generating the Same |
BR112014022855A2 (en) | 2012-03-16 | 2017-07-18 | Regeneron Pharma | genetically modified nonhuman animal, genetically modified mammal, and method for making a nonhuman animal |
EP2810955A1 (en) | 2013-06-07 | 2014-12-10 | Sanofi | Methods for inhibiting atherosclerosis by administering an inhibitor of PCSK9 |
EP3013422A1 (en) | 2013-06-28 | 2016-05-04 | Amgen Inc. | Methods for treating homozygous familial hypercholesterolemia |
US10035847B2 (en) | 2013-10-02 | 2018-07-31 | The Rockefeller University | Amyloid protofibril antibodies and methods of use thereof |
US20170173128A1 (en) * | 2013-12-06 | 2017-06-22 | Moderna TX, Inc. | Targeted adaptive vaccines |
US8945560B1 (en) | 2014-07-15 | 2015-02-03 | Kymab Limited | Method of treating rheumatoid arthritis using antibody to IL6R |
AU2015317899A1 (en) | 2014-09-16 | 2017-04-06 | Regeneron Pharmaceuticals, Inc. | Anti-glucagon antibodies and uses thereof |
EP3209332B1 (en) | 2014-10-23 | 2021-05-26 | Amgen Inc. | Reducing viscosity of pharmaceutical formulations |
CN105461809B (en) * | 2015-02-11 | 2018-10-12 | 康融东方(广东)医药有限公司 | PCSK9 antibody, its medical composition and its use |
CN105037554B (en) * | 2015-06-12 | 2019-04-12 | 成都贝爱特生物科技有限公司 | The preparation and application thereof of anti-human PCSK9 antibody |
CN106810609A (en) | 2015-11-27 | 2017-06-09 | 苏州君盟生物医药科技有限公司 | Anti- PCSK9 antibody and its application |
CN107266575B (en) * | 2016-04-07 | 2021-12-24 | 天士力生物医药股份有限公司 | Binding protein of proprotein convertase subtilisin kexin type 9 and application thereof |
US11016085B2 (en) | 2016-04-25 | 2021-05-25 | The Johns Hopkins University | ZNT8 assays for drug development and pharmaceutical compositions |
WO2017205377A2 (en) * | 2016-05-23 | 2017-11-30 | New York University | Compositions and methods for antibodies targeting staphylococcal leukotoxins |
CN107474140B (en) * | 2016-06-08 | 2022-06-03 | 常州博嘉生物医药科技有限公司 | PCSK9 specific binding protein MV072 and application thereof |
MX2018016057A (en) * | 2016-06-24 | 2019-05-06 | Hoffmann La Roche | Compositions and methods for treating cardiovascular disease. |
EP3522918A1 (en) | 2016-10-06 | 2019-08-14 | Amgen Inc. | Reduced viscosity protein pharmaceutical formulations |
US20190248888A1 (en) | 2016-10-20 | 2019-08-15 | Regeneron Pharmaceuticals, Inc. | Methods of lowering blood glucose levels |
JOP20190112A1 (en) | 2016-11-14 | 2019-05-14 | Amgen Inc | Combined therapies for atherosclerosis, including atherosclerotic cardiovascular disease |
JP7377596B2 (en) * | 2017-02-22 | 2023-11-10 | アムジエン・インコーポレーテツド | Low viscosity, high concentration evolocumab formulations and their manufacturing method |
AU2018258676B2 (en) | 2017-04-28 | 2024-09-19 | Amgen Inc. | N-acetylated and non-acetylated dipeptides containing arginine to reduce the viscosity of viscous compositions of therapeutic polypeptides |
US11892457B2 (en) | 2017-07-12 | 2024-02-06 | The Johns Hopkins University | Proteoliposome-based ZnT8 self-antigen for type 1 diabetes diagnosis |
US11525004B2 (en) | 2017-07-20 | 2022-12-13 | H. Lee Moffitt Cancer Center And Research Institute, Inc. | Recombinant CD123-binding antibodies |
ES2978396T3 (en) | 2017-08-01 | 2024-09-11 | Amgen Inc | Systems and methods for real-time preparation of a polypeptide sample for analysis by mass spectrometry |
CN110892267B (en) | 2017-08-01 | 2024-08-09 | 安进公司 | System and method for real-time glycan determination of a sample |
WO2019152599A1 (en) * | 2018-01-31 | 2019-08-08 | The Wistar Institute Of Anatomy And Biology | Nucleic acid monoclonal antibodies targeting pcsk9 and methods of use |
JP2021518748A (en) | 2018-02-23 | 2021-08-05 | アールイーエムディー バイオセラピューティクス,インコーポレイテッドREMD Biotherapeutics,Inc | Calcitonin gene-related peptide (CGRP) antagonist antibody |
SG11202008869UA (en) | 2018-03-13 | 2020-10-29 | Amgen Inc | Sequential digestion of polypeptides for mass spectrometric analysis |
JP2021516672A (en) | 2018-03-13 | 2021-07-08 | アムジエン・インコーポレーテツド | Methods for Preparing Trypsin-Resistant Polypeptides for Mass Spectroscopy Analysis |
CN113166241A (en) | 2018-08-16 | 2021-07-23 | 约翰霍普金斯大学 | Human ZNT8 antibodies |
US12000775B2 (en) | 2018-12-14 | 2024-06-04 | Amgen Inc. | System suitability method for use with protein concentration determination by slope |
US20220137061A1 (en) | 2019-02-14 | 2022-05-05 | Amgen Inc. | Systems And Methods For Preparing A Sample and Performing A Real-Time Assay Of The Sample |
JP7530376B2 (en) | 2019-02-20 | 2024-08-07 | アムジエン・インコーポレーテツド | Methods for determining protein stability |
US20230035363A1 (en) | 2019-03-04 | 2023-02-02 | Amgen Inc. | In vivo reversibility of high molecular weight species |
US12078701B2 (en) | 2019-03-27 | 2024-09-03 | Amgen Inc. | Methods of fingerprinting therapeutic proteins via a two-dimensional (2D) nuclear magnetic resonance technique at natural abundance for formulated biopharmaceutical products |
MA56121A (en) | 2019-06-05 | 2022-04-13 | Amgen Inc | METHODS FOR IDENTIFYING THERAPEUTIC PROTEIN ATTRIBUTES |
WO2021058597A1 (en) | 2019-09-24 | 2021-04-01 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Methods of determining whether a subject is at risk of developing arterial plaques |
AU2021253959A1 (en) * | 2020-04-09 | 2022-11-17 | Verve Therapeutics, Inc. | Base editing of PCSK9 and methods of using same for treatment of disease |
KR20210158693A (en) * | 2020-06-24 | 2021-12-31 | 바이오스트림테크놀러지스(주) | Anti pcsk9 antibody and use thereof |
US20230349912A1 (en) | 2020-09-18 | 2023-11-02 | Amgen Inc. | Methods of processing a sample for peptide mapping analysis |
AU2021376246A1 (en) | 2020-11-05 | 2023-06-08 | Amgen Inc. | Materials and methods for protein processing |
EP4387666A1 (en) * | 2021-08-20 | 2024-06-26 | The Johns Hopkins University | Cell-surface antibody to a specific biomarker of pancreatic beta-cells |
WO2023070103A1 (en) | 2021-10-21 | 2023-04-27 | Flagship Pioneering Innovations Vi, Llc | Modulators of proprotein convertase subtilisin/kexin type 9 (pcsk9) |
WO2023086793A1 (en) | 2021-11-09 | 2023-05-19 | Amgen Inc. | Production of therapeutic proteins |
WO2024040020A1 (en) | 2022-08-15 | 2024-02-22 | Absci Corporation | Quantitative affinity activity specific cell enrichment |
Citations (78)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US3773919A (en) | 1969-10-23 | 1973-11-20 | Du Pont | Polylactide-drug mixtures |
EP0003089A1 (en) | 1978-01-06 | 1979-07-25 | Bernard David | Drier for silkscreen printed sheets |
US4485045A (en) | 1981-07-06 | 1984-11-27 | Research Corporation | Synthetic phosphatidyl cholines useful in forming liposomes |
US4544545A (en) | 1983-06-20 | 1985-10-01 | Trustees University Of Massachusetts | Liposomes containing modified cholesterol for organ targeting |
US4676980A (en) | 1985-09-23 | 1987-06-30 | The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services | Target specific cross-linked heteroantibodies |
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4683195A (en) | 1986-01-30 | 1987-07-28 | Cetus Corporation | Process for amplifying, detecting, and/or-cloning nucleic acid sequences |
WO1987004462A1 (en) | 1986-01-23 | 1987-07-30 | Celltech Limited | Recombinant dna sequences, vectors containing them and method for the use thereof |
US4754065A (en) | 1984-12-18 | 1988-06-28 | Cetus Corporation | Precursor to nucleic acid probe |
GB2200651A (en) | 1987-02-07 | 1988-08-10 | Al Sumidaie Ayad Mohamed Khala | A method of obtaining a retrovirus-containing fraction from retrovirus-containing cells |
US4777127A (en) | 1985-09-30 | 1988-10-11 | Labsystems Oy | Human retrovirus-related products and methods of diagnosing and treating conditions associated with said retrovirus |
US4800159A (en) | 1986-02-07 | 1989-01-24 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences |
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
EP0345242A2 (en) | 1988-06-03 | 1989-12-06 | Smithkline Biologicals S.A. | Expression of gag proteins from retroviruses in eucaryotic cells |
WO1990007936A1 (en) | 1989-01-23 | 1990-07-26 | Chiron Corporation | Recombinant therapies for infection and hyperproliferative disorders |
WO1990011092A1 (en) | 1989-03-21 | 1990-10-04 | Vical, Inc. | Expression of exogenous polynucleotide sequences in a vertebrate |
WO1991000360A1 (en) | 1989-06-29 | 1991-01-10 | Medarex, Inc. | Bispecific reagents for aids therapy |
WO1991002805A2 (en) | 1989-08-18 | 1991-03-07 | Viagene, Inc. | Recombinant retroviruses delivering vector constructs to target cells |
US5013556A (en) | 1989-10-20 | 1991-05-07 | Liposome Technology, Inc. | Liposomes with enhanced circulation time |
US5047335A (en) | 1988-12-21 | 1991-09-10 | The Regents Of The University Of Calif. | Process for controlling intracellular glycosylation of proteins |
WO1991014445A1 (en) | 1990-03-21 | 1991-10-03 | Research Development Foundation | Heterovesicular liposomes |
WO1992020373A1 (en) | 1991-05-14 | 1992-11-26 | Repligen Corporation | Heteroconjugate antibodies for treatment of hiv infection |
EP0519596A1 (en) | 1991-05-17 | 1992-12-23 | Merck & Co. Inc. | A method for reducing the immunogenicity of antibody variable domains |
WO1993003769A1 (en) | 1991-08-20 | 1993-03-04 | THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES | Adenovirus mediated transfer of genes to the gastrointestinal tract |
