EP3013422A1 - Procédés de traitement d'une hypercholestérolémie familiale homozygote - Google Patents

Procédés de traitement d'une hypercholestérolémie familiale homozygote

Info

Publication number
EP3013422A1
EP3013422A1 EP13737090.4A EP13737090A EP3013422A1 EP 3013422 A1 EP3013422 A1 EP 3013422A1 EP 13737090 A EP13737090 A EP 13737090A EP 3013422 A1 EP3013422 A1 EP 3013422A1
Authority
EP
European Patent Office
Prior art keywords
seq
sequence
amino acid
pcsk9
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP13737090.4A
Other languages
German (de)
English (en)
Inventor
Scott Wasserman
Robert Andrew Donald Scott
Evan A. Stein
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Publication of EP3013422A1 publication Critical patent/EP3013422A1/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to methods for treatment of homozygous familial hypercholesterolemia using antigen binding proteins, including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9).
  • antigen binding proteins including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9).
  • LDL low density lipoprotein
  • PCSK9 proprotein convertase subtilisin/kexin 9
  • hypercholesterolemia new insights in pathogenesis and treatment. J Clin Invest 2003; 1 1 1 : 1795-803). While true genetic homozygous familial hypercholesterolemia is not uncommon, the majority of patients are compound heterozygotes (Usifo E, Leigh SE, Whittall RA, et al. Low-density lipoprotein receptor gene familial
  • hypercholesterolemia variant database update and pathological assessment. Ann Hum Genet 2012;76:387-401).
  • the residual LDL receptor activity is associated with severity of LDL cholesterol elevation and propensity for earlier cardiovascular disease (Goldstein JL, Hobbs HH, Brown MS, eds. Familial hypercholesterolemia. 8th Edition ed: McGraw- Hill; 2001).
  • mipomersen which both reduce hepatic lipoprotein production, have been approved solely for the treatment of homozygous familial hypercholesterolemia. Even with the introduction of these two new drugs, there remains a need to identify new methods for treating patients diagnosed with homozygous familial hypercholesterolemia.
  • the invention provided comprises a method of lowering serum LDL cholesterol in a patient diagnosed with homozygous familial hypercholesterolemia comprising administering at least one anti-PC SK9 antibody to the patient in need thereof at a dose of about 120 mg to about 3000 mg, thereby lowering said serum LDL cholesterol level by at least about 10%, as compared to a predose level of serum LDL cholesterol in the patient.
  • the serum LDL cholesterol level of said patient is lowered by at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%), at least about 50%>, at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%, at least about 75%, at least about 80%, at least about 85%, or at least about 90% as compared to a predose level of serum LDL cholesterol in the patient.
  • the anti-PCSK9 antibody is administered to a patient diagnosed with homozygous familial hypercholesterolemia at a dose of about 140 mg to about 3000 mg, of about 140 mg to about 2800 mg, of about 140 mg to about 2500 mg, of about 140 mg to about 2000 mg, of about 140 mg to about 1800 mg, of about 140 mg to about 1400 mg, of about 120 mg to about 1200 mg, of about 120 mg to about 1000 mg, of about 120 mg to about 700 mg, of about 140 mg to about 700 mg, of about 140 mg to about 600 mg, of about 140 mg to about 450 mg, of about 120 mg to about 450 mg, of about 120 mg to about 450 mg, of about 140 mg to about 450 mg, of about 210 mg to about 450 mg, or of about 280 mg to about 450 mg, of about 210 mg to about 420 mg, of about 280 mg to about 420 mg, of about 420 mg to about 3000 mg, of about 700 mg to about 3000 mg, of about 1000 mg of about 140 mg to about 3000 mg,
  • the anti-PCSK9 antibody is administered to a patient at a dose of about 35 mg, of about 45 mg, of about 70 mg, of about 105 mg, of about 120 mg of about 140 mg, of about 150 mg, of about 160 mg, of about 170 mg, of about 180 mg, of about 190 mg, of about 200 mg, of about 210 mg, of about 280 mg, of about 360 mg, of about 420 mg, of about 450 mg, of about 600 mg, of about 700 mg, of about 1200 mg, of about 1400 mg, of about 1800 mg, of about 2000 mg, of about 2500 mg, of about 2800 mg, or about 3000 mg.
  • the anti-PC SK9 antibody is administered to a patient on a schedule selected from the group consisting of: (1) once a week, (2) once every two weeks, (3) once a month, (4) once every other month, (5) once every three months (6)once every six months and (7) once every twelve months.
  • the ant-PCSK9 antibody is administered parenterally.
  • the anti-PC SK9 antibody is administered intravenously.
  • the anti-PCSK9 antibody is administered subcutaneously.
  • the anti-PCSK9 antibody comprises: A) one or more heavy chain complementary determining regions (CDRHs) selected from the group consisting of: (i) a CDRH1 from a CDRH1 in a sequence selected from the group consisting of SEQ ID NO: 49, 67, 459, 463 and 483; (ii) a CDRH2 from a CDRH2 in a sequence selected from the group consisting of SEQ ID NO: 49, 67, 459, 463 and 483; (iii) a CDRH3 from a CDRH3 in a sequence selected from the group consisting of SEQ ID NO: 49, 67, 459, 463 and 483; and (iv) a CDRH of (i), (ii), and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 4 amino acids; B) one or more light chain complementary determining regions (CDRLs) selected from the group consisting of: CDRLs (CDRLs
  • the isolated antigen binding protein comprises at least one CDRH of A) and at least one CDRL of B). In some embodiments, the isolated antigen binding protein comprises at least two CDRH of A) and at least two CDRL of B). In some embodiments, the isolated antigen binding protein comprises at least three CDRH of A) and at least three CDRL of B).
  • the isolated antigen binding protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:23, a CDRL2 of the CDRL2 sequence in SEQ ID NO:23, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:23, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 49, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 49, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 49.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:465, a CDRL2 of the CDRL2 sequence in SEQ ID NO:465, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:465, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 463, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 463, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:463.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO: 12, a CDRL2 of the CDRL2 sequence in SEQ ID NO: 12, and a CDRL3 of the CDRL3 sequence in SEQ ID NO: 12, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 67, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 67, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:67.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complimentarity region (CDR) of the CDRL1 sequence in SEQ ID NO:461, a CDRL2 of the CDRL2 sequence in SEQ ID NO:461, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:461, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 459, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 459, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:459.
  • CDR light chain complimentarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:485, a CDRL2 of the CDRL2 sequence in SEQ ID NO:485, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:485, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 483, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 483, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:483.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:582, a CDRL2 of the CDRL2 sequence in SEQ ID NO:582, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:582, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 583, a CDRLL? of the CDRH2 sequence in SEQ ID NO: 583, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:583.
  • CDR light chain complementarity region
  • the anti-PCSK9 antibody comprises: a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 23 and a heavy chain variable region that comprises and amino acid sequence that is at least 90% identical to that of SEQ ID NO:49; a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 12 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:67; a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 461 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:459; a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:465 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:463; a light chain variable region that comprises
  • the anti-PCSK9 antibody comprises: a light chain variable region that comprises an amino acid sequence, SEQ ID NO: 23, and a heavy chain variable region that comprises and amino acid sequence, SEQ ID NO:49; a light chain variable region that comprises an amino acid sequence, SEQ ID NO: 12, and a heavy chain variable region that comprises an amino acid sequence, SEQ ID NO:67; a light chain variable region that comprises amino acid sequence SEQ ID NO: 461 and a heavy chain variable region that comprises amino acid sequence SEQ ID NO:459; a light chain variable region that comprises the amino acid sequence of SEQ ID NO:465 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:463; a light chain variable region that comprises the amino acid sequence of SEQ ID NO: 485 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:483; a light chain variable region that comprises an amino acid sequence, SEQ ID NO: 582, and a heavy chain variable region that comprises and amino acid sequence
  • the invention comprises a method of treating a patient diagnosed with homozygous familial hypercholesterolemia comprising administering at least one anti-PCSK9 antibody to the patient in need thereof at a dose of about 120 mg to about 3000 mg, thereby treating the homozygous familial hypercholesterolemia in said patient.
  • the serum LDL cholesterol level of said patient diagnosed with homozygous familial hypercholesterolemia is lowered by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%), at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%>, at least about 75%>, at least about 80%>, at least about 85%>, or at least about 90%> as compared to a predose level of serum LDL cholesterol in said patient.
  • the anti-PCSK9 antibody comprises: A) one or more heavy chain complementary determining regions (CDRHs) selected from the group consisting of: (i) a CDRH1 from a CDRH1 in a sequence selected from the group consisting of SEQ ID NO: 49, 67, 459, 463 and 483; (ii) a CDRH2 from a CDRH2 in a sequence selected from the group consisting of SEQ ID NO: 49, 67, 459, 463 and 483; (iii) a CDRH3 from a CDRH3 in a sequence selected from the group consisting of SEQ ID NO: 49, 67, 459, 463 and 483; and (iv) a CDRH of (i), (ii), and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 4 amino acids; B) one or more light chain complementary determining regions (CDRLs) selected from the group consisting of: CDRLs (CDRLs
  • the isolated antigen binding protein comprises at least one CDRH of A) and at least one CDRL of B). In some embodiments, the isolated antigen binding protein comprises at least two CDRH of A) and at least two CDRL of B). In some embodiments, the isolated antigen binding protein comprises at least three CDRH of A) and at least three CDRL of B).
  • the isolated antigen binding protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:23, a CDRL2 of the CDRL2 sequence in SEQ ID NO:23, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:23, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 49, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 49, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 49.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:465, a CDRL2 of the CDRL2 sequence in SEQ ID NO:465, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:465, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 463, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 463, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:463.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO: 12, a CDRL2 of the CDRL2 sequence in SEQ ID NO: 12, and a CDRL3 of the CDRL3 sequence in SEQ ID NO: 12, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 67, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 67, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:67.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complimentarity region (CDR) of the CDRLl sequence in SEQ ID NO:461, a CDRL2 of the CDRL2 sequence in SEQ ID NO:461, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:461, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 459, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 459, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:459.
  • CDR light chain complimentarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:485, a CDRL2 of the CDRL2 sequence in SEQ ID NO:485, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:485, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 483, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 483, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:483.
  • CDR light chain complementarity region
  • the isolated antigen bindng protein comprises a light chain complementarity region (CDR) of the CDRLl sequence in SEQ ID NO:582, a CDRL2 of the CDRL2 sequence in SEQ ID NO:582, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:582, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 583, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 583, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:583.
  • CDR light chain complementarity region
  • the anti-PCSK9 antibody comprises: a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 23 and a heavy chain variable region that comprises and amino acid sequence that is at least 90% identical to that of SEQ ID NO:49; a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 12 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:67; a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 461 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:459; a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:465 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:463; a light chain variable region that comprises an amino acid sequence that is at least 90%
  • the anti-PCSK9 antibody comprises: a light chain variable region that comprises an amino acid sequence, SEQ ID NO: 23, and a heavy chain variable region that comprises and amino acid sequence, SEQ ID NO:49; a light chain variable region that comprises an amino acid sequence, SEQ ID NO: 12, and a heavy chain variable region that comprises an amino acid sequence, SEQ ID NO:67; a light chain variable region that comprises amino acid sequence SEQ ID NO: 461 and a heavy chain variable region that comprises amino acid sequence SEQ ID NO:459; a light chain variable region that comprises the amino acid sequence of SEQ ID NO:465 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:463; a light chain variable region that comprises the amino acid sequence of SEQ ID NO: 485 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:483; a light chain variable region that comprises an amino acid sequence, SEQ ID NO: 582, and a heavy chain variable region that comprises and amino acid sequence
  • the anti-PCSK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:23, a CDRL2 of the CDRL2 sequence in SEQ ID NO:23, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:23, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 49, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 49, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 49.
  • CDR light chain complementarity region
  • the anti-PCSK9 antibody comprises a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:23 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:49. In some embodiments the anti-PCSK9 antibody comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:23 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:49. In some embodiments the anti-PCSK9 antibody comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:591and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:590. In some embodiments, the anti-PCSK9 antibody is 21B12.
  • the anti-PCSK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:23, a CDRL2 of the CDRL2 sequence in SEQ ID NO:23, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:23, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 49, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 49, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 49, or an amino acid sequence that is at least 90% identical to that of SEQ ID NO:23 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:23 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of SEQ
  • the anti-PC SK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:23, a CDRL2 of the CDRL2 sequence in SEQ ID NO:23, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:23, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 49, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 49, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 49, or comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:23 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:23 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of
  • the anti-PC SK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:23, a CDRL2 of the CDRL2 sequence in SEQ ID NO:23, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:23, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 49, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 49, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 49, or comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:23 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:23 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:49 or the antibody is 2 IB 12, the anti-
  • the anti-PC SK9 antibody comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:23 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:23 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:49, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:591 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:590, or comprises the antibody is 21B12
  • the anti-PCSK9 antibody is administered to a patient at a dose of about 420 mg to about 3000 mg intraveneously every week, wherein the serum LDL cholesterol level of the patient is lowered 30-50% for about 7- 10 days; is administered to a patient at a dose of about 700 mg intraveneously every week, wherein the serum LDL cholesterol level of the patient is lowered 30-50%
  • the anti-PC SK9 antibody is 8 A3, 11F1 and 8A1.
  • the anti-PC SK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:465, a CDRL2 of the CDRL2 sequence in SEQ ID NO:465, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:465, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 463, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 463, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:463.
  • CDR light chain complementarity region
  • the anti-PCSK9 antibody comprises a light chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:465 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:463.