WO1993006213A1 (en) | 1991-09-23 | 1993-04-01 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
WO1993010218A1 (en) | 1991-11-14 | 1993-05-27 | The United States Government As Represented By The Secretary Of The Department Of Health And Human Services | Vectors including foreign genes and negative selective markers |
WO1993011230A1 (en) | 1991-12-02 | 1993-06-10 | Dynal As | Modified mammalian stem cell blocking viral replication |
US5219740A (en) | 1987-02-13 | 1993-06-15 | Fred Hutchinson Cancer Research Center | Retroviral gene transfer into diploid fibroblasts for gene therapy |
WO1993019191A1 (en) | 1992-03-16 | 1993-09-30 | Centre National De La Recherche Scientifique | Defective recombinant adenoviruses expressing cytokines for use in antitumoral treatment |
WO1993025234A1 (en) | 1992-06-08 | 1993-12-23 | The Regents Of The University Of California | Methods and compositions for targeting specific tissue |
WO1993025698A1 (en) | 1992-06-10 | 1993-12-23 | The United States Government As Represented By The | Vector particles resistant to inactivation by human serum |
US5278299A (en) | 1991-03-18 | 1994-01-11 | Scripps Clinic And Research Foundation | Method and composition for synthesizing sialylated glycosyl compounds |
WO1994003622A1 (en) | 1992-07-31 | 1994-02-17 | Imperial College Of Science, Technology & Medicine | D-type retroviral vectors, based on mpmv |
WO1994004690A1 (en) | 1992-08-17 | 1994-03-03 | Genentech, Inc. | Bispecific immunoadhesins |
WO1994012649A2 (en) | 1992-12-03 | 1994-06-09 | Genzyme Corporation | Gene therapy for cystic fibrosis |
WO1994023697A1 (en) | 1993-04-22 | 1994-10-27 | Depotech Corporation | Cyclodextrin liposomes encapsulating pharmacologic compounds and methods for their use |
WO1994028938A1 (en) | 1993-06-07 | 1994-12-22 | The Regents Of The University Of Michigan | Adenovirus vectors for gene therapy sponsorship |
WO1995000655A1 (en) | 1993-06-24 | 1995-01-05 | Mc Master University | Adenovirus vectors for gene therapy |
WO1995007994A2 (en) | 1993-09-15 | 1995-03-23 | Viagene, Inc. | Recombinant alphavirus vectors |
WO1995011984A2 (en) | 1993-10-25 | 1995-05-04 | Canji, Inc. | Recombinant adenoviral vector and methods of use |
WO1995013796A1 (en) | 1993-11-16 | 1995-05-26 | Depotech Corporation | Vesicles with controlled release of actives |
US5422120A (en) | 1988-05-30 | 1995-06-06 | Depotech Corporation | Heterovesicular liposomes |
WO1995030763A2 (en) | 1994-05-09 | 1995-11-16 | Chiron Viagene, Inc. | Retroviral vectors having a reduced recombination rate |
US5510261A (en) | 1991-11-21 | 1996-04-23 | The Board Of Trustees Of The Leland Stanford Juniot University | Method of controlling the degradation of glycoprotein oligosaccharides produced by cultured Chinese hamster ovary cells |
WO1996017072A2 (en) | 1994-11-30 | 1996-06-06 | Chiron Viagene, Inc. | Recombinant alphavirus vectors |
US5530101A (en) | 1988-12-28 | 1996-06-25 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5545806A (en) | 1990-08-29 | 1996-08-13 | Genpharm International, Inc. | Ransgenic non-human animals for producing heterologous antibodies |
US5545807A (en) | 1988-10-12 | 1996-08-13 | The Babraham Institute | Production of antibodies from transgenic animals |
US5565332A (en) | 1991-09-23 | 1996-10-15 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
US5569825A (en) | 1990-08-29 | 1996-10-29 | Genpharm International | Transgenic non-human animals capable of producing heterologous antibodies of various isotypes |
US5580859A (en) | 1989-03-21 | 1996-12-03 | Vical Incorporated | Delivery of exogenous DNA sequences in a mammal |
US5580717A (en) | 1990-05-01 | 1996-12-03 | Affymax Technologies N.V. | Recombinant library screening methods |
US5625126A (en) | 1990-08-29 | 1997-04-29 | Genpharm International, Inc. | Transgenic non-human animals for producing heterologous antibodies |
US5633425A (en) | 1990-08-29 | 1997-05-27 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
US5661016A (en) | 1990-08-29 | 1997-08-26 | Genpharm International Inc. | Transgenic non-human animals capable of producing heterologous antibodies of various isotypes |
WO1997042338A1 (en) | 1996-05-06 | 1997-11-13 | Chiron Corporation | Crossless retroviral vectors |
US5733743A (en) | 1992-03-24 | 1998-03-31 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
US5750373A (en) | 1990-12-03 | 1998-05-12 | Genentech, Inc. | Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants |
US5807715A (en) | 1984-08-27 | 1998-09-15 | The Board Of Trustees Of The Leland Stanford Junior University | Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin |
US5814482A (en) | 1993-09-15 | 1998-09-29 | Dubensky, Jr.; Thomas W. | Eukaryotic layered vector initiation systems |
US5866692A (en) | 1991-09-18 | 1999-02-02 | Kyowa Hakko Kogyo Co., Ltd. | Process for producing humanized chimera antibody |
US5981568A (en) | 1993-01-28 | 1999-11-09 | Neorx Corporation | Therapeutic inhibitor of vascular smooth muscle cells |
WO1999058572A1 (en) | 1998-05-08 | 1999-11-18 | Cambridge University Technical Services Limited | Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis |
US5997867A (en) | 1991-07-16 | 1999-12-07 | Waldmann; Herman | Method of using humanized antibody against CD18 |
WO2000009560A2 (en) | 1998-08-17 | 2000-02-24 | Abgenix, Inc. | Generation of modified molecules with increased serum half-lives |
US6054297A (en) | 1991-06-14 | 2000-04-25 | Genentech, Inc. | Humanized antibodies and methods for making them |
WO2000053211A2 (en) | 1999-03-09 | 2000-09-14 | University Of Southern California | Method of promoting myocyte proliferation and myocardial tissue repair |
US6180377B1 (en) | 1993-06-16 | 2001-01-30 | Celltech Therapeutics Limited | Humanized antibodies |
US6210671B1 (en) | 1992-12-01 | 2001-04-03 | Protein Design Labs, Inc. | Humanized antibodies reactive with L-selectin |
WO2001027160A1 (en) | 1999-10-14 | 2001-04-19 | Applied Molecular Evolution, Inc. | Methods of optimizing antibody variable region binding affinity |
US6265150B1 (en) | 1995-06-07 | 2001-07-24 | Becton Dickinson & Company | Phage antibodies |
US6350861B1 (en) | 1992-03-09 | 2002-02-26 | Protein Design Labs, Inc. | Antibodies with increased binding affinity |
US6376471B1 (en) | 1997-10-10 | 2002-04-23 | Johns Hopkins University | Gene delivery compositions and methods |
US6413942B1 (en) | 1989-03-21 | 2002-07-02 | Vical, Inc. | Methods of delivering a physiologically active polypeptide to a mammal |
US6436908B1 (en) | 1995-05-30 | 2002-08-20 | Duke University | Use of exogenous β-adrenergic receptor and β-adrenergic receptor kinase gene constructs to enhance myocardial function |
WO2004058184A2 (en) | 2002-12-24 | 2004-07-15 | Rinat Neuroscience Corp. | Anti-ngf antibodies and methods using same |
US7300754B2 (en) | 2003-04-25 | 2007-11-27 | Institut National De La Sante Et De La Recherche Medicale | Methods for detecting the presence of or predisposition to autosomal dominant hypercholesterolemia |
US7314622B2 (en) | 2005-04-15 | 2008-01-01 | Neogenix Oncology, Inc. | Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers |
Family Cites Families (21)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5824307A (en) | 1991-12-23 | 1998-10-20 | Medimmune, Inc. | Human-murine chimeric antibodies against respiratory syncytial virus |
DE69830492T2 (en) | 1997-02-12 | 2006-03-16 | Chugai Seiyaku K.K. | Antibodies as medicines against lymphocytic tumors (excluding myeloma) |
US6235311B1 (en) * | 1998-03-18 | 2001-05-22 | Bristol-Myers Squibb Company | Pharmaceutical composition containing a combination of a statin and aspirin and method |
AU2001241461A1 (en) * | 2000-02-07 | 2001-08-14 | Millennium Pharmaceuticals, Inc. | Narc-1, novel subtilase-like homologs |
CU23007A1 (en) | 2001-04-06 | 2004-12-17 | Ct De Inmunologia Molecular Ct De Inmunologia Mole | IMMUNOTHERAPEUTIC COMBINATIONS FOR THE TREATMENT IMMUNOTHERAPEUTIC COMBINATIONS FOR THE TREATMENT OF TUMORS THAT OVER-EXPRESS GANGLIOSIDES TO OF TUMORS THAT OVER-EXPRESS GANGLOSIDES |
AR047392A1 (en) * | 2002-10-22 | 2006-01-18 | Wyeth Corp | NEUTRALIZATION OF ANTIBODIES AGAINST GDF 8 AND ITS USE FOR SUCH PURPOSES |
AU2004210679A1 (en) * | 2003-02-10 | 2004-08-26 | Elan Pharmaceuticals, Inc. | Immunoglobulin formulation and method of preparation thereof |
WO2005023177A2 (en) * | 2003-05-21 | 2005-03-17 | Medarex, Inc. | Human monoclonal antibodies against bacillusanthracis protective antigen |
AR053026A1 (en) * | 2005-03-08 | 2007-04-18 | Pharmacia & Upjohn Co Llc | COMPOSITIONS OF ANTIBODIES ANTI FACTOR OF STIMULATION OF COLONIES OF MACROFAGOS (ANTI-M CSF) |
WO2006124269A2 (en) | 2005-05-16 | 2006-11-23 | Amgen Fremont Inc. | Human monoclonal antibodies that bind to very late antigen-1 for the treatment of inflammation and other disorders |
EP2639242A3 (en) * | 2006-03-06 | 2013-10-16 | MedImmune, Inc. | Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease |
CA2900402C (en) * | 2006-05-08 | 2018-01-16 | Adaerata, Limited Partnership | Pcsk9 polypeptide, ligand to said polypeptide, kits and methods using said ligands |
CA2668131A1 (en) | 2006-11-07 | 2008-05-15 | Merck & Co., Inc. | Antagonists of pcsk9 |