  • the anti- PCSK9 antibody comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:465 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:463.
  • the anti-PCSK9 antibody is 1 IF 1.
  • the anti-PC SK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:465, a CDRL2 of the CDRL2 sequence in SEQ ID NO:465, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:465, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 463, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 463, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:463, or comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:465 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:463, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:465 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:463, or comprises the antibody
  • the anti-PCSK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:465, a CDRL2 of the CDRL2 sequence in SEQ ID NO:465, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:465, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 463, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 463, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:463.
  • CDR light chain complementarity region
  • the anti-PCSK9 antibody comprises, or comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:465 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 463, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:465 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:463 or comprises the antibody is 11F1
  • the anti-PCSK9 antibody is administered to a patient at a dose of about 150 mg subcutaneous ly once a week wherein the serum LDL cholesterol level of the patient is lowered at least about 30-50%> for about 7-10 days, is administered to a patient at a dose of about 150 mg subcutaneous ly once every other week wherein the serum LDL cholesterol level of the patient is lowered at least about 30-50% for about 10-14 days; is administered to a patient at a dose of about 150 mg subcutaneously once every four weeks wherein the serum LDL cholesterol level of the patent is
  • the anti-PCSK9 antibody comprises a light chain complementarity region (CDR) of the CDRL1 sequence in SEQ ID NO:465, a CDRL2 of the CDRL2 sequence in SEQ ID NO:465, and a CDRL3 of the CDRL3 sequence in SEQ ID NO:465, and a heavy chain complementarity determining region (CDR) of the CDRH1 sequence in SEQ ID NO: 463, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 463, and a CDRH3 of the CDRH3 sequence in SEQ ID NO:463.
  • CDR light chain complementarity region
  • the anti-PCSK9 antibody comprises, or comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO:465 and a heavy chain variable region that comprises an amino acid sequence that is at least 90% identical to that of SEQ ID NO: 463, or comprises a light chain variable region that comprises the amino acid sequence of SEQ ID NO:465 and a heavy chain variable region that comprises the amino acid sequence of SEQ ID NO:463, or the antibody is 11F1
  • the anti-PCSK9 antibody is administered to a patient the anti-PCSK9 antibody is administered to a patient at a dose of about 420 mg to about 3000 mg intraveneously every week, wherein the serum LDL cholesterol level of the patient is lowered 30-50%> for about 7-10 days; is administered to a patient at a dose of about 700 mg intraveneously every week, wherein the serum LDL cholesterol level of the patient is lowered 30-50%> for about 7-10 days; is administered to a patient at a dose of about 1200 mg intraveneously every week, where
  • the at least one anti-PC SK9 antibody is administered to the patient before, after or concurrent with at least one other cholesterol-lowering agent.
  • Cholesterol lowering agents include statins, including, atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin, nicotinic acid (niacin), slow relese niacin (SLO-NIACIN), laropiprant (CORDAPTIVE), fibric acid (LOPID (Gemfibrozil), TRICOR (fenofibrate)), Bile acid sequestrants, sucha as cholestyramine (QUESTRAN), colesvelam (WELCHOL), COLESTID (colestipol)), cholesterol absorption inhibitor (ZETIA (ezetimibe)), lipid modifying agents, PPAR gamma agonsits, PPAR alpha/
  • the invention comprises a method of lowering the serum LDL cholesterol level in a patient diagnosed with homozygous familial hypercholesterolemia.
  • the method comprises administering to a patient diagnosed with homozygous familial hypercholesterolemia a dose of about 120 mg to about 3000 mg of at least one anti-PC SK9 antibody described herein.
  • the dose is about 120 mg to about 450 mg of at least one anti-PCSK9 antibody administered once weekly (QW).
  • the dose is about 140 mg to about 450 mg of at least one anti-PCSK9 antibody administered once weekly.
  • the dose is about 280 mg to about 450 mg of at least one anti-PCSK9 antibody administered once weekly.
  • the dose is aboutl20 mg to about 450 mg of at least one anti-PCSK9 antibody administered once every 2 weeks (Q2W). In some embodiments, the dose is about 140 mg to about 450 mg of at least one anti-PCSK9 antibody administered once every 2 weeks (Q2W). In some embodiments, the dose is about 280 mg to about 420 mg of at least one anti-PCSK9 antibody administered once every 2 weeks (Q2W). In some embodiments, the dose is about 400 mg to about 450 mg of at least one anti-PCSK9 antibody administered once every 2 weeks (Q2W). In some embodiments, the dose is about 420 mg of at least one anti-PCSK9 antibody administered once every 2 weeks (Q2W).
  • the dose is about 250 mg to about 480 mg of at least one anti-PCSK9 antibody administered once every 4 weeks (Q4W). In some embodiments, the dose is about 280 mg to about 420 mg of at least one anti-PCSK9 antibody administered once every 4 weeks (Q4W). In some embodiments, the dose is about 350 mg to about 420 mg of at least one anti-PCSK9 antibody administered once every 4 weeks (Q4W). In some embodiments, the dose is about 420 mg to about 3000 mg of at least one anti-PCSK9 antibody administered once every week (QW). In some embodiments, the dose is about 1000 mg to about 3000 mg of at least one anti-PC SK9 antibody administered once every week (QW).
  • the dose is about 2000 mg to about 3000 mg of at least one anti-PC SK9 antibody administered once every week (QW). In some embodiments, the dose is about 420 mg to about 3000 mg of at least one anti- PCSK9 antibody administered once every other week (Q2W). In some embodiments, the dose is about 1000 mg to about 3000 mg of at least one anti-PCSK9 antibody administered once every other week (Q2W). In some embodiments, the dose is about 2000 mg to about 3000 mg of at least one anti-PC SK9 antibody administered once every other week (Q2W). In some embodiments, the dose is about 420 mg to about 3000 mg of at least one anti-PCSK9 antibody administered once every month (Q4W).
  • the dose is about 1000 mg to about 3000 mg of at least one anti- PCSK9 antibody administered once every month (Q4W). In some embodiments, the dose is about 2000 mg to about 3000 mg of at least one anti-PCSK9 antibody administered once every month (Q4W).
  • the serum LDL cholesterol level is reduced by at least about 10% as compared to a predose serum LDL cholesterol level. In some embodiments, the serum LDL cholesterol level is reduced by at least about 15%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 20%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 25%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 30%>.
  • the serum LDL cholesterol level is reduced by at least about 35%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 40%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 45%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 50%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 55%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 60%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 75%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 70%). In some embodiments, the serum LDL cholesterol level is reduced by at least about 75%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 80%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 85%. %. In some embodiments, the serum LDL cholesterol level is reduced by at least about 90%>.
  • the invention comprises a method of lowering the serum LDL cholesterol level in a patient diagnosed with homozygous familial hypercholesterolemia, the method comprising administering to a patient in need thereof, a dose of at least one anti-PCSK9 antibody, and wherein the dose of anti- PCSK9 antibody is administered on a schedule selected from the group consisting of: (1) at least about 120 mg every week (QW); (2) at least an amount of about 140 mg every week (QW); (3) at least an amount of about 120 mg every two weeks or every other week (Q2W); (4) at least an amount of about 140 mg every two weeks or every other week (Q2W); (5) at least an amount of about 150 mg every two weeks or every other week (Q2W) (6) at least an amount of about 280 mg every two weeks or every other week (Q2W); (7) at least an amount of about 350 mg every two weeks or every other week (Q2W); (8) at least an amount of about 420 mg every two weeks or every other week (Q2W); and (9) at
  • the serum LDL cholesterol level is reduced by at least about 10%> as compared to a predose serum LDL cholesterol level. In some embodiments, the serum LDL cholesterol level is reduced by at least about 10%> as compared to a predose serum LDL cholesterol level. In some embodiments, the serum LDL cholesterol level is reduced by at least about 15%). In some embodiments, the serum LDL cholesterol level is reduced by at least about 20%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 25%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 30%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 35%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 40%.
  • the serum LDL cholesterol level is reduced by at least about 45%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 50%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 55%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 60%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 65%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 70%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 75%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 80%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 85%). In some embodiments, the serum LDL cholesterol level is reduced by at least about 90%.
  • the invention comprises a method of lowering the serum PCSK9 level in a patient diagnosed with homozygous familial hypercholesterolema, the method comprising administering to a patient in need thereof, a dose of at least one anti-PC SK9 antibody, and wherein the dose of anti-PC SK9 antibody is administered on a schedule selected from the group consisting of: 1) at least about 120 mg every week (QW); (2) at least an amount of about 140 mg every week (QW); (3) at least an amount of about 120 mg every two weeks or every other week (Q2W); (4) at least an amount of about 140 mg every two weeks or every other week (Q2W); (5) at least an amount of about 150 mg every two weeks or every other week (Q2W) (6) at least an amount of about 280 mg every two weeks or every other week (Q2W); (7) at least an amount of about 350 mg every two weeks or every other week (Q2W); (8) at least an amount of about 420 mg every two weeks or every other week (Q2W);
  • the serum PCSK9 value is reduced by at least about 20% as compared to a predose serum PCSK9 level. In some embodiments, the serum PCSK9 value is reduced by at least about 30%. In some embodiments, the serum PCSK9 value is reduced by at least about 40%. In some embodiments, the serum PCSK9 value is reduced by at least about 50%. In some embodiments, the serum PCSK9 value is reduced by at least about 60%. In some embodiments, the serum PCSK9 value is reduced by at least about 65%. In some embodiments, the serum PCSK9 value is reduced by at least about 70%. In some embodiments, the serum PCSK9 value is reduced by at least about 75%. In some embodiments, the serum PCSK9 value is reduced by at least about 80%. In some embodiments, the serum PCSK9 value is reduced by at least about 85%. In some embodiments, the serum PCSK9 value is reduced by at least about 90%>.
  • the invention comprises a method of lowering the total cholesterol level in a patient diagnosed with homozygous familial hypercholesterolemia, the method comprising administering to a patient in need thereof, a dose of at least one anti-PCSK9 antibody, and wherein the dose of anti- PCSK9 antibody is administered on a schedule selected from the group consisting of: (1) ) at least about 120 mg every week (QW); (2) at least an amount of about 140 mg every week (QW); (3) at least an amount of about 120 mg every two weeks or every other week (Q2W); (4) at least an amount of about 140 mg every two weeks or every other week (Q2W); (5) at least an amount of about 150 mg every two weeks or every other week (Q2W) (6) at least an amount of about 280 mg every two weeks or every other week (Q2W); (7) at least an amount of about 350 mg every two weeks or every other week (Q2W); (8) at least an amount of about 420 mg every two weeks or every other week (Q2W); and (9) at
  • the total cholesterol level is reduced by at least about 20% as compared to a predose total cholesterol level. In some embodiments, the total cholesterol level is reduced by at least about 25%. In some embodiments, the total cholesterol level is reduced by at least about 30%). In some embodiments, the total cholesterol level is reduced by at least about 35%o. In some embodiments, the total cholesterol level is reduced by at least about 40%). In some embodiments, the total cholesterol level is reduced by at least about 45%o. In some embodiments, the total cholesterol level is reduced by at least about 50%). In some embodiments, the total cholesterol level is reduced by at least about 55%o. In some embodiments, the total cholesterol level is reduced by at least about 60%.
  • the invention comprises a method of lowering the non-HDL cholesterol level in a patient diagnosed with homozygous familial hypercholesterolemia, the method comprising administering to a patient in need thereof, a dose of at least one anti-PCSK9 antibody, and wherein the dose of anti- PCSK9 antibody is administered on a schedule selected from the group consisting of: (1) at least about 120 mg every week (QW); (2) at least an amount of about 140 mg every week (QW); (3) at least an amount of about 120 mg every two weeks or every other week (Q2W); (4) at least an amount of about 140 mg every two weeks or every other week (Q2W); (5) at least an amount of about 150 mg every two weeks or every other week (Q2W) (6) at least an amount of about 280 mg every two weeks or every other week (Q2W); (7) at least an amount of about 350 mg every two weeks or every other week (Q2W); (8) at least an amount of about 420 mg every two weeks or every other week (Q2W); and (9)
  • the non- HDL cholesterol level is reduced by at least about 30% as compared to a predose non- HDL cholesterol level. In some embodiments, the non-HDL cholesterol level is reduced by at least about 35%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 40%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 45%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 50%>. In some embodiments, the non- HDL cholesterol level is reduced by at least about 55%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 60%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 65%.
  • the non-HDL cholesterol level is reduced by at least about 70%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 75%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 80%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 85%.
  • the invention comprises a method of lowering ApoB levels in a patient diagnosed with homozygous familial hypercholesterolemia, the method comprising administering to a patient in need thereof, a dose of at least one anti-PCSK9 antibody, and wherein the dose of anti-PCSK9 antibody is administered on a schedule selected from the group consisting of: (1) at least about 120 mg every week (QW); (2) at least an amount of about 140 mg every week (QW); (3) at least an amount of about 120 mg every two weeks or every other week (Q2W); (4) at least an amount of about 140 mg every two weeks or every other week (Q2W); (5) at least an amount of about 150 mg every two weeks or every other week (Q2W) (6) at least an amount of about 280 mg every two weeks or every other week (Q2W); (7) at least an amount of about 350 mg every two weeks or every other week (Q2W); (8) at least an amount of about 420 mg every two weeks or every other week (Q2W); and (9) at least an
  • the ApoB level is reduced by at least about 10% as compared to a predose ApoB level. In some embodiments, the ApoB level is reduced by at least about 15%. In some embodiments, the ApoB level is reduced by at least about 20%>. In some embodiments, the ApoB level is reduced by at least about 25%. In some embodiments, the ApoB level is reduced by at least about 30%. In some embodiments, the ApoB level is reduced by at least about 35%. In some embodiments, the ApoB level is reduced by at least about 40%. In some embodiments, the ApoB level is reduced by at least about 45%. In some embodiments, the ApoB level is reduced by at least about 50%.