WO2008057459A2 (en) * | 2006-11-07 | 2008-05-15 | Merck & Co., Inc. | Antagonists of pcsk9 |
CA2667869A1 (en) * | 2006-11-07 | 2008-05-15 | Merck & Co., Inc. | Antagonists of pcsk9 |
US20100040610A1 (en) * | 2006-11-07 | 2010-02-18 | Ayesha Sitlani | Antagonists of pcsk9 |
EP2137218A2 (en) | 2007-04-13 | 2009-12-30 | Novartis Ag | Molecules and methods for modulating proprotein convertase subtilisin/kexin type 9 (pcsk9) |
JOP20080381B1 (en) | 2007-08-23 | 2023-03-28 | Amgen Inc | Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9) |
US20130072665A1 (en) | 2007-08-23 | 2013-03-21 | Simon Mark Jackson | Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9) |
EP2205639B1 (en) * | 2007-10-26 | 2015-12-23 | Merck Sharp & Dohme Corp. | Anti-pcsk9 and methods for treating lipid and cholesterol disorders |
AR070316A1 (en) | 2008-02-07 | 2010-03-31 | Merck & Co Inc | PCSK9 ANTAGONISTS (SUBTILISINE-KEXINA TYPE 9 PROPROTEIN) |
-
2009
- 2009-09-10 TW TW098130562A patent/TWI445716B/en not_active IP Right Cessation
- 2009-09-10 TW TW103116072A patent/TWI516501B/en not_active IP Right Cessation
- 2009-09-11 CN CN200980145121.2A patent/CN102333542B/en not_active Expired - Fee Related
- 2009-09-11 RU RU2011109178/10A patent/RU2528735C2/en active
- 2009-09-11 AR ARP090103509A patent/AR073292A1/en active IP Right Grant
- 2009-09-11 JP JP2011526617A patent/JP4898976B2/en not_active Expired - Fee Related
- 2009-09-11 PE PE2015002084A patent/PE20151952A1/en unknown
- 2009-09-11 KR KR1020117008305A patent/KR101230675B1/en active IP Right Grant
- 2009-09-11 US US12/558,312 patent/US8080243B2/en active Active
- 2009-09-11 NZ NZ591541A patent/NZ591541A/en not_active IP Right Cessation
- 2009-09-11 WO PCT/IB2009/053990 patent/WO2010029513A2/en active Application Filing
- 2009-09-11 CA CA2736349A patent/CA2736349C/en not_active Expired - Fee Related
- 2009-09-11 MX MX2011002726A patent/MX2011002726A/en active IP Right Grant
- 2009-09-11 BR BRPI0919289-1A patent/BRPI0919289A2/en not_active Application Discontinuation
- 2009-09-11 EP EP09808999A patent/EP2344194A2/en not_active Ceased
- 2009-09-11 AU AU2009290438A patent/AU2009290438B2/en not_active Ceased
- 2009-09-11 PE PE2011000618A patent/PE20110802A1/en active IP Right Grant
-
2011
- 2011-03-02 IL IL211519A patent/IL211519A/en active IP Right Grant
- 2011-03-04 ZA ZA2011/01688A patent/ZA201101688B/en unknown
- 2011-03-28 CO CO11037695A patent/CO6351752A2/en active IP Right Grant
- 2011-09-02 US US13/225,119 patent/US8399646B2/en active Active
- 2011-09-02 US US13/225,265 patent/US8426363B2/en active Active
- 2011-12-26 JP JP2011283073A patent/JP5119359B2/en not_active Expired - Fee Related
-
2012
- 2012-02-28 HK HK12102005.5A patent/HK1161136A1/en not_active IP Right Cessation
- 2012-10-22 JP JP2012232726A patent/JP5750421B2/en not_active Expired - Fee Related
-
2013
- 2013-04-04 US US13/857,063 patent/US9175093B2/en active Active
- 2013-10-24 IL IL229059A patent/IL229059A/en active IP Right Grant
-
2014
- 2014-06-04 PH PH12014501261A patent/PH12014501261A1/en unknown
- 2014-07-07 RU RU2014127686A patent/RU2618869C2/en active
-
2015
- 2015-05-18 JP JP2015100693A patent/JP6060212B2/en not_active Expired - Fee Related
- 2015-09-24 US US14/864,750 patent/US20160096898A1/en not_active Abandoned
Patent Citations (86)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US3773919A (en) | 1969-10-23 | 1973-11-20 | Du Pont | Polylactide-drug mixtures |
EP0003089A1 (en) | 1978-01-06 | 1979-07-25 | Bernard David | Drier for silkscreen printed sheets |
US4485045A (en) | 1981-07-06 | 1984-11-27 | Research Corporation | Synthetic phosphatidyl cholines useful in forming liposomes |
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
US6331415B1 (en) | 1983-04-08 | 2001-12-18 | Genentech, Inc. | Methods of producing immunoglobulins, vectors and transformed host cells for use therein |
US4544545A (en) | 1983-06-20 | 1985-10-01 | Trustees University Of Massachusetts | Liposomes containing modified cholesterol for organ targeting |
US5807715A (en) | 1984-08-27 | 1998-09-15 | The Board Of Trustees Of The Leland Stanford Junior University | Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin |
US4754065A (en) | 1984-12-18 | 1988-06-28 | Cetus Corporation | Precursor to nucleic acid probe |
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4683202B1 (en) | 1985-03-28 | 1990-11-27 | Cetus Corp | |
US4676980A (en) | 1985-09-23 | 1987-06-30 | The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services | Target specific cross-linked heteroantibodies |
US4777127A (en) | 1985-09-30 | 1988-10-11 | Labsystems Oy | Human retrovirus-related products and methods of diagnosing and treating conditions associated with said retrovirus |
WO1987004462A1 (en) | 1986-01-23 | 1987-07-30 | Celltech Limited | Recombinant dna sequences, vectors containing them and method for the use thereof |
US4683195B1 (en) | 1986-01-30 | 1990-11-27 | Cetus Corp | |
US4683195A (en) | 1986-01-30 | 1987-07-28 | Cetus Corporation | Process for amplifying, detecting, and/or-cloning nucleic acid sequences |
US4800159A (en) | 1986-02-07 | 1989-01-24 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences |
GB2200651A (en) | 1987-02-07 | 1988-08-10 | Al Sumidaie Ayad Mohamed Khala | A method of obtaining a retrovirus-containing fraction from retrovirus-containing cells |
US5219740A (en) | 1987-02-13 | 1993-06-15 | Fred Hutchinson Cancer Research Center | Retroviral gene transfer into diploid fibroblasts for gene therapy |
US5422120A (en) | 1988-05-30 | 1995-06-06 | Depotech Corporation | Heterovesicular liposomes |
EP0345242A2 (en) | 1988-06-03 | 1989-12-06 | Smithkline Biologicals S.A. | Expression of gag proteins from retroviruses in eucaryotic cells |
US5545807A (en) | 1988-10-12 | 1996-08-13 | The Babraham Institute | Production of antibodies from transgenic animals |
US5047335A (en) | 1988-12-21 | 1991-09-10 | The Regents Of The University Of Calif. | Process for controlling intracellular glycosylation of proteins |
US5530101A (en) | 1988-12-28 | 1996-06-25 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5585089A (en) | 1988-12-28 | 1996-12-17 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5693761A (en) | 1988-12-28 | 1997-12-02 | Protein Design Labs, Inc. | Polynucleotides encoding improved humanized immunoglobulins |
US5693762A (en) | 1988-12-28 | 1997-12-02 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US6180370B1 (en) | 1988-12-28 | 2001-01-30 | Protein Design Labs, Inc. | Humanized immunoglobulins and methods of making the same |
WO1990007936A1 (en) | 1989-01-23 | 1990-07-26 | Chiron Corporation | Recombinant therapies for infection and hyperproliferative disorders |
WO1990011092A1 (en) | 1989-03-21 | 1990-10-04 | Vical, Inc. | Expression of exogenous polynucleotide sequences in a vertebrate |
US5580859A (en) | 1989-03-21 | 1996-12-03 | Vical Incorporated | Delivery of exogenous DNA sequences in a mammal |
US6413942B1 (en) | 1989-03-21 | 2002-07-02 | Vical, Inc. | Methods of delivering a physiologically active polypeptide to a mammal |
WO1991000360A1 (en) | 1989-06-29 | 1991-01-10 | Medarex, Inc. | Bispecific reagents for aids therapy |
WO1991002805A2 (en) | 1989-08-18 | 1991-03-07 | Viagene, Inc. | Recombinant retroviruses delivering vector constructs to target cells |
US5013556A (en) | 1989-10-20 | 1991-05-07 | Liposome Technology, Inc. | Liposomes with enhanced circulation time |
EP0524968A1 (en) | 1990-03-21 | 1993-02-03 | Res Dev Foundation | Heterovesicular liposomes. |
WO1991014445A1 (en) | 1990-03-21 | 1991-10-03 | Research Development Foundation | Heterovesicular liposomes |
US5580717A (en) | 1990-05-01 | 1996-12-03 | Affymax Technologies N.V. | Recombinant library screening methods |
US5661016A (en) | 1990-08-29 | 1997-08-26 | Genpharm International Inc. | Transgenic non-human animals capable of producing heterologous antibodies of various isotypes |
US5569825A (en) | 1990-08-29 | 1996-10-29 | Genpharm International | Transgenic non-human animals capable of producing heterologous antibodies of various isotypes |
US5625126A (en) | 1990-08-29 | 1997-04-29 | Genpharm International, Inc. | Transgenic non-human animals for producing heterologous antibodies |
US5545806A (en) | 1990-08-29 | 1996-08-13 | Genpharm International, Inc. | Ransgenic non-human animals for producing heterologous antibodies |
US5633425A (en) | 1990-08-29 | 1997-05-27 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
US5750373A (en) | 1990-12-03 | 1998-05-12 | Genentech, Inc. | Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants |
US5278299A (en) | 1991-03-18 | 1994-01-11 | Scripps Clinic And Research Foundation | Method and composition for synthesizing sialylated glycosyl compounds |
WO1992020373A1 (en) | 1991-05-14 | 1992-11-26 | Repligen Corporation | Heteroconjugate antibodies for treatment of hiv infection |
EP0519596A1 (en) | 1991-05-17 | 1992-12-23 | Merck & Co. Inc. | A method for reducing the immunogenicity of antibody variable domains |
US6054297A (en) | 1991-06-14 | 2000-04-25 | Genentech, Inc. | Humanized antibodies and methods for making them |
US5997867A (en) | 1991-07-16 | 1999-12-07 | Waldmann; Herman | Method of using humanized antibody against CD18 |
WO1993003769A1 (en) | 1991-08-20 | 1993-03-04 | THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES | Adenovirus mediated transfer of genes to the gastrointestinal tract |
US5866692A (en) | 1991-09-18 | 1999-02-02 | Kyowa Hakko Kogyo Co., Ltd. | Process for producing humanized chimera antibody |
WO1993006213A1 (en) | 1991-09-23 | 1993-04-01 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
US5565332A (en) | 1991-09-23 | 1996-10-15 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
WO1993010218A1 (en) | 1991-11-14 | 1993-05-27 | The United States Government As Represented By The Secretary Of The Department Of Health And Human Services | Vectors including foreign genes and negative selective markers |
US5510261A (en) | 1991-11-21 | 1996-04-23 | The Board Of Trustees Of The Leland Stanford Juniot University | Method of controlling the degradation of glycoprotein oligosaccharides produced by cultured Chinese hamster ovary cells |
WO1993011230A1 (en) | 1991-12-02 | 1993-06-10 | Dynal As | Modified mammalian stem cell blocking viral replication |
US6350861B1 (en) | 1992-03-09 | 2002-02-26 | Protein Design Labs, Inc. | Antibodies with increased binding affinity |
WO1993019191A1 (en) | 1992-03-16 | 1993-09-30 | Centre National De La Recherche Scientifique | Defective recombinant adenoviruses expressing cytokines for use in antitumoral treatment |
US5733743A (en) | 1992-03-24 | 1998-03-31 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1993025234A1 (en) | 1992-06-08 | 1993-12-23 | The Regents Of The University Of California | Methods and compositions for targeting specific tissue |
WO1993025698A1 (en) | 1992-06-10 | 1993-12-23 | The United States Government As Represented By The | Vector particles resistant to inactivation by human serum |
WO1994003622A1 (en) | 1992-07-31 | 1994-02-17 | Imperial College Of Science, Technology & Medicine | D-type retroviral vectors, based on mpmv |
WO1994004690A1 (en) | 1992-08-17 | 1994-03-03 | Genentech, Inc. | Bispecific immunoadhesins |
US6210671B1 (en) | 1992-12-01 | 2001-04-03 | Protein Design Labs, Inc. | Humanized antibodies reactive with L-selectin |
WO1994012649A2 (en) | 1992-12-03 | 1994-06-09 | Genzyme Corporation | Gene therapy for cystic fibrosis |
US5981568A (en) | 1993-01-28 | 1999-11-09 | Neorx Corporation | Therapeutic inhibitor of vascular smooth muscle cells |
WO1994023697A1 (en) | 1993-04-22 | 1994-10-27 | Depotech Corporation | Cyclodextrin liposomes encapsulating pharmacologic compounds and methods for their use |
WO1994028938A1 (en) | 1993-06-07 | 1994-12-22 | The Regents Of The University Of Michigan | Adenovirus vectors for gene therapy sponsorship |
US6180377B1 (en) | 1993-06-16 | 2001-01-30 | Celltech Therapeutics Limited | Humanized antibodies |
WO1995000655A1 (en) | 1993-06-24 | 1995-01-05 | Mc Master University | Adenovirus vectors for gene therapy |
WO1995007994A2 (en) | 1993-09-15 | 1995-03-23 | Viagene, Inc. | Recombinant alphavirus vectors |
US5814482A (en) | 1993-09-15 | 1998-09-29 | Dubensky, Jr.; Thomas W. | Eukaryotic layered vector initiation systems |
WO1995011984A2 (en) | 1993-10-25 | 1995-05-04 | Canji, Inc. | Recombinant adenoviral vector and methods of use |
WO1995013796A1 (en) | 1993-11-16 | 1995-05-26 | Depotech Corporation | Vesicles with controlled release of actives |
WO1995030763A2 (en) | 1994-05-09 | 1995-11-16 | Chiron Viagene, Inc. | Retroviral vectors having a reduced recombination rate |
WO1996017072A2 (en) | 1994-11-30 | 1996-06-06 | Chiron Viagene, Inc. | Recombinant alphavirus vectors |
US6436908B1 (en) | 1995-05-30 | 2002-08-20 | Duke University | Use of exogenous β-adrenergic receptor and β-adrenergic receptor kinase gene constructs to enhance myocardial function |
US6265150B1 (en) | 1995-06-07 | 2001-07-24 | Becton Dickinson & Company | Phage antibodies |
WO1997042338A1 (en) | 1996-05-06 | 1997-11-13 | Chiron Corporation | Crossless retroviral vectors |
US6376471B1 (en) | 1997-10-10 | 2002-04-23 | Johns Hopkins University | Gene delivery compositions and methods |
WO1999058572A1 (en) | 1998-05-08 | 1999-11-18 | Cambridge University Technical Services Limited | Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis |
WO2000009560A2 (en) | 1998-08-17 | 2000-02-24 | Abgenix, Inc. | Generation of modified molecules with increased serum half-lives |
WO2000053211A2 (en) | 1999-03-09 | 2000-09-14 | University Of Southern California | Method of promoting myocyte proliferation and myocardial tissue repair |
WO2001027160A1 (en) | 1999-10-14 | 2001-04-19 | Applied Molecular Evolution, Inc. | Methods of optimizing antibody variable region binding affinity |
WO2004058184A2 (en) | 2002-12-24 | 2004-07-15 | Rinat Neuroscience Corp. | Anti-ngf antibodies and methods using same |
US7300754B2 (en) | 2003-04-25 | 2007-11-27 | Institut National De La Sante Et De La Recherche Medicale | Methods for detecting the presence of or predisposition to autosomal dominant hypercholesterolemia |
US7314622B2 (en) | 2005-04-15 | 2008-01-01 | Neogenix Oncology, Inc. | Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers |
Non-Patent Citations (137)
Title |
---|
"Animal Cell Culture", 1987 |
"Antibodies: a practical approach", 1988, IRL PRESS |
"Cell and Tissue Culture: Laboratory Procedures", 1993, J. WILEY AND SONS |
"Cell Biology: A Laboratory Notebook", 1998, ACADEMIC PRESS |
"Current Protocols in Immunology", 1991 |
"Current Protocols in Molecular Biology", 1987 |
"Gene Transfer Vectors for Mammalian Cells", 1987 |
"Handbook of Experimental Immunology" |
"Methods in Enzymology", ACADEMIC PRESS, INC. |
"Methods in Molecular Biology", HUMANA PRESS |
"Monoclonal antibodies: a practical approach", 2000, OXFORD UNIVERSITY PRESS |
"Oligonucleotide Synthesis", 1984 |
"PCR: The Polymerase Chain Reaction", 1994 |
"PCR: The Polymerase Chain Reaction", 1994, BIRKAUSWER PRESS |
"Remington, The Science and Practice of Pharmacy", 2000, MACK PUBLISHING |
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO. |
"Short Protocols in Molecular Biology", 1999, WILEY AND SONS |
"The Antibodies", 1995, HARWOOD ACADEMIC PUBLISHERS |
A, Q, K; H. SEE, TAO ET AL., J. IMMUNOLOGY, vol. 143, 1989, pages 2595 - 2601 |
AALBERSE ET AL., IMMUNOLOGY, vol. 105, 2002, pages 9 - 19 |
AL-LAZIKANI ET AL., J. MOLEC. BIOL., vol. 273, 1997, pages 927 - 948 |
ARMOUR ET AL., MOLECULAR IMMUNOLOGY, vol. 40, 2003, pages 585 - 593 |
BALINT ET AL., GENE, vol. 137, no. 1, 1993, pages 109 - 18 |
BARBAS ET AL., PROC NAT. ACAD. SCI, USA, vol. 91, 1994, pages 3809 - 3813 |
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426 |
BOERNER ET AL., J. IMMUNOL., vol. 147, no. 1, 1991, pages 86 - 95 |
BOYD ET AL., MOL. IMMUNOL., vol. 32, 1996, pages 1311 - 1318 |
BROWN ET AL., CANCER RES., vol. 47, 1987, pages 3577 - 3583 |
BUCK, D. W. ET AL., IN VITRO, vol. 18, 1982, pages 377 - 381 |
C.A. JANEWAY; P. TRAVERS, IMMUNOBIOLOGY, 1997 |
CAMERON ET AL., HUMAN MOLECULAR GENETICS, vol. 15, 2006, pages 1551 - 1558 |
CAPEL ET AL., IMMUNOMETHODS, vol. 4, 1994, pages 25 - 34 |
CHIOU ET AL.: "Gene Therapeutics: Methods And Applications Of Direct Gene Transfer", 1994 |
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628 |
COLE; ALAN R. LISS ET AL., MONOCLONAL ANTIBODIES AND CANCER THERAPY, 1985, pages 77 |
COX ET AL., EUR. J. IMMUNOL., vol. 24, 1994, pages 827 - 836 |
CUNNINGHAM ET AL., NAT STRUCT MOL BIOL, vol. 14, no. 5, 2007, pages 413 - 419 |
CUNNINGHAM ET AL., NAT STRUCT MOL BIOL, vol. 14, no. 5, 2007, pages 413 - 9 |
CURIEL, HUM. GENE THER., vol. 3, 1992, pages 147 |
DAUGHERTY ET AL., NUCL. ACIDS RES., vol. 19, 1991, pages 2471 - 2476 |
DAYHOFF, M.O.: "Atlas of Protein Sequence and Structure", vol. 5, 1978, NATIONAL BIOMEDICAL RESEARCH FOUNDATION, article "A model of evolutionary change in proteins - Matrices for detecting distant relationships", pages: 345 - 358 |
DE HAAS ET AL., J. LAB. CLIN. MED., vol. 126, 1995, pages 330 - 41 |
E. HARLOW; D. LANE: "Using antibodies: a laboratory manual", 1999, COLD SPRING HARBOR LABORATORY PRESS |
EPSTEIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 3688 |
EUR. J. IMMUNOL., vol. 29, 1999, pages 2613 - 2624 |
FINDEIS ET AL., TRENDS BIOTECHNOL., vol. 11, 1993, pages 202 |
FISHER ET AL., JBC, vol. 282, no. 28, 2007, pages 20502 - 12 |
FREDRICKSON ET AL., N ENGL J MED., vol. 276, 1967, pages 34 - 42,94-103,148-156,215-225,273-281 |
GRIFFITH ET AL., EMBO J., vol. 12, 1993, pages 725 - 734 |
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587 |
HARLOW; LANE: "Using Antibodies, a Laboratory Manual", 1999, COLD SPRING HARBOR LABORATORY PRESS |
HAWKINS ET AL., J. MOL. BIOL., vol. 226, 1992, pages 889 - 896 |
HEIN J.: "Methods in Enzymology", vol. 183, 1990, ACADEMIC PRESS, INC., article "Unified Approach to Alignment and Phylogenes", pages: 626 - 645 |
HIGGINS, D.G.; SHARP, P.M., CABIOS, vol. 5, 1989, pages 151 - 153 |
HOLLIGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448 |
HOLLIGER, P., PROC. NAT!. ACAD SCI. USA, vol. 90, 1993, pages 6444 - 6448 |
HOMER ET AL., ATHEROSCLEROSIS, vol. 196, 2008, pages 659 - 666 |
HOOGENBOOM; WINTER, J. MOL. BIOL., vol. 227, 1991, pages 381 |
HSE ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 9062 - 9070 |
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883 |