  • the ApoB level is reduced by at least about 55%. In some embodiments, the ApoB level is reduced by at least about 60%. In some embodiments, the ApoB level is reduced by at least about 65%. In some embodiments, the ApoB level is reduced by at least about 70%. In some embodiments, the ApoB level is reduced by at least about 75%.
  • the invention comprises a method of lowering Lipoprotein A ("Lp(a)”) levels in a patient diagnosed with homozygous familial hypercholesterolemia, the method comprising administering to a patient in need thereof, a dose of at least one anti-PC SK9 antibody, and wherein the dose of anti-PC SK9 antibody is administered on a schedule selected from the group consisting of: (1) at least about 120 mg every week (QW); (2) at least an amount of about 140 mg every week (QW); (3) at least an amount of about 120 mg every two weeks or every other week (Q2W); (4) at least an amount of about 140 mg every two weeks or every other week (Q2W); (5) at least an amount of about 150 mg every two weeks or every other week (Q2W) (6) at least an amount of about 280 mg every two weeks or every other week (Q2W); (7) at least an amount of about 350 mg every two weeks or every other week (Q2W); (8) at least an amount of about 420 mg every two weeks or every other week (
  • the Lp(a) level is reduced by at least about 10% as compared to a predose Lp(a) level. In some embodiments, the Lp(a) level is reduced by at least about 15%. In some embodiments, the Lp(a) level is reduced by at least about 20%). In some embodiments, the Lp(a) level is reduced by at least about 25%. In some embodiments, the Lp(a) level is reduced by at least about 30%>. In some embodiments, the Lp(a) level is reduced by at least about 35%. In some embodiments, the Lp(a) level is reduced by at least about 40%>. In some embodiments, the Lp(a) level is reduced by at least about 45%.
  • the Lp(a) level is reduced by at least about 50%>. In some embodiments, the Lp(a) level is reduced by at least about 55%). In some embodiments, the Lp(a) level is reduced by at least about 60%>. In some embodiments, the Lp(a) level is reduced by at least about 65%>.
  • the invention comprises a method for treating a patient diagnosed with homozyougous familial hypercholesterolemia, the method comprising administering to a patient diagnosed with homozygous familial hypercholesterolemia a dose of about 120 mg to about 3000 mg of at least one anti-PCSK9 antibody described herein.
  • the dose is about 120 mg to about 450 mg of at least one anti- PCSK9 antibody administered once weekly (QW).
  • the dose is about 140 mg to about 450 mg of at least one anti-PCSK9 antibody administered once weekly.
  • the dose is about 120 mg to about 450 mg of at least one anti-PCSK9 antibody administered once every two weeks (Q2W).
  • the dose is about 140 mg to about 420 mg of at least one anti-PCSK9 antibody administered once every two weeks (Q2W). In some embodiments, the dose is about 105 mg to about 350 mg of at least one anti-PC SK9 antibody administered once every two weeks (Q2W). In some embodiments, the dose is about 140 mg to about 280 mg of at least one anti-PCSK9 antibody administered once every two weeks (Q2W). In some embodiments, the dose is about 150 mg to about 280 mg of at least one anti-PCSK9 antibody administered once every two weeks (Q2W). In some embodiments, the dose is about 150 mg to about 200 mg of at least one anti-PCSK9 antibody administered once every two weeks (Q2W).
  • the dose is about 400 mg to about 450 mg of at least one anti-PC SK9 antibody administered once every two weeks (Q2W). In some embodiments, the dose is about 420 mg of at least one anti-PC SK9 antibody administered once every two weeks (Q2W). In some embodiments, the dose is about 120 mg to about 480 mg of at least one anti-PCSK9 antibody administered once every four weeks (Q4W). In some embodiments, the dose is about 150 mg to about 420 mg of at least one anti-PC SK9 antibody administered once every four weeks (Q4W). In some embodiments, the dose is about 400 mg to about 450 mg of at least one anti-PCSK9 antibody administered once every four weeks (Q4W).
  • the dose is about 250 mg to about 480 mg of at least one anti-PCSK9 antibody administered once every four weeks (Q4W). In some embodiments, the dose is about 280 mg to about 420 mg of at least one anti-PCSK9 antibody administered once every four weeks (Q4W). In some embodiments, the dose is about 350 mg to about 420 mg of at least one anti-PC SK9 antibody administered once every four weeks. In some embodiments, the dose is about 1000 mg every four weeks (Q4W). In some embodiments, the dose is about about 2000 mg every four weeks (Q4W). In some embodiments, the dose is about 3000 mg every four weeks (Q4W).
  • the serum LDL cholesterol level is reduced by at least about 10% as compared to a predose serum LDL cholesterol level. In some embodiments, the serum LDL cholesterol level is reduced by at least about 15%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 20%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 25%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 30%). In some embodiments, the serum LDL cholesterol level is reduced by at least about 35%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 40%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 45%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 50%.
  • the serum LDL cholesterol level is reduced by at least about 55%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 60%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 65%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 70%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 75%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 80%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 85%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 90%.
  • the anti-PCSK9 antibody is 21B12, 26H5, 31H4, 8A3, 1 1F1 and/or 8A1.
  • FIG. 1 A depicts an amino acid sequence of the mature form of the PCSK9 with the pro-domain underlined.
  • FIGs. IB 1 -IB 4 depict amino acid and nucleic acid sequences of PCSK9 with the pro-domain underlined and the signal sequence in bold.
  • FIGs. 2A-2D are sequence comparison tables of various light chains of various antigen binding proteins.
  • FIG. 2C continues the sequence started in FIG. 2A.
  • FIG. 2D continues the sequence started on FIG. 2B.
  • FIGs. 3A-3D are sequence comparison tables of various heavy chains of various antigen binding proteins.
  • FIG. 3C continues the sequence started in FIG. 3 A.
  • FIG. 3D continues the sequence started on FIG. 3B.
  • FIGs. 3E-3JJ depict the amino acid and nucleic acid sequences for the variable domains of some embodiments of the antigen binding proteins.
  • FIG. 3K depicts the amino acid sequences for various constant domains.
  • FIGs. 3LL-3BBB depict the amino acid and nucleic acid sequences for the variable domains of some embodiments of the antigen binding proteins.
  • FIGs. 3CCC-3JJJ are sequence comparison tables of various heavy and light chains of some embodiments of the antigen binding proteins.
  • FIG. 3LLL is a set of ABP sequences identifying various differences between the human ABP sequences and the ABP sequences that were raised in E. coli.. (U.S. P. 8,030,457).
  • FIG. 4A is a binding curve of an antigen binding protein to human PCSK9.
  • FIG. 4B is a binding curve of an antigen binding protein to human PCSK9.
  • FIG. 4C is a binding curve of an antigen binding protein to cynomolgus PCSK9.
  • FIG. 4D is a binding curve of an antigen binding protein to cynomolgus
  • FIG. 4E is a binding curve of an antigen binding protein to mouse PCSK9.
  • FIG. 4F is a binding curve of an antigen binding protein to mouse PCSK9.
  • FIG. 5A depicts the results of an SDS PAGE experiment involving PCSK9 and various antigen binding proteins demonstrating the relative purity and concentration of the proteins.
  • FIG. 5B and 5C depict graphs from Biacore solution equilibrium assays for
  • FIG. 5D depicts the graph of the kinetics from a Biacore capture assay.
  • FIG. 6 A is an inhibition curve of antigen binding protein 31H4 IgG2 to PCSK9 in an in vitro PCSK9:LDLR binding assay
  • FIG. 6B is an inhibition curve of antigen binding protein 31H4 IgG4 to PCSK9 in an in vitro PCSK9:LDLR binding assay.
  • FIG. 6C is an inhibition curve of antigen binding protein 2 IB 12 IgG2 to PCSK9 in an in vitro PCSK9:LDLR binding assay.
  • FIG. 6D is an inhibition curve of antigen binding protein 2 IB 12 IgG4 to
  • FIG. 7A is an inhibition curve of antigen binding protein 31H4 IgG2 in the cell LDL uptake assay showing the effect of the ABP to reduce the LDL uptake blocking effects of PCSK9
  • FIG. 7B is an inhibition curve of antigen binding protein 31H4 IgG4 in the cell
  • FIG. 7C is an inhibition curve of antigen binding protein 2 IB 12 IgG2 in the cell LDL uptake assay showing the effect of the ABP to reduce the LDL uptake blocking effects of PCSK9
  • FIG. 7D is an inhibition curve of antigen binding protein 2 IB 12 IgG4 in the cell LDL uptake assay showing the effect of the ABP to reduce the LDL uptake blocking effects of PCSK9
  • FIG. 8 A is a graph depicting the serum cholesterol lowering ability in mice of ABP 31H4, changes relative to the IgG control treated mice (* p ⁇ 0.01).
  • FIG. 8C is a graph depicting the effect of ABP 31H4 on HDL cholesterol levels in C57Bl/6 mice (* p ⁇ 0.01).
  • FIG. 8D is a graph depicting the effect of ABP 31H4 on HDL cholesterol levels in C57B1/6 mice (# p ⁇ 0.05).
  • FIG. 9 depicts a western blot analysis of the ability of ABP 31H4 to enhance the amount of liver LDLR protein present after various time points.
  • FIG. 10A is a graph depicting the ability of an antigen binding protein 31H4 to lower total serum cholesterol in wild type mice, relative.
  • FIG. 1 OB is a graph depicting the ability of an antigen binding protein 31H4 to lower HDL in wild type mice.
  • FIG. IOC is a graph depicting the serum cholesterol lowering ability of various antigen binding proteins 31H4 and 16F12.
  • FIG. 11A depicts an injection protocol for testing the duration and ability of antigen binding proteins to lower serum cholesterol.
  • FIG. 1 IB is a graph depicting the results of the protocol in FIG. 11 A.
  • FIG. 12A depicts LDLR levels in response to the combination of a statin and ABP 2 IB 12 in HepG2 cells.
  • FIG. 12B depicts LDLR levels in response to the combination of a statin and ABP 31H4 in HepG2 cells.
  • FIG. 12C depicts LDLR levels in response to the combination of a statin and ABP 25A7.1, a non-neutralizing antibody, (in contrast the "25 A7" a neutralizing antibody) in HepG2 cells.
  • FIG. 12D depicts LDLR levels in response to the combination of a statin and ABP 21B12 in HepG2 cells over expressing PCSK9.
  • FIG. 12E depicts LDLR levels in response to the combination of a statin and ABP 31H4 in HepG2 cells over expressing PCSK9.
  • FIG. 12F depicts LDLR levels in response to the combination of a statin and ABP 25A7.1, a non-neutralizing antibody, (in contrast the "25 A7" a neutralizing antibody) in HepG2 cells over expressing PCSK9.
  • FIG. 13A depicts the various light chain amino acid sequences of various ABPs to PCSK9.
  • the dots (.) indicate no amino acid.
  • FIG. 13B depicts a light chain cladogram for various ABPs to PCSK9.
  • FIG. 13C depicts the various heavy chain amino acid sequences of various ABPs to PCSK9.
  • the dots (.) indicate no amino acid.
  • FIG. 13D depicts a heavy chain dendrogram for various ABPs to PCSK9.
  • FIG. 13E depicts a comparison of light and heavy CDRs and designation of groups from which to derive consensus.
  • FIG. 13F depicts the consensus sequences for Groups 1 and 2.
  • FIG. 13G depicts the consensus sequences for Groups 3 and 4.
  • FIG. 13H depicts the consensus sequences for Groups 1 and 2. The dots (.) indicated identical residues.
  • FIG. 131 depicts the consensus sequences for Group 2.
  • FIG. 13 J depicts the consensus sequences for Groups 3 and 4. The dots (.) indicated identical residues.
  • FIG. 14 is a graph illustrating the binding specificity of 1 IF 1 in a competition assay with PCSKP, PCSK2, PCSK1 PCSK7 and Furin with OD 450 plotted on the vertical axis and concentration of PCSK9 (ug/ml) plotted on the horizontal axis.
  • FIG. 15 is a graph showing the dose response curve for inhibition of LDLR:D374Y PCSK9 binding by 11F1 in a competition assay with OD 450 plotted on the vertical axis and Log [11F1] (pM) plotted on the horizontal axis.
  • FIG. 16 is a graph depicting the dose response curve for the inhibition of LDLR: WT PCSK9 binding by 1 lFlin a competition assay with OD 4 so plotted on the vertical axis and Log [1 lfl] (pM) plotted on the horizontal axis.
  • FIG. 17 is a graph depicting the dose response curve for the ability of 11F1 to block human D374Y PCSK9-mediated reduction of LDL uptake in HepG2 cells with relative fluorescence units (xlO 4 ) plotted on the vertical axis and Log [11F1] (nM) plotted on the horizontal axis.
  • FIG. 18 is a graph depicting the dose response curve for the ability of 11F1 to block human WT PCSK9-mediated reduction of LDL uptake in HepG2 cells with relative fluorescence units plotted (xlO 4 ) on the vertical axis and Log [11F1] (nM) plotted on the horizontal axis.