HWANG ET AL., PROC. NATL ACAD. SCI. USA, vol. 77, 1980, pages 4030 |
IDUSOGIE ET AL., J. IMMUNOLOGY, vol. 164, 2000, pages 4178 - 4184 |
III ET AL., PROTEIN ENG., vol. 10, 1997, pages 949 - 57 |
J.P. MATHER; P.E. ROBERTS: "Introduction to Cell and Tissue Culture", 1998, PLENUM PRESS |
JACKSON ET AL., J. IMMUNOL., vol. 154, no. 7, 1995, pages 3310 - 9 |
JEFFERIS ET AL., IMMUNOLOGICAL REVIEWS, vol. 163, 1998, pages 59 - 76 |
JEFFERIS; LUND, CHEM. IMMUNOL., vol. 65, 1997, pages 111 - 128 |
JOHNSON, KEVIN S.; CHISWELL, DAVID J., CURRENT OPINION IN STRUCTURAL BIOLOGY, vol. 3, 1993, pages 564 - 571 |
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525 |
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH |
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH |
KABAT, E. A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, NIH PUBL. |
KABAT, E. A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, NIH PUBL. NO. 91-3242 |
KARLSSON, R.; ROOS, H.; FAGERSTAM, L.; PETERSSON, B., METHODS ENZYMOLOGY, vol. 6, 1994, pages 99 - 110 |
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249 |
KOHLER, B.; MILSTEIN, C., NATURE, vol. 256, 1975, pages 495 - 497 |
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 |
KOSTELNY ET AL., J. IMMUNOL., vol. 148, 1992, pages 1547 - 1553 |
KWON ET AL., PNAS, vol. 105, 2008, pages 1820 - 1825 |
LAGACE ET AL., J CLIN INV, vol. 116, no. 11, 2006, pages 2995 - 3005 |
LAMBERT ET AL., TRENDS IN ENDOCRINOLOGY AND METABOLISM, vol. 17, 2006, pages 79 - 81 |
LOBUGLIO ET AL., PROC. NAT. ACAD. SCI. USA, vol. 86, 1989, pages 4220 - 4224 |
LONBERG, N.; D. HUSZAR, INT. REV. IMMUNOL, vol. 13, 1995, pages 65 |
LUND ET AL., J. IMMUNOLOGY, vol. 157, 1996, pages 4963 - 4969 |
MARK ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597 |
MARKS ET AL., BIO/TECHNOL., vol. 10, 1992, pages 779 - 783 |
MARKS ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 779 - 783 |
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 |
MARTIN, EMBO J., vol. 13, 1994, pages 5303 - 9 |
MAXWELL ET AL., CURR. OPIN. LIPIDOL., vol. 16, 2005, pages 167 - 172 |
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 553 |
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 554 |
MILLSTEIN; CUELLO, NATURE, vol. 305, 1983, pages 537 - 539 |
MORGAN ET AL., IMMUNOLOGY, vol. 86, 1995, pages 319 - 324 |
MORRISON ET AL., PROC. NAT. ACAD. SCI., vol. 81, 1984, pages 6851 |
MYERS, E.W.; MULLER W., CABIOS, vol. 4, 1988, pages 11 - 17 |
P. FINCH, ANTIBODIES, 1997 |
PEETERS ET AL., VACCINE, vol. 19, 2001, pages 2756 |
PHILIP, MOL. CELL BIOL., vol. 14, 1994, pages 2411 |
POLJAK, R. J. ET AL., STRUCTURE, vol. 2, 1994, pages 1121 - 1123 |
POLLOCK ET AL., J IMMUNOL METHODS, vol. 231, 1999, pages 147 |
RASHID ET AL., PROC NATL ACAD SCI USA, vol. 102, 2005, pages 5374 |
RAVETCH; KINET, ANN. REV. IMMUNOL., vol. 9, 1991, pages 457 - 92 |
REMINGTON: "The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS AND WILKINS |
REMINGTON: "The Science and Practice of Pharmacy", 2000, MACK PUBLISHING |
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327 |
ROBINSON, E.D., COMB. THEOR., vol. 11, 1971, pages 105 |
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR PRESS |
SANTOU, N.; NES, M., MOL. BIOL. EVOL., vol. 4, 1987, pages 406 - 425 |
SCHIER ET AL., GENE, vol. 169, 1995, pages 147 - 155 |
SHAW ET AL., J IMMUNOL., vol. 138, 1987, pages 4534 - 4538 |
SHEETS ET AL., PROC. NATL. ACAD. SCI. (USA), vol. 95, 1998, pages 6157 - 6162 |
SNEATH, P.H.A.; SOKAL, R.R.: "Numerical Taxonomy the Principles and Practice of Numerical Taxonomy", 1973, FREEMAN PRESS |
SONGSIVILAI; LACHMANN, CLIN. EXP. IMMUNOL., vol. 79, 1990, pages 315 - 321 |
SURESH ET AL., METHODS IN ENZYMOLOGY, vol. 121, 1986, pages 210 |
TAO ET AL., J. IMMUNOLOGY, vol. 143, 1989, pages 2595 - 2601 |
TILLER ET AL., J. IMMUNOL. METHODS, vol. 329, 2008, pages 112 |
TOMLINSON ET AL., J. MOL. BIOL., vol. 227, 1992, pages 776 - 798 |
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655 - 3659 |
TRAUNECKER ET AL., INT. J. CANCER, vol. 7, 1992, pages 51 - 52 |
UMANA ET AL., NATURE BIOTECH., vol. 17, 1999, pages 176 - 180 |
VAUGHAN ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 309 - 314 |
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536 |
WATERHOUSE ET AL., NUCL. ACIDS RES., vol. 21, 1993, pages 2265 - 2266 |
WILBUR, W.J.; LIPMAN, D.J., PROC. NAT!. ACAD. SCI. USA, vol. 80, 1983, pages 726 - 730 |
WINTER ET AL., ANNU. REV. IMMUNOL., vol. 12, 1994, pages 433 - 455 |
WINTER ET AL., NATURE, vol. 349, 1991, pages 293 - 299 |
WITTWE; HOWARD, BIOCHEM., vol. 29, 1990, pages 4175 - 4180 |
WOFFENDIN, PROC. NATL. ACAD. SCI., vol. 91, 1994, pages 1581 |
WRIGHT; MORRISON, TIBTECH, vol. 15, 1997, pages 26 - 32 |
WU ET AL., J. BIOL. CHEM., vol. 263, 1988, pages 621 |
WU ET AL., J. BIOL. CHEM., vol. 266, 1991, pages 338 |
WU ET AL., J. BIOL. CHEM., vol. 269, 1994, pages 542 |
WU, J., BIOL. CHEM., vol. 264, 1989, pages 16985 |
WYSS; WAGNER, CURRENT OPIN. BIOTECH., vol. 7, 1996, pages 409 - 416 |
YELTON ET AL., J. IMMUNOL., vol. 155, 1995, pages 1994 - 2004 |
ZENKE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 3655 |
Cited By (264)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US8344114B2 (en) | 2006-11-07 | 2013-01-01 | Merck Sharp & Dohme Corp. | Antagonists of PCSK9 |
US8168762B2 (en) | 2007-08-23 | 2012-05-01 | 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) |
US8563698B2 (en) | 2007-08-23 | 2013-10-22 | Amgen Inc. | Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9) |
US8829165B2 (en) | 2007-08-23 | 2014-09-09 | Amgen, Inc. | Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9) |
US8859741B2 (en) | 2007-08-23 | 2014-10-14 | 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) |
US8981064B2 (en) | 2007-08-23 | 2015-03-17 | 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) |
US8030457B2 (en) | 2007-08-23 | 2011-10-04 | 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) |
US9920134B2 (en) | 2007-08-23 | 2018-03-20 | Amgen Inc. | Monoclonal antibodies 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) |
US8889834B2 (en) | 2007-08-23 | 2014-11-18 | Amgen Inc. | Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9) |
US8697070B2 (en) | 2008-02-07 | 2014-04-15 | Merck Sharp & Dohme Corp. | 1D05 PCSK9 antagonists |
US8188233B2 (en) | 2008-02-07 | 2012-05-29 | Merck Sharp & Dohme Corp. | 1B20 PCSK9 antagonists |
US8188234B2 (en) | 2008-02-07 | 2012-05-29 | Merck Sharp & Dohme Corp. | 1D05 PCSK9 antagonists |
US8957194B2 (en) | 2008-02-07 | 2015-02-17 | Merck Sharpe & Dohme Corp. | 1B20 PCSK9 antagonists |
US10611835B2 (en) * | 2008-07-10 | 2020-04-07 | Toray Industries, Inc. | Pharmaceutical composition for treatment and prevention of cancer |
US20110129478A1 (en) * | 2008-07-10 | 2011-06-02 | Toray Industries, Inc. | Pharmaceutical composition for treatment and prevention of cancer |
US10941210B2 (en) | 2008-12-15 | 2021-03-09 | Regeneron Pharmaceuticals, Inc. | Anti-PCSK9 antibodies |
US9724411B2 (en) | 2008-12-15 | 2017-08-08 | Regeneron Pharmaceuticals, Inc. | Methods for treating hypercholesterolemia and reducing LDL-C using antibodies to PCSK9 |
US9550837B2 (en) | 2008-12-15 | 2017-01-24 | Regeneron Pharmaceuticals, Inc. | Therapeutic uses of anti-PCSK9 antibodies |
US10023654B2 (en) | 2008-12-15 | 2018-07-17 | Regeneron Pharmaceuticals, Inc. | Anti-PCSK9 antibodies |
EP2493507A4 (en) * | 2009-10-30 | 2013-11-20 | Merck Sharp & Dohme | Ax213 and ax132 pcsk9 antagonists and variants |
US8877900B2 (en) | 2009-10-30 | 2014-11-04 | Merck Sharp & Dohme Corp. | AX132 PCSK9 antagonists |
EP2493507A2 (en) * | 2009-10-30 | 2012-09-05 | Merck Sharp & Dohme Corp. | Ax213 and ax132 pcsk9 antagonists and variants |
EP3424949A1 (en) * | 2010-04-13 | 2019-01-09 | Bristol-Myers Squibb Company | Fibronectin based scaffold domain proteins that bind pcsk9 |
GB2481373A (en) * | 2010-06-21 | 2011-12-28 | Weiming Xu | Treatment of hypercholesterolaemia by ubiquitination of PCSK9 |
JP2014511106A (en) * | 2010-12-22 | 2014-05-08 | ジェネンテック, インコーポレイテッド | Anti-PCSK9 antibody and method of use |
WO2012088313A1 (en) * | 2010-12-22 | 2012-06-28 | Genentech, Inc. | Anti-pcsk9 antibodies and methods of use |
EP2668211A1 (en) * | 2011-01-28 | 2013-12-04 | Sanofi | Pharmaceutical compositions comprising human antibodies to pcsk9 |
RU2721279C2 (en) * | 2011-01-28 | 2020-05-18 | Санофи Байотекнолоджи | Human antibodies to pcsk9 for use in methods of treating specific groups of individuals |
AU2012210480B2 (en) * | 2011-01-28 | 2017-05-18 | Sanofi Biotechnology | Human antibodies to PCSK9 for use in methods of treating particular groups of subjects |
JP2014508142A (en) * | 2011-01-28 | 2014-04-03 | サノフイ | Pharmaceutical composition comprising a human antibody against PCSK9 |
JP2017095490A (en) * | 2011-01-28 | 2017-06-01 | サノフィ・バイオテクノロジー | Human antibody against pcsk9 for use in method of treating particular group of subjects |
RU2603481C2 (en) * | 2011-01-28 | 2016-11-27 | Санофи Байотекнолоджи | Human antibodies to pcsk9 for use in methods of treating particular groups of subjects |
JP2017095491A (en) * | 2011-01-28 | 2017-06-01 | サノフィ・バイオテクノロジー | Pharmaceutical composition comprising human antibody to psck9 |
JP2014511361A (en) * | 2011-01-28 | 2014-05-15 | サノフイ | Human antibodies to PCSK9 for use in a method of treating a particular group of subjects |
EP3326648A1 (en) * | 2011-01-28 | 2018-05-30 | Sanofi Biotechnology | Pharmaceutical compositions comprising human antibodies to pcsk9 |
US9682013B2 (en) | 2011-01-28 | 2017-06-20 | Sanofi Biotechnology | Pharmaceutical compositions comprising human antibodies to PCSK9 |
US11246925B2 (en) | 2011-01-28 | 2022-02-15 | Sanofi Biotechnology | Human antibodies to PCSK9 for use in methods of treating particular groups of subjects |
WO2012101252A3 (en) * | 2011-01-28 | 2012-11-15 | Sanofi | Human antibodies to pcsk9 for use in methods of treating particular groups of subjects |
EP3395836A1 (en) * | 2011-01-28 | 2018-10-31 | Sanofi Biotechnology | Human antibodies to pcsk9 for use in methods of treating particular groups of subjects |
US9561155B2 (en) | 2011-01-28 | 2017-02-07 | Sanofi Biotechnology | Method of reducing cholesterol levels using a human anti-PCSK9 antibody |
US12083176B2 (en) | 2011-01-28 | 2024-09-10 | Sanofi Biotechnology | Human antibodies to PCSK9 for use in methods of treating particular groups of subjects |
US20160152734A1 (en) * | 2011-07-14 | 2016-06-02 | Pfizer Inc. | Treatment with anti-pcsk9 antibodies |
WO2013008185A1 (en) | 2011-07-14 | 2013-01-17 | Pfizer Inc. | Treatment with anti-pcsk9 antibodies |
JP2017145256A (en) * | 2011-07-14 | 2017-08-24 | ファイザー・インク | Treatment with anti-pcsk9 antibodies |
JP2013023499A (en) * | 2011-07-14 | 2013-02-04 | Pfizer Inc | Treatment with anti-pcsk9 antibody |
CN104093423A (en) * | 2011-07-14 | 2014-10-08 | 辉瑞公司 | Treatment with anti-pcsk9 antibodies |
US11673967B2 (en) | 2011-07-28 | 2023-06-13 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-PCSK9 antibodies |
US10752701B2 (en) | 2011-07-28 | 2020-08-25 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-PCSK9 antibodies |
US10472425B2 (en) | 2011-07-28 | 2019-11-12 | Regeneron Pharmaceuticals, Inc. | Stabilized formulations containing anti-PCSK9 antibodies |
US11116839B2 (en) | 2011-09-16 | 2021-09-14 | Regeneron Pharmaceuticals, Inc. | Methods for reducing lipoprotein(a) levels by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9) |
US8530414B2 (en) | 2011-09-16 | 2013-09-10 | Eli Lilly And Company | Antibodies to PCSK9 and uses thereof |
JP2017206520A (en) * | 2011-09-16 | 2017-11-24 | リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. | METHODS FOR REDUCING LIPOPROTEIN (a) LEVELS BY ADMINISTERING AN INHIBITOR OF PROPROTEIN CONVERTASE SUBTILISIN KEXIN-9 (PCSK9) |
JP2014530188A (en) * | 2011-09-16 | 2014-11-17 | イーライ リリー アンド カンパニー | Antibodies against PCSK9 and uses thereof |
US10076571B2 (en) | 2011-09-16 | 2018-09-18 | Regeneron Pharmaceuticals, Inc. | Methods for reducing lipoprotein(a) levels by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9) |
JP2014527967A (en) * | 2011-09-16 | 2014-10-23 | リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. | Method of reducing lipoprotein (a) levels by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9) |
WO2013039958A1 (en) | 2011-09-16 | 2013-03-21 | Eli Lilly And Company | Antibodies to pcsk9 and uses thereof |
WO2013086443A1 (en) | 2011-12-08 | 2013-06-13 | Amgen Inc. | Agonistic human lcat antigen binding proteins and their use in therapy |
WO2013091103A1 (en) * | 2011-12-20 | 2013-06-27 | Adaerata, Limited Partnership | Single domain antibodies as inhibitors of pcsk9 |
US9879093B2 (en) | 2011-12-20 | 2018-01-30 | Adaerata, Limited Partnershp | Single domain antibodies as inhibitors of PCSK9 |
CN104169304A (en) * | 2011-12-20 | 2014-11-26 | 阿达拉塔合伙有限公司 | Single domain antibodies as inhibitors of PCSK9 |
KR20150006471A (en) * | 2012-05-08 | 2015-01-16 | 앨더바이오 홀딩스 엘엘씨 | Anti-pcsk9 antibodies and use thereof |
US10259885B2 (en) | 2012-05-08 | 2019-04-16 | Alderbio Holdings Llc | Anti-PCSK9 antibodies and use thereof |
EP2844673A4 (en) * | 2012-05-08 | 2016-04-20 | Alderbio Holdings Llc | Anti-pcsk9 antibodies and use thereof |
KR102137291B1 (en) * | 2012-05-08 | 2020-07-23 | 앨더바이오 홀딩스 엘엘씨 | Anti-pcsk9 antibodies and use thereof |
US9255154B2 (en) | 2012-05-08 | 2016-02-09 | Alderbio Holdings, Llc | Anti-PCSK9 antibodies and use thereof |
WO2013188855A1 (en) * | 2012-06-15 | 2013-12-19 | Genentech, Inc. | Anti-pcsk9 antibodies, formulations, dosing, and methods of use |
US9266961B2 (en) | 2012-06-15 | 2016-02-23 | Genentech, Inc. | Anti-PCSK9 antibodies, formulations, dosing, and methods of use |
US9540449B2 (en) | 2012-08-13 | 2017-01-10 | Regeneron Pharmaceuticals, Inc. | Anti-PCSK9 antibodies with pH-dependent binding characteristics |
EP3081249A1 (en) | 2012-11-21 | 2016-10-19 | Amgen, Inc | Drug delivery device |
US10682474B2 (en) | 2012-11-21 | 2020-06-16 | Amgen Inc. | Drug delivery device |
EP4234694A2 (en) | 2012-11-21 | 2023-08-30 | Amgen Inc. | Drug delivery device |
WO2014081780A1 (en) | 2012-11-21 | 2014-05-30 | Amgen Inc. | Drug delivery device |
US11458247B2 (en) | 2012-11-21 | 2022-10-04 | Amgen Inc. | Drug delivery device |
US12115341B2 (en) | 2012-11-21 | 2024-10-15 | Amgen Inc. | Drug delivery device |
EP3072548A1 (en) | 2012-11-21 | 2016-09-28 | Amgen, Inc | Drug delivery device |
US11439745B2 (en) | 2012-11-21 | 2022-09-13 | Amgen Inc. | Drug delivery device |
US11344681B2 (en) | 2012-11-21 | 2022-05-31 | Amgen Inc. | Drug delivery device |
EP3656426A1 (en) | 2012-11-21 | 2020-05-27 | Amgen, Inc | Drug delivery device |
WO2014144096A1 (en) | 2013-03-15 | 2014-09-18 | Amgen Inc. | Drug cassette, autoinjector, and autoinjector system |
WO2014143770A1 (en) | 2013-03-15 | 2014-09-18 | Amgen Inc. | Body contour adaptable autoinjector device |
WO2014150983A3 (en) * | 2013-03-15 | 2014-12-04 | Amgen Inc. | Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9 (pcsk9) |
EP3593839A1 (en) | 2013-03-15 | 2020-01-15 | Amgen Inc. | Drug cassette |
WO2014149699A1 (en) | 2013-03-15 | 2014-09-25 | Eli Lilly And Company | Bifunctional protein |
WO2014150983A2 (en) * | 2013-03-15 | 2014-09-25 | Amgen Inc. | Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9 |
WO2014149357A1 (en) | 2013-03-22 | 2014-09-25 | Amgen Inc. | Injector and method of assembly |
EP3831427A1 (en) | 2013-03-22 | 2021-06-09 | Amgen Inc. | Injector and method of assembly |
US10111953B2 (en) | 2013-05-30 | 2018-10-30 | Regeneron Pharmaceuticals, Inc. | Methods for reducing remnant cholesterol and other lipoprotein fractions by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9) |
WO2014197752A1 (en) | 2013-06-07 | 2014-12-11 | Regeneron Pharmaceuticals, Inc. | Methods fo inhibting atherosclerosis by administering an inhibitor of pcsk9 |
US10494442B2 (en) | 2013-06-07 | 2019-12-03 | Sanofi Biotechnology | Methods for inhibiting atherosclerosis by administering an inhibitor of PCSK9 |
US10995150B2 (en) | 2013-06-07 | 2021-05-04 | Regeneron Pharmaceuticals, Inc. | Methods for inhibiting atherosclerosis by administering an anti-PCSK9 antibody |
EP2862877A1 (en) | 2013-10-18 | 2015-04-22 | Sanofi | Methods for inhibiting atherosclerosis by administering an inhibitor of PCSK9 |
WO2015061389A1 (en) | 2013-10-24 | 2015-04-30 | Amgen Inc. | Drug delivery system with temperature-sensitive control |
WO2015061386A1 (en) | 2013-10-24 | 2015-04-30 | Amgen Inc. | Injector and method of assembly |
EP3501575A1 (en) | 2013-10-24 | 2019-06-26 | Amgen, Inc | Drug delivery system with temperature-sensitive-control |
EP3957345A1 (en) | 2013-10-24 | 2022-02-23 | Amgen, Inc | Drug delivery system with temperature-sensitive control |
EP3421066A1 (en) | 2013-10-24 | 2019-01-02 | Amgen, Inc | Injector and method of assembly |
EP3789064A1 (en) | 2013-10-24 | 2021-03-10 | Amgen, Inc | Injector and method of assembly |
US10428157B2 (en) | 2013-11-12 | 2019-10-01 | Sanofi Biotechnology | Dosing regimens for use with PCSK9 inhibitors |
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 |
US8883157B1 (en) | 2013-12-17 | 2014-11-11 | Kymab Limited | Targeting rare human PCSK9 variants for cholesterol treatment |
DE202014010499U1 (en) | 2013-12-17 | 2015-10-20 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
US10611849B2 (en) | 2013-12-17 | 2020-04-07 | Kymab Limited | Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment |
DE202014010421U1 (en) | 2013-12-17 | 2015-11-12 | Kymab Limited | Human goals |
US10618971B2 (en) | 2013-12-17 | 2020-04-14 | Kymab Limited | Targeting rare human PCSK9 variants for cholesterol treatment |
WO2015119906A1 (en) | 2014-02-05 | 2015-08-13 | Amgen Inc. | Drug delivery system with electromagnetic field generator |
US11773175B2 (en) | 2014-03-04 | 2023-10-03 | Kymab Limited | Antibodies, uses and methods |
US11753479B2 (en) | 2014-03-04 | 2023-09-12 | Kymab Limited | Nucleic acids encoding anti-OX40L antibodies |
EP3785749A1 (en) | 2014-05-07 | 2021-03-03 | Amgen Inc. | Autoinjector with shock reducing elements |
WO2015171777A1 (en) | 2014-05-07 | 2015-11-12 | Amgen Inc. | Autoinjector with shock reducing elements |
US11738146B2 (en) | 2014-06-03 | 2023-08-29 | Amgen Inc. | Drug delivery system and method of use |
WO2015187799A1 (en) | 2014-06-03 | 2015-12-10 | Amgen Inc. | Systems and methods for remotely processing data collected by a drug delivery device |
WO2015187797A1 (en) | 2014-06-03 | 2015-12-10 | Amgen Inc. | Controllable drug delivery system and method of use |