  • FIG. 19 is a bar graph depicting the effect of 1 IF 1 and 8 A3 on serum non-HDL cholesterol in mice expressing human PCSK9 by AAV with non-HDL-C serum concentration (mg/ml) on the vertical axis and time following injection (days) plotted on the horizontal axis.
  • FIG. 20 is a bar graph depicting the effect of 1 IF 1 and 8A3 on Serum Total Cholesterol in mice expressing human PCSK9 by AAV with Serum Total Cholesterol (mg/ml) on the vertical axis and time following injection (days) plotted on the horizontal axis.
  • FIG. 21 is a bar graph depicting the effect of 1 IF 1 and 8 A3 on Serum HDL
  • HDL-C Cholesterol
  • FIG. 22 is a graph depicting IgG2, 8A3 and 1 IF 1 antibody concentration profiles in mice expressing human PCSK9 by AAV with serum antibody concentration (ng/mL) plotted on the vertical axis and time following injection in days plotted on the horizontal axis.
  • FIG. 23 is a table summarizing PK parameters for IgG2, 11F1 and 8A3 in mice expressing human PCSK9 by AAV.
  • FIG. 24 is a graph depicting the effect of a single subcutaneous administration of an ant-KLH antibody (control), 2 IB 12, 8 A3 and 1 IF 1 on serum LDL concentration (LDL-C) in cynomolgus monkeys with LDL-C (mg/dl) plotted on the vertical axis and time following administration in days on the horizontal axis.
  • FIG. 25 is a graph depicting the effect of a single subcutaneous administration of an ant-KLH antibody (control), 2 IB 12, 8 A3 and 11F1 on Serum Total Cholesterol in cynomolgus monkeys with Total Cholesterol concentration (mg/dl) plotted on the vertical axis and time following administration in days on the horizontal axis.
  • FIG. 26 is a graph depicting the effect of a single subcutaneous administration of an ant-KLH antibody (control), 2 IB 12, 8 A3 and 1 IF 1 on Serum HDL Cholesterol in cynomolgus monkeys with HDL-C (mg/dl) plotted on the vertical axis and time following administration in days on the horizontal axis.
  • FIG. 27 is a graph depicting the effect of a single subcutaneous administration of an ant-KLH antibody (control), 2 IB 12, 8 A3 and 1 IF 1 on Serum Triglycerides in cynomolgus monkeys with Serum Triglyceride concentration (mg/dl) plotted on the vertical axis and time following administration in days on the horizontal axis.
  • FIG. 28 is a graph depicting the effect of a single subcutaneous administration of an ant-KLH antibody (control), 2 IB 12, 8 A3 and 1 IF 1 on Apolipoprotein B (ApoB) in cynomolgus monkeys with APOB concentration (mg/dl) plotted on the vertical axis and time following administration in days on the horizontal axis.
  • FIG. 29 is a graph depicting the mean pharmacokinetic profiles for the anti— KLH antibody (control), 2 IB 12, 8 A3 and 1 IF 1 in cynomolgus monkeys with antibody concentrations (ng/ml) plotted on the vertical axis and time following administration in days on the horizontal axis.
  • FIG. 30 is a table summarizing PK parameters for the anti— KLH antibody (control), 21B12, 8A3 and 1 IF 1 in cynomolgus monkeys.
  • FIG. 31A depicts a comparison of light chain amino acid sequences of 8A1, 8 A3 and 1 IF 1, as well as a consensus sequence derived from the the comparison. CDR sequences are underlined.
  • FIG. 3 IB depicts a comparison of heavy chain amino acid sequences of 8A1, 8 A3 and 1 IF 1, as well as a consensus sequence derived from the the comparison. CDR sequences are underlined.
  • a method of treating a patient diagnosed with homozygous familial hypercholesterolemia comprises administering at least one antigen binding protein, including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9) to the patient.
  • at least one antigen binding protein including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9)
  • methods of lowering serum LDL cholesterol in a patient diagnosed with homozygous familial hypercholesterolemia using antigen binding proteins, including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9) are provided herein.
  • proprotein convertase subtilisin kexin type 9 refers to a polypeptide as set forth in SEQ ID NO: 1 and/or 3 or fragments thereof, as well as related polypeptides, which include, but are not limited to, allelic variants, splice variants, derivative variants, substitution variants, deletion variants, and/or insertion variants including the addition of an N-terminal methionine, fusion polypeptides, and interspecies homologs.
  • a PCSK9 polypeptide includes terminal residues, such as, but not limited to, leader sequence residues, targeting residues, amino terminal methionine residues, lysine residues, tag residues and/or fusion protein residues.
  • PCSK9 has also been referred to as FH3, NARC1, HCHOLA3, proprotein convertase subtilisin/kexin type 9, and neural apoptosis regulated convertase 1.
  • the PCSK9 gene encodes a proprotein convertase protein that belongs to the proteinase K subfamily of the secretory subtilase family.
  • PCSK9 denotes both the proprotein and the product generated following autocatalysis of the proprotein.
  • PCSK9 When only the autocatalyzed product is being referred to (such as for an antigen binding protein that selectively binds to the cleaved PCSK9), the protein can be referred to as the "mature,” “cleaved”, “processed” or “active” PCSK9. When only the inactive form is being referred to, the protein can be referred to as the "inactive”, “pro-form”, or "unprocessed” form of PCSK9.
  • PCSK9 as used herein also includes naturally occurring alleles, such as the mutations D374Y, S127R and F216L.
  • PCSK9 also encompasses PCSK9 molecules incorporating post-translational modifications of the PCSK9 amino acid sequence, such as PCSK9 sequences that have been glycosylated, PEGylated, PCSK9 sequences from which its signal sequence has been cleaved, PCSK9 sequence from which its pro domain has been cleaved from the catalytic domain but not separated from the catalytic domain (e.g., FIGs. 1A and IB).
  • PCSK9 activity includes any biological effect of PCSK9.
  • PCSK9 activity includes the ability of PCSK9 to interact or bind to a substrate or receptor.
  • PCSK9 activity is represented by the ability of PCSK9 to bind to a LDL receptor (LDLR).
  • LDLR LDL receptor
  • PCSK9 binds to and catalyzes a reaction involving LDLR.
  • PCSK9 activity includes the ability of PCSK9 to alter (e.g., reduce) the availability of LDLR.
  • PCSK9 activity includes the ability of PCSK9 to increase the amount of LDL in a subject.
  • PCSK9 activity includes the ability of PCSK9 to decrease the amount of LDLR that is available to bind to LDL.
  • PCSK9 activity includes any biological activity resulting from PCSK9 signaling. Exemplary activities include, but are not limited to, PCSK9 binding to LDLR, PCSK9 enzyme activity that cleaves LDLR or other proteins, PCSK9 binding to proteins other than LDLR that facilitate PCSK9 action, PCSK9 altering APOB secretion (Sun X-M et al, "Evidence for effect of mutant PCSK9 on apoliprotein B secretion as the cause of unusually severe dominant hypercholesterolemia, Human Molecular Genetics 14: 1161-1169, 2005 and Ouguerram K et al, "Apolipoprotein B100 metabolism in autosomal-dominant hypercholesterolemia related to mutations in PCSK9, Arterioscler thromb Vase Biol.
  • hypocholesterolemia refers to a condition in which cholesterol levels are elevated above a desired level. In some embodiments, this denotes that serum cholesterol levels are elevated. In some embodiments, the desired level takes into account various "risk factors" that are known to one of skill in the art (and are described or referenced herein).
  • homozygous familial hypercholesterolemia refers a cholesterol-related disorder that is determined by genetic confirmation or clinical diagnosis (such as history of an untreated LDL-cholesterol concentration greater than 13 mmol/L plus either xanthoma before 10 years of age or evidence of heterozygous familial hypercholesterolaemia in both parents).
  • polynucleotide or “nucleic acid” includes both single-stranded and double-stranded nucleotide polymers.
  • the nucleotides comprising the polynucleotide can be ribonucleotides or deoxyribonucleotides or a modified form of either type of nucleotide.
  • Said modifications include base modifications such as bromouridine and inosine derivatives, ribose modifications such as 2',3'-dideoxyribose, and internucleotide linkage modifications such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate and phosphoroamidate.
  • oligonucleotide means a polynucleotide comprising 200 or fewer nucleotides. In some embodiments, oligonucleotides are 10 to 60 bases in length. In other embodiments, oligonucleotides are 12, 13, 14, 15, 16, 17, 18, 19, or 20 to 40 nucleotides in length. Oligonucleotides can be single stranded or double stranded, e.g., for use in the construction of a mutant gene. Oligonucleotides can be sense or antisense oligonucleotides.
  • An oligonucleotide can include a label, including a radiolabel, a fluorescent label, a hapten or an antigenic label, for detection assays. Oligonucleotides can be used, for example, as PCR primers, cloning primers or hybridization probes.
  • isolated nucleic acid molecule means a DNA or R A of genomic, mRNA, cDNA, or synthetic origin or some combination thereof which is not associated with all or a portion of a polynucleotide in which the isolated polynucleotide is found in nature, or is linked to a polynucleotide to which it is not linked in nature.
  • a nucleic acid molecule comprising a particular nucleotide sequence does not encompass intact chromosomes.
  • Isolated nucleic acid molecules "comprising" specified nucleic acid sequences can include, in addition to the specified sequences, coding sequences for up to ten or even up to twenty other proteins or portions thereof, or can include operably linked regulatory sequences that control expression of the coding region of the recited nucleic acid sequences, and/or can include vector sequences.
  • the left-hand end of any single-stranded polynucleotide sequence discussed herein is the 5 ' end; the left-hand direction of double-stranded polynucleotide sequences is referred to as the 5 ' direction.
  • the direction of 5 ' to 3 ' addition of nascent R A transcripts is referred to as the transcription direction; sequence regions on the DNA strand having the same sequence as the RNA transcript that are 5 ' to the 5 ' end of the RNA transcript are referred to as "upstream sequences;" sequence regions on the DNA strand having the same sequence as the RNA transcript that are 3 ' to the 3 ' end of the RNA transcript are referred to as "downstream sequences.”
  • control sequence refers to a polynucleotide sequence that can affect the expression and processing of coding sequences to which it is ligated. The nature of such control sequences can depend upon the host organism.
  • control sequences for prokaryotes can include a promoter, a ribosomal binding site, and a transcription termination sequence.
  • control sequences for eukaryotes can include promoters comprising one or a plurality of recognition sites for transcription factors, transcription enhancer sequences, and transcription termination sequence.
  • Control sequences can include leader sequences and/or fusion partner sequences.
  • vector means any molecule or entity (e.g., nucleic acid, plasmid, bacteriophage or virus) used to transfer protein coding information into a host cell.
  • expression vector refers to a vector that is suitable for transformation of a host cell and contains nucleic acid sequences that direct and/or control (in conjunction with the host cell) expression of one or more heterologous coding regions operatively linked thereto.
  • An expression construct can include, but is not limited to, sequences that affect or control transcription, translation, and, if introns are present, affect RNA splicing of a coding region operably linked thereto.
  • operably linked means that the components to which the term is applied are in a relationship that allows them to carry out their inherent functions under suitable conditions.
  • a control sequence in a vector that is "operably linked" to a protein coding sequence is ligated thereto so that expression of the protein coding sequence is achieved under conditions compatible with the transcriptional activity of the control sequences.
  • the term "host cell” means a cell that has been transformed, or is capable of being transformed, with a nucleic acid sequence and thereby expresses a gene of interest.
  • the term includes the progeny of the parent cell, whether or not the progeny is identical in morphology or in genetic make-up to the original parent cell, so long as the gene of interest is present.
  • transfection means the uptake of foreign or exogenous DNA by a cell, and a cell has been "transfected" when the exogenous DNA has been introduced inside the cell membrane.
  • transfection techniques are well known in the art and are disclosed herein. See, e.g., Graham et al, 1973, Virology 52:456; Sambrook et al, 2001, Molecular Cloning: A Laboratory Manual, supra; Davis et al, 1986, Basic Methods in Molecular Biology, Elsevier; Chu et al, 1981, Gene 13: 197.
  • Such techniques can be used to introduce one or more exogenous DNA moieties into suitable host cells.
  • transformation refers to a change in a cell's genetic characteristics, and a cell has been transformed when it has been modified to contain new DNA or RNA.
  • a cell is transformed where it is genetically modified from its native state by introducing new genetic material via transfection, transduction, or other techniques.
  • the transforming DNA can recombine with that of the cell by physically integrating into a chromosome of the cell, or can be maintained transiently as an episomal element without being replicated, or can replicate independently as a plasmid.
  • a cell is considered to have been "stably transformed” when the transforming DNA is replicated with the division of the cell.
  • polypeptide or "protein” means a macromolecule having the amino acid sequence of a native protein, that is, a protein produced by a naturally-occurring and non-recombinant cell; or it is produced by a genetically-engineered or recombinant cell, and comprise molecules having the amino acid sequence of the native protein, or molecules having deletions from, additions to, and/or substitutions of one or more amino acids of the native sequence.
  • the term also includes amino acid polymers in which one or more amino acids are chemical analogs of a corresponding naturally- occurring amino acid and polymers.
  • polypeptide and protein specifically encompass PCSK9 antigen binding proteins, antibodies, or sequences that have deletions from, additions to, and/or substitutions of one or more amino acid of antigen-binding protein.
  • polypeptide fragment refers to a polypeptide that has an amino-terminal deletion, a carboxyl-terminal deletion, and/or an internal deletion as compared with the full-length native protein. Such fragments can also contain modified amino acids as compared with the native protein. In certain embodiments, fragments are about five to 500 amino acids long. For example, fragments can be at least 5, 6, 8, 10, 14, 20, 50, 70, 100, 110, 150, 200, 250, 300, 350, 400, or 450 amino acids long.