US11213624B2 (en) | 2014-06-03 | 2022-01-04 | Amgen Inc. | Controllable drug delivery system and method of use |
WO2015187793A1 (en) | 2014-06-03 | 2015-12-10 | Amgen Inc. | Drug delivery system and method of use |
EP4036924A1 (en) | 2014-06-03 | 2022-08-03 | Amgen, Inc | Devices and methods for assisting a user of a drug delivery device |
EP4362039A2 (en) | 2014-06-03 | 2024-05-01 | Amgen Inc. | Controllable drug delivery system and method of use |
US11992659B2 (en) | 2014-06-03 | 2024-05-28 | Amgen Inc. | Controllable drug delivery system and method of use |
US9914769B2 (en) | 2014-07-15 | 2018-03-13 | Kymab Limited | Precision medicine 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 |
US8980273B1 (en) | 2014-07-15 | 2015-03-17 | 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 |
US8986691B1 (en) | 2014-07-15 | 2015-03-24 | Kymab Limited | Method of treating atopic dermatitis or asthma using antibody to IL4RA |
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 |
US9017678B1 (en) | 2014-07-15 | 2015-04-28 | Kymab Limited | Method of treating rheumatoid arthritis using antibody to IL6R |
US9023359B1 (en) | 2014-07-15 | 2015-05-05 | Kymab Limited | Targeting rare human PCSK9 variants for cholesterol treatment |
US9034331B1 (en) | 2014-07-15 | 2015-05-19 | 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 |
US11555066B2 (en) | 2014-07-15 | 2023-01-17 | Kymab Limited | Precision medicine for cholesterol treatment |
US9045548B1 (en) | 2014-07-15 | 2015-06-02 | Kymab Limited | Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment |
EP3332790A1 (en) | 2014-07-15 | 2018-06-13 | Kymab Limited | Antibodies for use in treating conditions related to specific pcsk9 variants in specific patients populations |
US9045545B1 (en) | 2014-07-15 | 2015-06-02 | Kymab Limited | Precision medicine by targeting PD-L1 variants for treatment of cancer |
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 |
US9068012B1 (en) | 2014-07-15 | 2015-06-30 | Kymab Limited | Targeting rare human PCSK9 variants for cholesterol treatment |
DE202015009002U1 (en) | 2014-07-15 | 2016-08-18 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
US9067998B1 (en) | 2014-07-15 | 2015-06-30 | Kymab Limited | Targeting PD-1 variants for treatment of cancer |
US9109034B1 (en) | 2014-07-15 | 2015-08-18 | Kymab Limited | Precision medicine by targeting PD-L1 variants for treatment of cancer |
US9394568B2 (en) | 2014-07-15 | 2016-07-19 | Kymab Limited | Methods of treating anaemia |
US9139648B1 (en) | 2014-07-15 | 2015-09-22 | Kymab Limited | Precision medicine by targeting human NAV1.9 variants for treatment of pain |
DE202015009006U1 (en) | 2014-07-15 | 2016-08-19 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
US9439963B2 (en) | 2014-07-15 | 2016-09-13 | Kymab Limited | Methods of treating anaemia |
US9187562B1 (en) | 2014-07-15 | 2015-11-17 | Kymab Limited | Methods for treating anaemia |
EP2975059A1 (en) | 2014-07-15 | 2016-01-20 | Kymab Limited | Antibodies for use in treating conditions related to specific pcsk9 variants in specific patients populations |
WO2016008899A1 (en) | 2014-07-15 | 2016-01-21 | Kymab Limited | Targeting human pcsk9 for cholesterol treatment |
DE202015008988U1 (en) | 2014-07-15 | 2016-06-30 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
DE202015009007U1 (en) | 2014-07-15 | 2016-08-19 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
US9428578B2 (en) | 2014-07-15 | 2016-08-30 | Kymab Limited | Methods of treating anaemia |
DE202015008974U1 (en) | 2014-07-15 | 2016-06-30 | Kymab Limited | Targeting of human PCSK9 for cholesterol treatment |
US10618955B2 (en) | 2014-07-15 | 2020-04-14 | Kymab Limited | Methods for treating neurodegenerative disease using anti-PD-1 antibodies |
US10711059B2 (en) | 2014-07-15 | 2020-07-14 | Kymab Limited | Methods for treating neurodegenerative diseases using anti-PD-L1 antibodies |
US9303089B2 (en) | 2014-07-15 | 2016-04-05 | Kymab Limited | Methods of treating anaemia |
US10544232B2 (en) | 2014-07-16 | 2020-01-28 | Sanofi Biotechnology | Methods for treating patients with heterozygous familial hypercholesterolemia (heFH) with an anti-PCSK9 antibody |
US11306155B2 (en) | 2014-07-16 | 2022-04-19 | Sanofi Biotechnology | Methods for treating patients with heterozygous familial hypercholesterolemia (heFH) with an anti-PCSK9 antibody |
WO2016020798A1 (en) | 2014-08-06 | 2016-02-11 | Rinat Neuroscience Corp. | Methods for reducing ldl-cholesterol |
WO2016020799A1 (en) | 2014-08-06 | 2016-02-11 | Rinat Neuroscience Corp. | Methods for reducing ldl-cholesterol |
WO2016023916A1 (en) | 2014-08-12 | 2016-02-18 | Kymab Limited | Treatment of disease using ligand binding to targets of interest |
WO2016046684A1 (en) | 2014-09-23 | 2016-03-31 | Pfizer Inc. | Treatment with anti-pcsk9 antibodies |
US10472424B2 (en) | 2014-09-23 | 2019-11-12 | Pfizer Inc. | Treatment with anti-PCSK9 antibodies |
WO2016061220A2 (en) | 2014-10-14 | 2016-04-21 | Amgen Inc. | Drug injection device with visual and audio indicators |
EP3943135A2 (en) | 2014-10-14 | 2022-01-26 | Amgen Inc. | Drug injection device with visual and audible indicators |
WO2016071701A1 (en) | 2014-11-07 | 2016-05-12 | Kymab Limited | Treatment of disease using ligand binding to targets of interest |
WO2016085820A1 (en) * | 2014-11-24 | 2016-06-02 | Shire Human Genetic Therapies, Inc. | Lysosomal targeting and uses thereof |
EP3848072A1 (en) | 2014-12-19 | 2021-07-14 | Amgen Inc. | Drug delivery device with proximity sensor |
US11357916B2 (en) | 2014-12-19 | 2022-06-14 | Amgen Inc. | Drug delivery device with live button or user interface field |
US11944794B2 (en) | 2014-12-19 | 2024-04-02 | Amgen Inc. | Drug delivery device with proximity sensor |
US10765801B2 (en) | 2014-12-19 | 2020-09-08 | Amgen Inc. | Drug delivery device with proximity sensor |
WO2016100055A1 (en) | 2014-12-19 | 2016-06-23 | Amgen Inc. | Drug delivery device with live button or user interface field |
WO2016100781A1 (en) | 2014-12-19 | 2016-06-23 | Amgen Inc. | Drug delivery device with proximity sensor |
EP3689394A1 (en) | 2014-12-19 | 2020-08-05 | Amgen Inc. | Drug delivery device with live button or user interface field |
US10799630B2 (en) | 2014-12-19 | 2020-10-13 | Amgen Inc. | Drug delivery device with proximity sensor |
EP3556411A1 (en) | 2015-02-17 | 2019-10-23 | Amgen Inc. | Drug delivery device with vacuum assisted securement and/or feedback |
WO2016133947A1 (en) | 2015-02-17 | 2016-08-25 | Amgen Inc. | Drug delivery device with vacuum assisted securement and/or feedback |
EP3981450A1 (en) | 2015-02-27 | 2022-04-13 | Amgen, Inc | Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement |
WO2016138434A1 (en) | 2015-02-27 | 2016-09-01 | Amgen Inc. | Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement |
US10688119B2 (en) | 2015-03-20 | 2020-06-23 | Aarhus Universitet | Inhibitors of PCSK9 for treatment of lipoprotein metabolism disorders |
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 |
US10772956B2 (en) | 2015-08-18 | 2020-09-15 | Regeneron Pharmaceuticals, Inc. | Methods for reducing or eliminating the need for lipoprotein apheresis in patients with hyperlipidemia by administering alirocumab |
WO2017039786A1 (en) | 2015-09-02 | 2017-03-09 | Amgen Inc. | Syringe assembly adapter for a syringe |
WO2017055966A1 (en) | 2015-10-01 | 2017-04-06 | Pfizer Inc. | Low viscosity antibody compositions |
WO2017100501A1 (en) | 2015-12-09 | 2017-06-15 | Amgen Inc. | Auto-injector with signaling cap |
RU2739208C2 (en) * | 2015-12-31 | 2020-12-21 | Цзянсу Хэнжуй Медицин Ко., Лтд. | Anti-pcsk9 antibody, its antigen-binding domain and medical application thereof |
EP3398968A4 (en) * | 2015-12-31 | 2019-08-07 | Jiangsu Hengrui Medicine Co., Ltd. | Pcsk9 antibody, antigen-binding fragment thereof, and medicinal application thereof |
EP3401336A4 (en) * | 2016-01-05 | 2020-01-22 | Jiangsu Hengrui Medicine Co., Ltd. | Pcsk9 antibody, antigen-binding fragment thereof, and medical uses thereof |
WO2017120178A1 (en) | 2016-01-06 | 2017-07-13 | Amgen Inc. | Auto-injector with signaling electronics |
US10822385B2 (en) | 2016-01-13 | 2020-11-03 | Novo Nordisk A/S | EGF(A) analogues with fatty acid substituents |
EP4035711A1 (en) | 2016-03-15 | 2022-08-03 | Amgen Inc. | Reducing probability of glass breakage in drug delivery devices |
EP3721922A1 (en) | 2016-03-15 | 2020-10-14 | Amgen Inc. | Reducing probability of glass breakage in drug delivery devices |
WO2017160799A1 (en) | 2016-03-15 | 2017-09-21 | Amgen Inc. | Reducing probability of glass breakage in drug delivery devices |
WO2017189089A1 (en) | 2016-04-29 | 2017-11-02 | Amgen Inc. | Drug delivery device with messaging label |
WO2017192287A1 (en) | 2016-05-02 | 2017-11-09 | Amgen Inc. | Syringe adapter and guide for filling an on-body injector |
WO2017197222A1 (en) | 2016-05-13 | 2017-11-16 | Amgen Inc. | Vial sleeve assembly |
WO2017200989A1 (en) | 2016-05-16 | 2017-11-23 | Amgen Inc. | Data encryption in medical devices with limited computational capability |
WO2017209899A1 (en) | 2016-06-03 | 2017-12-07 | Amgen Inc. | Impact testing apparatuses and methods for drug delivery devices |
WO2018004842A1 (en) | 2016-07-01 | 2018-01-04 | Amgen Inc. | Drug delivery device having minimized risk of component fracture upon impact events |
WO2018034784A1 (en) | 2016-08-17 | 2018-02-22 | Amgen Inc. | Drug delivery device with placement detection |
EP3515950A4 (en) * | 2016-09-20 | 2020-10-28 | Wuxi Biologics Ireland Limited. | Novel anti-pcsk9 antibodies |
US11173177B2 (en) | 2016-09-20 | 2021-11-16 | Aarhus Universitet | Compounds for treatment of lipoprotein metabolism disorders |
WO2018054240A1 (en) * | 2016-09-20 | 2018-03-29 | Wuxi Biologics (Shanghai) Co. Ltd. | Novel anti-pcsk9 antibodies |
US11111313B2 (en) | 2016-09-20 | 2021-09-07 | WuXi Biologics Ireland Limited | Anti-PCSK9 antibodies |
EP3515949A4 (en) * | 2016-09-20 | 2020-10-28 | Wuxi Biologics Ireland Limited. | Novel anti-pcsk9 antibodies |
WO2018081234A1 (en) | 2016-10-25 | 2018-05-03 | Amgen Inc. | On-body injector |
US11779604B2 (en) | 2016-11-03 | 2023-10-10 | Kymab Limited | Antibodies, combinations comprising antibodies, biomarkers, uses and methods |
US11485795B2 (en) | 2016-12-24 | 2022-11-01 | Innovent Biologics (Suzhou) Co., Ltd | Anti-PCSK9 antibody and use thereof |
WO2018113781A1 (en) | 2016-12-24 | 2018-06-28 | 信达生物制药(苏州)有限公司 | Anti-pcsk9 antibody and application thereof |
WO2018136398A1 (en) | 2017-01-17 | 2018-07-26 | Amgen Inc. | Injection devices and related methods of use and assembly |
WO2018151890A1 (en) | 2017-02-17 | 2018-08-23 | Amgen Inc. | Drug delivery device with sterile fluid flowpath and related method of assembly |
WO2018152073A1 (en) | 2017-02-17 | 2018-08-23 | Amgen Inc. | Insertion mechanism for drug delivery device |
WO2018165143A1 (en) | 2017-03-06 | 2018-09-13 | Amgen Inc. | Drug delivery device with activation prevention feature |
WO2018164829A1 (en) | 2017-03-07 | 2018-09-13 | Amgen Inc. | Needle insertion by overpressure |
WO2018165499A1 (en) | 2017-03-09 | 2018-09-13 | Amgen Inc. | Insertion mechanism for drug delivery device |
EP4241807A2 (en) | 2017-03-28 | 2023-09-13 | Amgen Inc. | Plunger rod and syringe assembly system and method |
WO2018183039A1 (en) | 2017-03-28 | 2018-10-04 | Amgen Inc. | Plunger rod and syringe assembly system and method |
WO2018226565A1 (en) | 2017-06-08 | 2018-12-13 | Amgen Inc. | Torque driven drug delivery device |
WO2018226515A1 (en) | 2017-06-08 | 2018-12-13 | Amgen Inc. | Syringe assembly for a drug delivery device and method of assembly |
WO2018236619A1 (en) | 2017-06-22 | 2018-12-27 | Amgen Inc. | Device activation impact/shock reduction |
WO2018237225A1 (en) | 2017-06-23 | 2018-12-27 | Amgen Inc. | Electronic drug delivery device comprising a cap activated by a switch assembly |
WO2019014014A1 (en) | 2017-07-14 | 2019-01-17 | Amgen Inc. | Needle insertion-retraction system having dual torsion spring system |
US11130794B2 (en) | 2017-07-19 | 2021-09-28 | Novo Nordisk A/S | Bifunctional compounds |
WO2019018169A1 (en) | 2017-07-21 | 2019-01-24 | Amgen Inc. | Gas permeable sealing member for drug container and methods of assembly |
EP4292576A2 (en) | 2017-07-21 | 2023-12-20 | Amgen Inc. | Gas permeable sealing member for drug container and methods of assembly |
EP4085942A1 (en) | 2017-07-25 | 2022-11-09 | Amgen Inc. | Drug delivery device with gear module and related method of assembly |
WO2019022951A1 (en) | 2017-07-25 | 2019-01-31 | Amgen Inc. | Drug delivery device with gear module and related method of assembly |
WO2019022950A1 (en) | 2017-07-25 | 2019-01-31 | Amgen Inc. | Drug delivery device with container access system and related method of assembly |
WO2019032482A2 (en) | 2017-08-09 | 2019-02-14 | Amgen Inc. | Hydraulic-pneumatic pressurized chamber drug delivery system |
WO2019036181A1 (en) | 2017-08-18 | 2019-02-21 | Amgen Inc. | Wearable injector with sterile adhesive patch |
WO2019040548A1 (en) | 2017-08-22 | 2019-02-28 | Amgen Inc. | Needle insertion mechanism for drug delivery device |
WO2019070472A1 (en) | 2017-10-04 | 2019-04-11 | Amgen Inc. | Flow adapter for drug delivery device |
WO2019070552A1 (en) | 2017-10-06 | 2019-04-11 | Amgen Inc. | Drug delivery device with interlock assembly and related method of assembly |
EP4257164A2 (en) | 2017-10-06 | 2023-10-11 | Amgen Inc. | Drug delivery device with interlock assembly and related method of assembly |
WO2019074579A1 (en) | 2017-10-09 | 2019-04-18 | Amgen Inc. | Drug delivery device with drive assembly and related method of assembly |
WO2019090079A1 (en) | 2017-11-03 | 2019-05-09 | Amgen Inc. | System and approaches for sterilizing a drug delivery device |
WO2019090086A1 (en) | 2017-11-03 | 2019-05-09 | Amgen Inc. | Systems and approaches for sterilizing a drug delivery device |
WO2019089178A1 (en) | 2017-11-06 | 2019-05-09 | Amgen Inc. | Drug delivery device with placement and flow sensing |
WO2019090303A1 (en) | 2017-11-06 | 2019-05-09 | Amgen Inc. | Fill-finish assemblies and related methods |
WO2019094138A1 (en) | 2017-11-10 | 2019-05-16 | Amgen Inc. | Plungers for drug delivery devices |
WO2019099324A1 (en) | 2017-11-16 | 2019-05-23 | Amgen Inc. | Door latch mechanism for drug delivery device |
WO2019099322A1 (en) | 2017-11-16 | 2019-05-23 | Amgen Inc. | Autoinjector with stall and end point detection |
WO2019231582A1 (en) | 2018-05-30 | 2019-12-05 | Amgen Inc. | Thermal spring release mechanism for a drug delivery device |
WO2019231618A1 (en) | 2018-06-01 | 2019-12-05 | Amgen Inc. | Modular fluid path assemblies for drug delivery devices |
WO2020023444A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Delivery devices for administering drugs |
WO2020023451A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Delivery devices for administering drugs |
WO2020023336A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Hybrid drug delivery devices with grip portion |
WO2020023220A1 (en) | 2018-07-24 | 2020-01-30 | Amgen Inc. | Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation |
WO2020028009A1 (en) | 2018-07-31 | 2020-02-06 | Amgen Inc. | Fluid path assembly for a drug delivery device |
WO2020068623A1 (en) | 2018-09-24 | 2020-04-02 | Amgen Inc. | Interventional dosing systems and methods |
WO2020068476A1 (en) | 2018-09-28 | 2020-04-02 | Amgen Inc. | Muscle wire escapement activation assembly for a drug delivery device |
WO2020072577A1 (en) | 2018-10-02 | 2020-04-09 | Amgen Inc. | Injection systems for drug delivery with internal force transmission |
WO2020072846A1 (en) | 2018-10-05 | 2020-04-09 | Amgen Inc. | Drug delivery device having dose indicator |
WO2020081480A1 (en) | 2018-10-15 | 2020-04-23 | Amgen Inc. | Platform assembly process for drug delivery device |
WO2020081479A1 (en) | 2018-10-15 | 2020-04-23 | Amgen Inc. | Drug delivery device having damping mechanism |
WO2020091981A1 (en) | 2018-11-01 | 2020-05-07 | Amgen Inc. | Drug delivery devices with partial drug delivery member retraction |
WO2020092056A1 (en) | 2018-11-01 | 2020-05-07 | Amgen Inc. | Drug delivery devices with partial needle retraction |
WO2020091956A1 (en) | 2018-11-01 | 2020-05-07 | Amgen Inc. | Drug delivery devices with partial drug delivery member retraction |
WO2020219482A1 (en) | 2019-04-24 | 2020-10-29 | Amgen Inc. | Syringe sterilization verification assemblies and methods |
WO2021041067A2 (en) | 2019-08-23 | 2021-03-04 | Amgen Inc. | Drug delivery device with configurable needle shield engagement components and related methods |
WO2022246055A1 (en) | 2021-05-21 | 2022-11-24 | Amgen Inc. | Method of optimizing a filling recipe for a drug container |
Also Published As
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US9175093B2 (en) | PCSK9 antagonists | |
US20200024366A1 (en) | ANTIBODIES WITH pH DEPENDENT ANTIGEN BINDING | |
AU2013200743B2 (en) | PCSK9 antagonists | |
AU2015200427B2 (en) | PCSK9 antagonists | |
AU2013204981A1 (en) | Antibodies with pH dependent antigen binding |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 200980145121.2 Country of ref document: CN |
|
WWE | Wipo information: entry into national phase |
Ref document number: 591541 Country of ref document: NZ |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2736349 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2009290438 Country of ref document: AU Ref document number: 12011500507 Country of ref document: PH |
|
ENP | Entry into the national phase |
Ref document number: 2011526617 Country of ref document: JP Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2011109178 Country of ref document: RU Ref document number: 1816/DELNP/2011 Country of ref document: IN Ref document number: 000618-2011 Country of ref document: PE Ref document number: MX/A/2011/002726 Country of ref document: MX |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 11037695 Country of ref document: CO |
|
ENP | Entry into the national phase |
Ref document number: 2009290438 Country of ref document: AU Date of ref document: 20090911 Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 20117008305 Country of ref document: KR Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2009808999 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 002084-2015 Country of ref document: PE |
|
ENP | Entry into the national phase |
Ref document number: PI0919289 Country of ref document: BR Kind code of ref document: A2 Effective date: 20110311 |