  • Useful polypeptide fragments include immunologically functional fragments of antibodies, including binding domains.
  • useful fragments include but are not limited to a CDR region, a variable domain of a heavy and/or light chain, a portion of an antibody chain or just its variable region including two CDRs, and the like.
  • isolated protein means that a subject protein (1) is free of at least some other proteins with which it would normally be found, (2) is essentially free of other proteins from the same source, e.g., from the same species, (3) is expressed by a cell from a different species, (4) has been separated from at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is associated in nature, (5) is operably associated (by covalent or noncovalent interaction) with a polypeptide with which it is not associated in nature, or (6) does not occur in nature.
  • an "isolated protein" constitutes at least about 5%, at least about 10%, at least about 25%, or at least about 50% of a given sample.
  • Genomic DNA, cDNA, mRNA or other RNA, of synthetic origin, or any combination thereof can encode such an isolated protein.
  • the isolated protein is substantially free from proteins or polypeptides or other contaminants that are found in its natural environment that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.
  • amino acid includes its normal meaning in the art.
  • a "variant" of a polypeptide comprises an amino acid sequence wherein one or more amino acid residues are inserted into, deleted from and/or substituted into the amino acid sequence relative to another polypeptide sequence.
  • Variants include fusion proteins.
  • identity refers to a relationship between the sequences of two or more polypeptide molecules or two or more nucleic acid molecules, as determined by aligning and comparing the sequences. "Percent identity” means the percent of identical residues between the amino acids or nucleotides in the compared molecules and is calculated based on the size of the smallest of the molecules being compared. For these calculations, gaps in alignments (if any) are preferably addressed by a particular mathematical model or computer program (i.e., an "algorithm”). Methods that can be used to calculate the identity of the aligned nucleic acids or polypeptides include those described in Computational Molecular Biology, (Lesk, A.
  • the sequences being compared are typically aligned in a way that gives the largest match between the sequences.
  • One example of a computer program that can be used to determine percent identity is the GCG program package, which includes GAP (Devereux et al, 1984, Nucl. Acid Res. 12:387; Genetics Computer Group, University of Wisconsin, Madison, WI).
  • GAP is used to align the two polypeptides or polynucleotides for which the percent sequence identity is to be determined.
  • the sequences are aligned for optimal matching of their respective amino acid or nucleotide (the "matched span", as determined by the algorithm).
  • a gap opening penalty (which is calculated as 3x the average diagonal, wherein the "average diagonal” is the average of the diagonal of the comparison matrix being used; the “diagonal” is the score or number assigned to each perfect amino acid match by the particular comparison matrix) and a gap extension penalty (which is usually 1/10 times the gap opening penalty), as well as a comparison matrix such as PAM 250 or BLOSum 62 are used in conjunction with the algorithm.
  • a standard comparison matrix (see, Dayhoff et al., 1978, Atlas of Protein Sequence and Structure 5:345-352 for the PAM 250 comparison matrix; Henikoff et al, 1992, Proc. Natl. Acad. Sci. U.S.A. 89:10915-10919 for the BLOSum 62 comparison matrix) is also used by the algorithm.
  • Certain alignment schemes for aligning two amino acid sequences may result in matching of only a short region of the two sequences, and this small aligned region may have very high sequence identity even though there is no significant relationship between the two full-length sequences. Accordingly, the selected alignment method (GAP program) can be adjusted if so desired to result in an alignment that spans at least 50 or other number of contiguous amino acids of the target polypeptide.
  • the twenty conventional (e.g., naturally occurring) amino acids and their abbreviations follow conventional usage. See Immunology— A Synthesis (2nd Edition, E. S. Golub and D. R. Gren, Eds., Sinauer Associates, Sunderland, Mass. (1991)), which is incorporated herein by reference for any purpose. Stereoisomers (e.g., D-amino acids) of the twenty conventional amino acids, unnatural amino acids such as ⁇ -, ⁇ -disubstituted amino acids, N-alkyl amino acids, lactic acid, and other unconventional amino acids can also be suitable components for polypeptides of the present invention.
  • Examples of unconventional amino acids include: 4-hydroxyproline, ⁇ -carboxyglutamate, ⁇ - ⁇ , ⁇ , ⁇ -trimethyllysine, ⁇ - ⁇ -acetyllysine, O-phosphoserine, N- acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, ⁇ - ⁇ - methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxyproline).
  • the left-hand direction is the amino terminal direction and the right-hand direction is the carboxy-terminal direction, in accordance with standard usage and convention.
  • the left-hand end of single-stranded polynucleotide sequences is the 5' end; the left-hand direction of double-stranded polynucleotide sequences is referred to as the 5' direction.
  • the direction of 5' to 3' addition of nascent R A transcripts is referred to as the transcription direction; sequence regions on the DNA strand having the same sequence as the RNA and which are 5' to the 5' end of the RNA transcript are referred to as "upstream sequences"; sequence regions on the DNA strand having the same sequence as the RNA and which are 3' to the 3' end of the RNA transcript are referred to as "downstream sequences.”
  • amino acid residues can encompass non-naturally occurring amino acid residues, which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. These include peptidomimetics and other reversed or inverted forms of amino acid moieties.
  • Naturally occurring residues can be divided into classes based on common side chain properties:
  • non-conservative substitutions can involve the exchange of a member of one of these classes for a member from another class.
  • substituted residues can be introduced, for example, into regions of a human antibody that are homologous with non-human antibodies, or into the non-homologous regions of the molecule.
  • the hydropathic index of amino acids can be considered.
  • Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. They are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (- 3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
  • hydropathic amino acid index in conferring interactive biological function on a protein is understood in the art. Kyte et al., J. Mol. Biol., 157: 105-131 (1982). It is known that certain amino acids can be substituted for other amino acids having a similar hydropathic index or score and still retain a similar biological activity. In making changes based upon the hydropathic index, in certain embodiments, the substitution of amino acids whose hydropathic indices are within ⁇ 2 is included. In certain embodiments, those which are within ⁇ 1 are included, and in certain embodiments, those within ⁇ 0.5 are included.
  • the substitution of like amino acids can be made effectively on the basis of hydrophilicity, particularly where the biologically functional protein or peptide thereby created is intended for use in immunological embodiments, as in the present case.
  • the greatest local average hydrophilicity of a protein as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigenicity, i.e., with a biological property of the protein.
  • hydrophilicity values have been assigned to these amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 ⁇ 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (- 1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5) and tryptophan (-3.4).
  • the substitution of amino acids whose hydrophilicity values are within ⁇ 2 is included, in certain embodiments, those which are within ⁇ 1 are included, and in certain embodiments, those within ⁇ 0.5 are included.
  • derivative refers to a molecule that includes a chemical modification other than an insertion, deletion, or substitution of amino acids (or nucleic acids).
  • derivatives comprise covalent modifications, including, but not limited to, chemical bonding with polymers, lipids, or other organic or inorganic moieties.
  • a chemically modified antigen binding protein can have a greater circulating half-life than an antigen binding protein that is not chemically modified.
  • a chemically modified antigen binding protein can have improved targeting capacity for desired cells, tissues, and/or organs.
  • a derivative antigen binding protein is covalently modified to include one or more water soluble polymer attachments, including, but not limited to, polyethylene glycol, polyoxyethylene glycol, or polypropylene glycol. See, e.g., U.S. Patent Nos: 4,640,835, 4,496,689, 4,301 ,144, 4,670,417, 4,791 ,192 and 4,179,337.
  • a derivative antigen binding protein comprises one or more polymer, including, but not limited to, monomethoxy-polyethylene glycol, dextran, cellulose, or other carbohydrate based polymers, poly-(N-vinyl pyrrolidone)- polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol, as well as mixtures of such polymers.
  • polymer including, but not limited to, monomethoxy-polyethylene glycol, dextran, cellulose, or other carbohydrate based polymers, poly-(N-vinyl pyrrolidone)- polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol, as well
  • a derivative is covalently modified with polyethylene glycol (PEG) subunits.
  • PEG polyethylene glycol
  • one or more water-soluble polymer is bonded at one or more specific position, for example at the amino terminus, of a derivative.
  • one or more water-soluble polymer is randomly attached to one or more side chains of a derivative.
  • PEG is used to improve the therapeutic capacity for an antigen binding protein.
  • PEG is used to improve the therapeutic capacity for a humanized antibody.
  • Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed "peptide mimetics” or “peptidomimetics.” Fauchere, J., Adv. Drug Res., 15:29 (1986); Veber & Freidinger, TINS, p.392 (1985); and Evans et al., J. Med. Chem., 30: 1229 (1987), which are incorporated herein by reference for any purpose. Such compounds are often developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides can be used to produce a similar therapeutic or prophylactic effect.
  • a paradigm polypeptide i.e., a polypeptide that has a biochemical property or pharmacological activity
  • Systematic substitution of one or more amino acids of a consensus sequence with a D-amino acid of the same type can be used in certain embodiments to generate more stable peptides.
  • constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation can be generated by methods known in the art (Rizo and Gierasch, Ann. Rev. Biochem., 61 :387 (1992), incorporated herein by reference for any purpose); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the peptide.
  • naturally occurring refers to materials which are found in nature or a form of the materials that is found in nature.
  • an “antigen binding protein” as used herein means any protein that binds a specified target antigen.
  • the specified target antigen is the PCSK9 protein or fragment thereof.
  • Antigen binding protein includes but is not limited to antibodies and binding parts thereof, such as immunologically functional fragments. Peptibodies are another example of antigen binding proteins.
  • immunologically functional fragment of an antibody or immunoglobulin chain (heavy or light chain) antigen binding protein, as used herein, is a species of antigen binding protein comprising a portion (regardless of how that portion is obtained or synthesized) of an antibody that lacks at least some of the amino acids present in a full-length chain but which is still capable of specifically binding to an antigen.
  • fragments are biologically active in that they bind to the target antigen and can compete with other antigen binding proteins, including intact antibodies, for binding to a given epitope.
  • the fragments are neutralizing fragments.
  • the fragments can block or reduce the likelihood of the interaction between LDLR and PCSK9.
  • such a fragment will retain at least one CDR present in the full-length light or heavy chain, and in some embodiments will comprise a single heavy chain and/or light chain or portion thereof.
  • These biologically active fragments can be produced by recombinant DNA techniques, or can be produced by enzymatic or chemical cleavage of antigen binding proteins, including intact antibodies.
  • Immunologically functional immunoglobulin fragments include, but are not limited to, Fab, a diabody (heavy chain variable domain on the same polypeptide as a light chain variable domain, connected via a short peptide linker that is too short to permit pairing between the two domains on the same chain), Fab', F(ab') 2 , Fv, domain antibodies and single-chain antibodies, and can be derived from any mammalian source, including but not limited to human, mouse, rat, camelid or rabbit.
  • an antigen binding protein can include nonprotein components.
  • ABPs are described herein in terms of "number/letter/number" (e.g., 25 A7). In these cases, the exact name denotes a specific antibody (e.g., 25A7 versus 21B12).
  • an ABP named 25A7 is not necessarily the same as an antibody named 25A7.1, (unless they are explicitly taught as the same in the specification, e.g., 25 A7 and 25A7.3). Unless otherwise stated, the ABP name is understood to be a generic designation denoting an antibody.
  • antigen binding proteins described herein are antibodies or are derived from antibodies.
  • the polypeptide structure of the antigen binding proteins is based on antibodies, including, but not limited to, monoclonal antibodies, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), chimeric antibodies, humanized antibodies, human antibodies, antibody fusions (sometimes referred to herein as "antibody conjugates”), and fragments thereof, respectively.
  • the ABP comprises or consists of avimers (tightly binding peptide). These various antigen binding proteins are further described herein.
  • examples of antibodies are provided in e.g., U.S.P. 8,030,457, U.S.P.
  • An "Fc" region comprises two heavy chain fragments comprising the C H I and C H 2 domains of an antibody.
  • the two heavy chain fragments are held together by two or more disulfide bonds and by hydrophobic interactions of the C H 3 domains.
  • a "Fab fragment” comprises one light chain and the C R I and variable regions of one heavy chain.
  • the heavy chain of a Fab molecule cannot form a disulfide bond with another heavy chain molecule.
  • a “Fab' fragment” comprises one light chain and a portion of one heavy chain that contains the VH domain and the C R I domain and also the region between the C R I and C H 2 domains, such that an interchain disulfide bond can be formed between the two heavy chains of two Fab' fragments to form an F(ab') 2 molecule.
  • a “F(ab') 2 fragment” contains two light chains and two heavy chains containing a portion of the constant region between the C R I and C R 2 domains, such that an interchain disulfide bond is formed between the two heavy chains.
  • a F(ab') 2 fragment thus is composed of two Fab' fragments that are held together by a disulfide bond between the two heavy chains.
  • the "Fv region” comprises the variable regions from both the heavy and light chains, but lacks the constant regions.
  • Single-chain antibodies are Fv molecules in which the heavy and light chain variable regions have been connected by a flexible linker to form a single polypeptide chain, which forms an antigen binding region.
  • Single chain antibodies are discussed in detail in International Patent Application Publication No. WO 88/01649 and United States Patent Nos. 4,946,778 and No. 5,260,203, the disclosures of which are incorporated by reference.
  • a “domain antibody” is an immunologically functional immunoglobulin fragment containing only the variable region of a heavy chain or the variable region of a light chain.
  • two or more V H regions are covalently joined with a peptide linker to create a bivalent domain antibody.
  • the two V H regions of a bivalent domain antibody can target the same or different antigens.
  • a “bivalent antigen binding protein” or “bivalent antibody” comprises two antigen binding sites. In some instances, the two binding sites have the same antigen specificities. Bivalent antigen binding proteins and bivalent antibodies can be bispecific, see, infra. A bivalent antibody other than a “multispecific” or “multifunctional” antibody, in certain embodiments, typically is understood to have each of its binding sites identical.
  • a “multispecific antigen binding protein” or “multispecific antibody” is one that targets more than one antigen or epitope.
  • a "bispecific,” “dual-specific” or “bifunctional” antigen binding protein or antibody is a hybrid antigen binding protein or antibody, respectively, having two different antigen binding sites.
  • Bispecific antigen binding proteins and antibodies are a species of multispecific antigen binding protein antibody and can be produced by a variety of methods including, but not limited to, fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai and Lachmann, 1990, Clin. Exp. Immunol. 79:315-321; Kostelny et al, 1992, J. Immunol. 148: 1547-1553.
  • the two binding sites of a bispecific antigen binding protein or antibody will bind to two different epitopes, which can reside on the same or different protein targets.
  • An antigen binding protein is said to "specifically bind” its target antigen when the dissociation constant (Ka) is ⁇ 10 ⁇ 7 M.
  • the ABP specifically binds antigen with "high affinity” when the Ka is ⁇ 5 x 10 ⁇ 9 M, and with "very high affinity” when the Ka is ⁇ 5x 10 "10 M.
  • the ABP has a K d of ⁇ 10 ⁇ 9 M.
  • the off-rate is ⁇ 1 x 10 ⁇ 5 .
  • the ABPs will bind to human PCSK9 with a IQ of between about 10 ⁇ 9 M and 10 ⁇ 13 M, and in yet another embodiment the ABPs will bind with a Ka ⁇ 5 x 10 ⁇ 10 .
  • any or all of the antigen binding fragments can specifically bind to PCSK9.
  • An antigen binding protein is "selective" when it binds to one target more tightly than it binds to a second target.
  • Antigen binding region means a protein, or a portion of a protein, that specifically binds a specified antigen (e.g., a paratope). For example, that portion of an antigen binding protein that contains the amino acid residues that interact with an antigen and confer on the antigen binding protein its specificity and affinity for the antigen is referred to as "antigen binding region.”
  • An antigen binding region typically includes one or more “complementary binding regions” (“CDRs"). Certain antigen binding regions also include one or more "framework” regions.
  • CDR is an amino acid sequence that contributes to antigen binding specificity and affinity. "Framework” regions can aid in maintaining the proper conformation of the CDRs to promote binding between the antigen binding region and an antigen.
  • framework regions can be located in antibodies between CDRs. Examples of framework and CDR regions are shown in FIGs. 2A-3D, 3CCC-3JJJ.
  • the sequences for CDRs for the light chain of antibody 3B6 are as follows: CDR1 TLSSGYSSYEVD (SEQ ID NO: 279); CDR2 VDTGGIVGSKGE (SEQ ID NO: 280); CDR3 GADHGSGTNFVVV (SEQ ID NO: 281), and the FRs are as follows: FR1 QPVLTQPLFASASLGASVTLTC (SEQ ID NO: 282); FR2 WYQQRPGKGPRFVMR (SEQ ID NO: 283); FR3 GIPDRFSVLGSGLNRYLTIK IQEEDESDYHC (SEQ ID NO: 284); and FR4 FGGGTKLTVL (SEQ ID NO: 285).
  • recombinant antigen binding proteins that bind PCSK9, for example human PCSK9
  • a "recombinant antigen binding protein” is a protein made using recombinant techniques, i.e., through the expression of a recombinant nucleic acid as described herein. Methods and techniques for the production of recombinant proteins are well known in the art.
  • antibody refers to an intact immunoglobulin of any isotype, or a fragment thereof that can compete with the intact antibody for specific binding to the target antigen, and includes, for instance, chimeric, humanized, fully human, and bispecific antibodies.
  • An "antibody” is a species of an antigen binding protein.
  • An intact antibody will generally comprise at least two full-length heavy chains and two full-length light chains, but in some instances can include fewer chains such as antibodies naturally occurring in camelids which can comprise only heavy chains.
  • Antibodies can be derived solely from a single source, or can be "chimeric,” that is, different portions of the antibody can be derived from two different antibodies as described further below.
  • antigen binding proteins, antibodies, or binding fragments can be produced in hybridomas, by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact antibodies.
  • antibody includes, in addition to antibodies comprising two full-length heavy chains and two full-length light chains, derivatives, variants, fragments, and muteins thereof, examples of which are described below.
  • antibodies include monoclonal antibodies, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), chimeric antibodies, humanized antibodies, human antibodies, antibody fusions (sometimes referred to herein as "antibody conjugates”), and fragments thereof, respectively.
  • the term also encompasses peptibodies.
  • Naturally occurring antibody structural units typically comprise a tetramer.
  • Each such tetramer typically is composed of two identical pairs of polypeptide chains, each pair having one full-length "light” (in certain embodiments, about 25 kDa) and one full-length "heavy” chain (in certain embodiments, about 50-70 kDa).
  • the amino- terminal portion of each chain typically includes a variable region of about 100 to 110 or more amino acids that typically is responsible for antigen recognition.
  • the carboxy- terminal portion of each chain typically defines a constant region that can be responsible for effector function.
  • Human light chains are typically classified as kappa and lambda light chains.
  • Heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • IgG has several subclasses, including, but not limited to, IgGl, IgG2, IgG3, and IgG4.
  • IgM has subclasses including, but not limited to, IgMl and IgM2.
  • IgA is similarly subdivided into subclasses including, but not limited to, IgAl and IgA2.
  • variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D” region of about 10 more amino acids.
  • J Fundamental Immunology
  • the variable regions of each light/heavy chain pair typically form the antigen binding site.
  • variable regions typically exhibit the same general structure of relatively conserved framework regions (FR) joined by three hyper variable regions, also called complementarity determining regions or CDRs.
  • the CDRs from the two chains of each pair typically are aligned by the framework regions, which can enable binding to a specific epitope.
  • both light and heavy chain variable regions typically comprise the domains FRl, CDRl, FR2, CDR2, FR3, CDR3 and FR4.
  • the assignment of amino acids to each domain is typically in accordance with the definitions of Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk, J. Mol. Biol., 196:901-917 (1987); Chothia et al, Nature, 342:878-883 (1989).
  • an antibody heavy chain binds to an antigen in the absence of an antibody light chain. In certain embodiments, an antibody light chain binds to an antigen in the absence of an antibody heavy chain. In certain embodiments, an antibody binding region binds to an antigen in the absence of an antibody light chain. In certain embodiments, an antibody binding region binds to an antigen in the absence of an antibody heavy chain. In certain embodiments, an individual variable region specifically binds to an antigen in the absence of other variable regions.
  • definitive delineation of a CDR and identification of residues comprising the binding site of an antibody is accomplished by solving the structure of the antibody and/or solving the structure of the antibody-ligand complex. In certain embodiments, that can be accomplished by any of a variety of techniques known to those skilled in the art, such as X-ray crystallography. In certain embodiments, various methods of analysis can be employed to identify or approximate the CDR regions. Examples of such methods include, but are not limited to, the Kabat definition, the Chothia definition, the AbM definition and the contact definition.
  • the Kabat definition is a standard for numbering the residues in an antibody and is typically used to identify CDR regions. See, e.g., Johnson & Wu, Nucleic Acids Res., 28: 214-8 (2000).
  • the Chothia definition is similar to the Kabat definition, but the Chothia definition takes into account positions of certain structural loop regions. See, e.g., Chothia et al, J. Mol. Biol, 196: 901-17 (1986); Chothia et al, Nature, 342: 877-83 (1989).
  • the AbM definition uses an integrated suite of computer programs produced by Oxford Molecular Group that model antibody structure.
  • the AbM definition models the tertiary structure of an antibody from primary sequence using a combination of knowledge databases and ab initio methods, such as those described by Samudrala et al, "Ab Initio Protein Structure Prediction Using a Combined Hierarchical Approach,” in PROTEINS, Structure, Function and Genetics SuppL, 3: 194-198 (1999).
  • the contact definition is based on an analysis of the available complex crystal structures. See, e.g., MacCallum et al, J. Mol. Biol., 5:732-45 (1996).
  • the CDR regions in the heavy chain are typically referred to as HI, H2, and H3 and are numbered sequentially in the direction from the amino terminus to the carboxy terminus.
  • the CDR regions in the light chain are typically referred to as LI, L2, and L3 and are numbered sequentially in the direction from the amino terminus to the carboxy terminus.
  • the term "light chain” includes a full-length light chain and fragments thereof having sufficient variable region sequence to confer binding specificity.
  • a full-length light chain includes a variable region domain, V L , and a constant region domain, C L .
  • the variable region domain of the light chain is at the amino-terminus of the polypeptide.
  • Light chains include kappa chains and lambda chains.
  • heavy chain includes a full-length heavy chain and fragments thereof having sufficient variable region sequence to confer binding specificity.
  • a full- length heavy chain includes a variable region domain, V R , and three constant region domains, C H I , C H 2, and C H 3.
  • the V H domain is at the amino-terminus of the polypeptide, and the C H domains are at the carboxyl-terminus, with the C H 3 being closest to the carboxy-terminus of the polypeptide.
  • Heavy chains can be of any isotype, including IgG (including IgGl, IgG2, IgG3 and IgG4 subtypes), IgA (including IgAl and IgA2 subtypes), IgM and IgE.
  • a bispecific or bifunctional antibody typically is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including, but not limited to, fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai et ah, Clin. Exp. Immunol, 79: 315-321 (1990); Kostelny et al, J. Immunol, 148: 1547-1553 (1992).
  • Some species of mammals also produce antibodies having only a single heavy chain.
  • Each individual immunoglobulin chain is typically composed of several "immunoglobulin domains," each consisting of roughly 90 to 110 amino acids and having a characteristic folding pattern. These domains are the basic units of which antibody polypeptides are composed.
  • the IgA and IgD isotypes contain four heavy chains and four light chains; the IgG and IgE isotypes contain two heavy chains and two light chains; and the IgM isotype contains five heavy chains and five light chains.
  • the heavy chain C region typically comprises one or more domains that can be responsible for effector function. The number of heavy chain constant region domains will depend on the isotype.
  • IgG heavy chains for example, contain three C region domains known as C H I , C H 2 and C H 3.
  • the antibodies that are provided can have any of these isotypes and subtypes.
  • an anti-PCSK9 antibody is of the IgG2 or IgG4 subtype.
  • variable region refers to a portion of the light and/or heavy chains of an antibody, typically including approximately the amino- terminal 120 to 130 amino acids in the heavy chain and about 100 to 110 amino terminal amino acids in the light chain.
  • variable regions of different antibodies differ extensively in amino acid sequence even among antibodies of the same species.
  • the variable region of an antibody typically determines specificity of a particular antibody for its target
  • neutralizing antigen binding protein or “neutralizing antibody” refers to an antigen binding protein or antibody, respectively, that binds to a ligand and prevents or reduces the biological effect of that ligand. This can be done, for example, by directly blocking a binding site on the ligand or by binding to the ligand and altering the ligand's ability to bind through indirect means (such as structural or energetic alterations in the ligand).
  • the term can also denote an antigen binding protein that prevents the protein to which it is bound from performing a biological function.
  • an antibody or fragment in assessing the binding and/or specificity of an antigen binding protein, e.g., an antibody or immunologically functional fragment thereof, can substantially inhibit binding of a ligand to its binding partner when an excess of antibody reduces the quantity of binding partner bound to the ligand by at least about 1-20, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-85%, 85- 90%, 90-95%, 95-97%, 97-98%, 98-99% or more (as measured in an in vitro competitive binding assay).
  • such a neutralizing molecule can diminish the ability of PCSK9 to bind the LDLR.
  • the neutralizing ability is characterized and/or described via a competition assay. In some embodiments, the neutralizing ability is described in terms of an IC 50 or EC 50 value.
  • ABPs 27B2, 13H1, 13B5 and 3C4 are non-neutralizing ABPs, 3B6, 9C9 and 31A4 are weak neutralizers, and the remaining ABPs in Table 2 are strong neutralizers.
  • the antibodies or antigen binding proteins neutralize by binding to PCSK9 and preventing PCSK9 from binding to LDLR (or reducing the ability of PCSK9 to bind to LDLR).
  • the antibodies or ABPs neutralize by binding to PCSK9, and while still allowing PCSK9 to bind to LDLR, preventing or reducing the PCSK9 mediated degradation of LDLR.
  • a neutralizing ABP or antibody can still permit PCSK9/LDLR binding, but will prevent (or reduce) subsequent PCSK9 involved degradation of LDLR.
  • target refers to a molecule or a portion of a molecule capable of being bound by an antigen binding protein.
  • a target can have one or more epitopes.
  • a target is an antigen.
  • the use of "antigen” in the phrase “antigen binding protein” simply denotes that the protein sequence that comprises the antigen can be bound by an antibody. In this context, it does not require that the protein be foreign or that it be capable of inducing an immune response.
  • antigen binding proteins e.g., neutralizing antigen binding proteins or neutralizing antibodies
  • competition means competition between antigen binding proteins as determined by an assay in which the antigen binding protein (e.g., antibody or immunologically functional fragment thereof) being tested prevents or inhibits (e.g., reduces) specific binding of a reference antigen binding protein (e.g. , a ligand, or a reference antibody) to a common antigen (e.g., PCSK9 or a fragment thereof).
  • a reference antigen binding protein e.g. , a ligand, or a reference antibody
  • RIA solid phase direct or indirect radioimmunoassay
  • EIA solid phase direct or indirect enzyme immunoassay
  • sandwich competition assay see, e.g., Stahli et al, 1983, Methods in Enzymology 9:242-253
  • solid phase direct biotin-avidin EIA see, e.g., Kirkland et al., 1986, J. Immunol.
  • solid phase direct labeled assay solid phase direct labeled sandwich assay (see, e.g., Harlow and Lane, 1988, Antibodies, A Laboratory Manual, Cold Spring Harbor Press); solid phase direct label RIA using 1-125 label (see, e.g., Morel et al., 1988, Molec. Immunol. 25:7-15); solid phase direct biotin-avidin EIA (see, e.g., Cheung, et al, 1990, Virology 176:546-552); and direct labeled RIA (Moldenhauer et al, 1990, Scand. J. Immunol. 32:77-82).
  • such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabelled test antigen binding protein and a labeled reference antigen binding protein.
  • Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test antigen binding protein.
  • the test antigen binding protein is present in excess.
  • Antigen binding proteins identified by competition assay include antigen binding proteins binding to the same epitope as the reference antigen binding proteins and antigen binding proteins binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antigen binding protein for steric hindrance to occur. Additional details regarding methods for determining competitive binding are provided in the examples herein.
  • a competing antigen binding protein when present in excess, it will inhibit (e.g. , reduce) specific binding of a reference antigen binding protein to a common antigen by at least 40-45%, 45-50%, 50-55%, 55-60%, 60-65%, 65-70%), 70-75%) or 75% or more. In some instances, binding is inhibited by at least 80-85%, 85-90%, 90-95%, 95-97%, or 97% or more.
  • antigen refers to a molecule or a portion of a molecule capable of being bound by a selective binding agent, such as an antigen binding protein (including, e.g., an antibody or immunological functional fragment thereof).
  • an antigen binding protein including, e.g., an antibody or immunological functional fragment thereof.
  • the antigen is capable of being used in an animal to produce antibodies capable of binding to that antigen.
  • An antigen can possess one or more epitopes that are capable of interacting with different antigen binding proteins, e.g., antibodies.
  • epitope includes any determinant capable being bound by an antigen binding protein, such as an antibody or to a T-cell receptor.
  • An epitope is a region of an antigen that is bound by an antigen binding protein that targets that antigen, and when the antigen is a protein, includes specific amino acids that directly contact the antigen binding protein. Most often, epitopes reside on proteins, but in some instances can reside on other kinds of molecules, such as nucleic acids.
  • Epitope determinants can include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl or sulfonyl groups, and can have specific three dimensional structural characteristics, and/or specific charge characteristics.
  • substantially pure means that the described species of molecule is the predominant species present, that is, on a molar basis it is more abundant than any other individual species in the same mixture.
  • a substantially pure molecule is a composition wherein the object species comprises at least 50% (on a molar basis) of all macromolecular species present.
  • a substantially pure composition will comprise at least 80%, 85%, 90%, 95%, or 99% of all macromolecular species present in the composition.
  • the object species is purified to essential homogeneity wherein contaminating species cannot be detected in the composition by conventional detection methods and thus the composition consists of a single detectable macromolecular species.
  • agent is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
  • label refers to incorporation of a detectable marker, e.g., by incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotin moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods).
  • marked avidin e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods.
  • the label or marker can also be therapeutic.
  • Various methods of labeling polypeptides and glycoproteins are known in the art and can be used.
  • labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, U1 ln, 125 I, 131 I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, ⁇ -galactosidase, luciferase, alkaline phosphatase), chemiluminescent, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags).
  • labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
  • biological sample includes, but is not limited to, any quantity of a substance from a living thing or formerly living thing.
  • living things include, but are not limited to, humans, mice, monkeys, rats, rabbits, and other animals.
  • substances include, but are not limited to, blood, serum, urine, cells, organs, tissues, bone, bone marrow, lymph nodes, and skin.
  • pharmaceutical agent composition refers to a chemical compound, composition, agent or drug capable of inducing a desired therapeutic effect when properly administered to a patient. It does not necessarily require more than one type of ingredient.
  • therapeutically effective amount refers to the amount of a PCSK9 antigen binding protein determined to produce a therapeutic response in a mammal. Such therapeutically effective amounts are readily ascertained by one of ordinary skill in the art.
  • modulator is a compound that changes or alters the activity or function of a molecule.
  • a modulator can cause an increase or decrease in the magnitude of a certain activity or function of a molecule compared to the magnitude of the activity or function observed in the absence of the modulator.
  • a modulator is an inhibitor, which decreases the magnitude of at least one activity or function of a molecule.
  • Certain exemplary activities and functions of a molecule include, but are not limited to, binding affinity, enzymatic activity, and signal transduction.
  • Certain exemplary inhibitors include, but are not limited to, proteins, peptides, antibodies, peptibodies, carbohydrates or small organic molecules. Peptibodies are described in, e.g., U.S. Patent No. 6,660,843 (corresponding to PCT Application No. WO 01/83525).
  • patient and “subject” are used interchangeably and include human and non-human animal subjects as well as those with formally diagnosed disorders, those without formally recognized disorders, those receiving medical attention, those at risk of developing the disorders, etc.
  • treat and treatment includes therapeutic treatments, prophylactic treatments, and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses embodiments in which one reduces symptoms or underlying risk factors.
  • prevent does not require the 100% elimination of the possibility of an event. Rather, it denotes that the likelihood of the occurrence of the event has been reduced in the presence of the compound or method.
  • Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection).
  • Enzymatic reactions and purification techniques can be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein.
  • the foregoing techniques and procedures can be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.
  • PCSK9 Proprotein convertase subtilisin kexin type 9
  • LDLR low density lipoprotein receptor
  • PCSK9 is a prohormone -proprotein convertase in the subtilisin (S8) family of serine proteases (Seidah et al., 2003).
  • An exemplary human PCSK9 amino acid sequence is presented as SEQ ID NO: 1 in FIG. 1A (depicting the "pro" domain of the protein as underlined) and SEQ ID NO:3 in FIG. IB (depicting the signal sequence in bold and the pro domain underlined).
  • PCSK9 proteins can also include fragments of the full length PCSK9 protein.
  • the structure of the PCSK9 protein was solved by two groups (Cunningham et al., Nature Structural & Molecular Biology, 2007, and Piper et al., Structure, 15: 1-8, 2007), the entireties of both of which are herein incorporated by reference.
  • PCSK9 includes a signal sequence, a N-terminal prodomain, a subtilisin-like catalytic domain and a C-terminal domain.
  • Antigen binding proteins that bind PCSK9, including human PCSK9, are used in the methods provided herein.
  • the antigen binding proteins are polypeptides which comprise one or more complementary determining regions (CDRs), as described herein.
  • CDRs complementary determining regions
  • the CDRs are embedded into a "framework" region, which orients the CDR(s) such that the proper antigen binding properties of the CDR(s) is achieved.
  • antigen binding proteins that can be used in the methods provided herein can interfere with, block, reduce or modulate the interaction between PCSK9 and LDLR.
  • antigen binding proteins are denoted as “neutralizing.”
  • binding between PCSK9 and LDLR can still occur, even though the antigen binding protein is neutralizing and bound to PCSK9.
  • the ABP useful in the methods provided herein prevents or reduces the adverse influence of PCSK9 on LDLR without blocking the LDLR binding site on PCSK9.
  • the ABP modulates or alters PCSK9's ability to result in the degradation of LDLR, without having to prevent the binding interaction between PCSK9 and LDLR.
  • Such ABPs can be specifically described as "non-competitively neutralizing" ABPs.
  • the neutralizing ABP binds to PCSK9 in a location and/or manner that prevents PCSK9 from binding to LDLR.
  • ABPs can be specifically described as “competitively neutralizing" ABPs. Both of the above neutralizers can result in a greater amount of free LDLR being present in a subject, which results in more LDLR binding to LDL (thereby reducing the amount of LDL in the subject). In turn, this results in a reduction in the amount of serum cholesterol present in a subject.
  • the antigen binding proteins provided herein are capable of inhibiting PCSK9-mediated activity (including binding). In some embodiments, antigen binding proteins binding to these epitopes inhibit, inter alia, interactions between PCSK9 and LDLR and other physiological effects mediated by PCSK9. In some embodiments, the antigen binding proteins are human, such as fully human antibodies to PCSK9.
  • the ABP binds to the catalytic domain of PCSK9. In some embodiments, the ABP binds to the mature form of PCSK9. In some embodiments the ABP binds in the prodomain of PCSK9. In some embodiments, the ABP selectively binds to the mature form of PCSK9. In some embodiments, the ABP binds to the catalytic domain in a manner such that PCSK9 cannot bind or bind as efficiently to LDLR. In some embodiments, the antigen binding protein does not bind to the c-terminus of the catalytic domain. In some embodiments, the antigen binding protein does not bind to the n-terminus of the catalytic domain.
  • the ABP does not bind to the n- or c-terminus of the PCSK9 protein. In some embodiments, the ABP binds to any one of the epitopes bound by the antibodies discussed herein. In some embodiments, this can be determined by competition assays between the antibodies disclosed herein and other antibodies. In some embodiments, the ABP binds to an epitope bound by one of the antibodies described in Table 2. In some embodiments, the antigen binding proteins bind to a specific conformational state of PCSK9 so as to prevent PCSK9 from interacting with LDLR. In some embodiments, the ABP binds to the V domain of PCSK9.
  • the ABP binds to the V domain of PCSK9 and prevents (or reduces) PCSK9 from binding to LDLR. In some embodiments, the ABP binds to the V domain of PCSK9, and while it does not prevent (or reduce) the binding of PCSK9 to LDLR, the ABP prevents or reduces the adverse activities mediated through PCSK9 on LDLR.
  • the disclosed antigen binding proteins that are useful in the methods provided herein have a variety of utilities. Some of the antigen binding proteins, for instance, are useful in specific binding assays, affinity purification of PCSK9, in particular human PCSK9 or its ligands and in screening assays to identify other antagonists of PCSK9 activity. Some of the antigen binding proteins are useful for inhibiting binding of PCSK9 to LDLR, or inhibiting PCSK9-mediated activities.
  • the antigen binding proteins that are useful in the methods provided herein comprise one or more CDRs (e.g., 1 , 2, 3, 4, 5 or 6 CDRs).
  • the antigen binding protein comprises (a) a polypeptide structure and (b) one or more CDRs that are inserted into and/or joined to the polypeptide structure.
  • the polypeptide structure can take a variety of different forms. For example, it can be, or comprise, the framework of a naturally occurring antibody, or fragment or variant thereof, or can be completely synthetic in nature. Examples of various polypeptide structures are further described below.
  • the polypeptide structure of the antigen binding proteins is an antibody or is derived from an antibody, including, but not limited to, monoclonal antibodies, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), chimeric antibodies, humanized antibodies, antibody fusions (sometimes referred to as "antibody conjugates"), and portions or fragments of each, respectively.
  • the antigen binding protein is an immunological fragment of an antibody (e.g., a Fab, a Fab', a F(ab') 2 , or a scFv).
  • the antigen binding protein specifically and/or selectively binds to human PCSK9 protein having and/or consisting of residues 153-692 of SEQ ID NO: 3.
  • the ABP specifically and/or selectively binds to human PCSK9 having and/or consisting of residues 31-152 of SEQ ID NO: 3.
  • the ABP selectively binds to a human PCSK9 protein as depicted in FIG. 1A (SEQ ID NO: 1).
  • the antigen binding protein specifically binds to at least a fragment of the PCSK9 protein and/or a full length PCSK9 protein, with or without a signal sequence.
  • an antigen binding protein can inhibit, interfere with or modulate one or more biological activities of PCSK9.
  • an antigen binding protein binds specifically to human PCSK9 and/or substantially inhibits binding of human PCSK9 to LDLR by at least about 20%-40%, 40-60%, 60-80%, 80-85%, or more (for example, by measuring binding in an in vitro competitive binding assay).
  • Some of the antigen binding proteins that are provided herein are antibodies.
  • the ABP has a 3 ⁇ 4 of less (binding more tightly) than 10 "7 , 10 "8 , 10 "9 , 10 "10 , 10 "11 , 10 "12 , 10 ⁇ 13 M.
  • the ABP has an IC 50 for blocking the binding of LDLR to PCSK9 (D374Y, high affinity variant) of less than 1 microM, 1000 nM to 100 nM, ⁇ to 10 nM, ⁇ to 1 nM, ⁇ to 500pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM.
  • IC 50 for blocking the binding of LDLR to PCSK9 D374Y, high affinity variant
  • IgG2 heavy chain constant domain of an anti-PCSK9 antibody of the present invention has the amino acid sequence as shown in SEQ ID NO: 154, FIG. 3K .
  • IgG4 heavy chain constant domain of an anti-PCSK9 antibody of the present invention has the amino acid sequence as shown in SEQ ID NO: 155, FIG. 3K .
  • One example of a kappa light chain constant domain of an anti-PC SK9 antibody has the amino acid sequence as shown in SEQ ID NO: 157, FIG. 3K .
  • lambda light chain constant domain of an anti-PCSK9 antibody has the amino acid sequence as shown in SEQ ID NO: 156, FIG. 3K .
  • Variable regions of immunoglobulin chains generally exhibit the same overall structure, comprising relatively conserved framework regions (FR) joined by three hypervariable regions, more often called “complementarity determining regions” or CDRs.
  • the CDRs from the two chains of each heavy chain/light chain pair mentioned above typically are aligned by the framework regions to form a structure that binds specifically with a specific epitope on the target protein (e.g., PCSK9).
  • target protein e.g., PCSK9
  • From N- terminal to C-terminal, naturally-occurring light and heavy chain variable regions both typically conform with the following order of these elements: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • a numbering system has been devised for assigning numbers to amino acids that occupy positions in each of these domains.
  • each of these variable regions can be attached to the above heavy and light chain constant regions to form a complete antibody heavy and light chain, respectively. Further, each of the so generated heavy and light chain sequences can be combined to form a complete antibody structure.
  • variable regions of the light and heavy chains of the antibodies that are provided and their corresponding amino acid sequences are summarized in TABLE 2.
  • each of 8A1 485/483 the exemplary variable heavy chains listed in Table 2 can be combined with any of the exemplary variable light chains shown in Table 2 to form an antibody.
  • Table 2 shows exemplary light and heavy chain pairings found in several of the antibodies disclosed herein.
  • the antibodies include at least one variable heavy chain and one variable light chain from those listed in Table 2.
  • the antibodies contain two identical light chains and two identical heavy chains.
  • an antibody or antigen binding protein can include a heavy chain and a light chain, two heavy chains, or two light chains.
  • the antigen binding protein comprises (and/or consists) of 1, 2, and/or 3 heavy and/or light CDRs from at least one of the sequences listed in Table 2 (CDRs for the sequences are outlined in FIGs. 2A-3D, and other embodiments in FIGs. 3CCC-3JJJ and 3KK and 13A-13J).
  • CDRs for the sequences are outlined in FIGs. 2A-3D, and other embodiments in FIGs. 3CCC-3JJJ and 3KK and 13A-13J).
  • all 6 CDRs CDRl-3 from the light (CDRLl, CDRL2, CDRL3) and CDRl-3 from the heavy (CDRHl, CDRH2, and CDRH3)) are part of the ABP.
  • 1, 2, 3, 4, 5, or more CDRs are included in the ABP.
  • one heavy and one light CDR from the CDRs in the sequences in Table 2 is included in the ABP (CDRs for the sequences in Table 2 are outlined in FIGs. 2A-3D).
  • CDRs for the sequences in Table 2 are outlined in FIGs. 2A-3D.
  • additional sections e.g., as depicted in FIG. 2A-2D, 3A-3D, and other embodiments in 3CCC-3JJJ and 3LLL and 13A-13J
  • Examples of CDRs and FRs for the heavy and light chains noted in Table 2 are outlined in FIGs. 2A-3D (and other embodiments in FIGs. 3CCC-3JJJ and 3LLL and 13 A- J).
  • Optional light chain variable sequences can be selected from the following: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 28, 30, 31, 32, 33, 35, 36, 37, 38, 39, 40, 42, 44, 46, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, 461,465, 469, 473, 477, 481, and 485.
  • Optional heavy chain variable sequences can be selected from the following: 74, 85, 71 , 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81,60, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, 463, 467, 471, 475, 479, and 483.
  • 3CCC-3JJJ and 3LLL variations of the sequences or alternative boundaries of the CDRs and FRs are identified. These alternatives are identified with a "vl" following the ABP name. As most of these alternatives are minor in nature, only sections with differences are displayed in the table. It is understood that the remaining section of the light or heavy chain is the same as shown for the base ABP in the other panels. Thus, for example, 19H9vl in FIG. 2C has the same FR1, CDR1, and FR2 as 19H9 in FIG. 2A as the only difference is noted in FIG. 2C.
  • additional alternative nucleic acid sequences are provided in the figures. As will be appreciated by one of skill in the art, no more than one such sequence need actually be used in the creation of an antibody or ABP. Indeed, in some embodiments, only one or neither of the specific heavy or light chain nucleic acids need be present.
  • the antibodies useful in the methods described herein include the antibodies provided in U.S.P. 8,030,457 or U.S.P. 8,168,762. Further examples of antibodies that are useful in the methods described herein are provided in e.g., U.S.P. 8,188,233, U.S.P. 8,188,234, U.S.P. 8,080,243, U.S.P. 8,062,640, WO 2008/06332, WO 2009/055783, WO 201 1/053759, WO 2012/054438, WO 2012/088313, WO2012/109530, and WO 2013/039958.
  • the ABP is encoded by a nucleic acid sequence that can encode any of the protein sequences in Table 2.
  • the ABP binds selectively to the form of PCSK9 that binds to LDLR (e.g., the autocatalyzed form of the molecule).
  • the antigen binding protein does not bind to the c-terminus of the catalytic domain (e.g., the 5. 5-10, 10-15, 15-20, 20-25, 25-30, 30-40 most amino acids in the c-terminus).
  • the antigen binding protein does not bind to the n-terminus of the catalytic domain (e.g., the 5. 5-10, 10-15, 15-20, 20-25, 25-30, 30-40 most amino acids in the n-terminus).
  • the ABP binds to amino acids within amino acids 1-100 of the mature form of PCSK9. In some embodiments, the ABP binds to amino acids within (and/or amino acid sequences consisting of) amino acids 31-100, 100-200, 31-152, 153-692, 200-300, 300-400, 452-683, 400-500, 500-600, 31-692, 31- 449, and/or 600-692. In some embodiments, the ABP binds to the catalytic domain. In some embodiments, the neutralizing and/or non-neutralizing ABP binds to the prodomain. In some embodiments, the ABP binds to both the catalytic and pro domains.
  • the ABP binds to the catalytic domain so as to obstruct an area on the catalytic domain that interacts with the pro domain. In some embodiments, the ABP binds to the catalytic domain at a location or surface that the pro-domain interacts with as outlined in Piper et al. (Structure 15 : 1-8 (2007), the entirety of which is hereby incorporated by reference, including the structural representations therein). In some embodiments, the ABP binds to the catalytic domain and restricts the mobility of the prodomain. In some embodiments, the ABP binds to the catalytic domain without binding to the pro-domain.
  • the ABP binds to the catalytic domain, without binding to the pro-domain, while preventing the pro-domain from reorienting to allow PCSK9 to bind to LDLR. In some embodiments, the ABP binds in the same epitope as those surrounding residues 149- 152 of the pro-domain in Piper et al. In some embodiments, the ABPs bind to the groove (as outlined in Piper et al.) on the V domain. In some embodiments, the ABPs bind to the histidine-rich patch proximal to the groove on the V domain. In some embodiments, such antibodies (that bind to the V domain) are not neutralizing.
  • antibodies that bind to the V domain are neutralizing.
  • the neutralizing ABPs prevent the binding of PCSK9 to LDLR.
  • the neutralizing ABPs while preventing the PCSK9 degradation of LDLR, do not prevent the binding of PCSK9 to LDLR (for example ABP 31A4).
  • the ABP binds to or blocks at least one of the histidines depicted in Figure 4 of the Piper et al. paper.
  • the ABP blocks the catalytic triad in PCSK9.
  • the antibody binds selectively to variant PCSK9 proteins, e.g., D374Y over wild type PCSK9.
  • these antibodies bind to the variant at least twice as strongly as the wild type, and preferably 2-5, 5-10, 10-100, 100-1000, 1000-10,000 fold or more to the mutant than the wild type (as measured via a IQ).
  • the antibody selectively inhibits variant D374Y PCSK9 from interacting with LDLR over wild type PCSK9's ability to interact with LDLR.
  • these antibodies block the variant's ability to bind to LDLR more strongly than the wild type's ability, e.g., at least twice as strongly as the wild type, and preferably 2-5, 5-10, 10-100, 100-1000 fold or more to the mutant than the wild type (as measured via an IC 50 ).
  • the antibody binds to and neutralizes both wild type PCSK9 and variant forms of PCSK9, such as D374Y at similar levels.
  • the antibody binds to PCSK9 to prevent variants of LDLR from binding to PCSK9.
  • the variants of LDLR are at least 50% identical to human LDLR.
  • variants of LDLR are known to those of skill in the art ⁇ e.g., Brown MS et al, "Calcium cages, acid baths and recycling receptors” Nature 388: 629-630, 1997).
  • the ABP can raise the level of effective LDLR in heterozygote familial hypercholesterolemia (where a loss-of function variant of LDLR is present).
  • the ABP binds to (but does not block) variants of PCSK9 that are at least 50%, 50-60, 60-70, 70-80, 80-90, 90-95, 95-99, or greater percent identity to the form of PCSK9 depicted in FIG. 1A and/or FIG. IB. In some embodiments, the ABP binds to (but does not block) variants of PCSK9 that are at least 50%, 50-60, 60-70, 70-80, 80-90, 90-95, 95-99, or greater percent identity to the mature form of PCSK9 depicted in FIG. 1A and/or FIG. IB.
  • the ABP binds to and prevents variants of PCSK9 that are at least 50%, 50-60, 60-70, 70-80, 80-90, 90-95, 95-99, or greater percent identity to the form of PCSK9 depicted in FIG. 1A and/or FIG. IB from interacting with LDLR. In some embodiments, the ABP binds to and prevents variants of PCSK9 that are at least 50, 50-60, 60-70, 70-80, 80-90, 90-95, 95-99, or greater percent identity to the mature form of PCSK9 depicted in FIG. IB from interacting with LDLR.
  • the variant of PCSK9 is a human variant, such as variants at position 474, E620G, and/or E670G.
  • the amino acid at position 474 is valine (as in other humans) or threonine (as in cyno and mouse). Given the cross-reactivity data presented herein, it is believed that the present antibodies will readily bind to the above variants.
  • the ABP binds to an epitope bound by one of the antibodies described in Table 2.
  • the antigen binding proteins bind to a specific conformational state of PCSK9 so as to prevent PCSK9 from interacting with LDLR.
  • Humanized Antigen Binding Proteins e.g.. Antibodies
  • an antigen binding protein to PCSK9 can comprise a humanized antibody and/or part thereof.
  • An important practical application of such a strategy is the "humanization" of the mouse humoral immune system.
  • a humanized antibody is substantially non- immunogenic in humans.
  • a humanized antibody has substantially the same affinity for a target as an antibody from another species from which the humanized antibody is derived. See, e.g., U.S. Patent 5,530,101, U.S. Patent 5,693,761; U.S. Patent 5,693,762; U.S. Patent 5,585,089.
  • amino acids of an antibody variable domain that can be modified without diminishing the native affinity of the antigen binding domain while reducing its immunogenicity are identified. See, e.g., U.S. Patent Nos. 5,766,886 and 5,869,619.
  • modification of an antibody by methods known in the art is typically designed to achieve increased binding affinity for a target and/or to reduce immunogenicity of the antibody in the recipient.
  • humanized antibodies are modified to eliminate glycosylation sites in order to increase affinity of the antibody for its cognate antigen. See, e.g., Co et al, Mol. Immunol., 30: 1361-1367 (1993).
  • techniques such as "reshaping,” “hyperchimerization,” or “veneering/resurfacing” are used to produce humanized antibodies. See, e.g., Vaswami et al, Annals of Allergy, Asthma, & Immunol.
  • such techniques typically reduce antibody immunogenicity by reducing the number of foreign residues, but do not prevent anti- idiotypic and anti-allotypic responses following repeated administration of the antibodies.
  • Certain other methods for reducing immunogenicity are described, e.g., in Gilliland et al, J. Immunol ., 62(6): 3663-71 (1999).
  • humanizing antibodies results in a loss of antigen binding capacity.
  • humanized antibodies are "back mutated.”
  • the humanized antibody is mutated to include one or more of the amino acid residues found in the donor antibody. See, e.g., Saldanha et al, Mol Immunol 36:709-19 (1999).
  • the complementarity determining regions (CDRs) of the light and heavy chain variable regions of an antibody to PCSK9 can be grafted to framework regions (FRs) from the same, or another, species.
  • the CDRs of the light and heavy chain variable regions of an antibody to PCSK9 can be grafted to consensus human FRs.
  • consensus human FRs In certain embodiments, FRs from several human heavy chain or light chain amino acid sequences are aligned to identify a consensus amino acid sequence.
  • the FRs of an antibody to PCSK9 heavy chain or light chain are replaced with the FRs from a different heavy chain or light chain.
  • rare amino acids in the FRs of the heavy and light chains of an antibody to PCSK9 are not replaced, while the rest of the FR amino acids are replaced. Rare amino acids are specific amino acids that are in positions in which they are not usually found in FRs.
  • the grafted variable regions from an antibody to PCSK9 can be used with a constant region that is different from the constant region of an antibody to PCSK9. In certain embodiments, the grafted variable regions are part of a single chain Fv antibody. CDR grafting is described, e.g., in U.S. Patent Nos.
  • Human Antigen Binding Proteins e.g.. Antibodies
  • an antigen binding protein that binds to PCSK9 can comprise a human ⁇ i.e., fully human) antibody and/or part thereof.
  • nucleotide sequences encoding, and amino acid sequences comprising, heavy and light chain immunoglobulin molecules, particularly sequences corresponding to the variable regions are provided.
  • sequences corresponding to complementarity determining regions (CDR's), specifically from CDRl through CDR3, are provided.
  • a hybridoma cell line expressing such an immunoglobulin molecule is provided.
  • a hybridoma cell line expressing such a monoclonal antibody is provided.
  • a hybridoma cell line is selected from at least one of the cell lines described in Table 2, e.g., 21B12, 16F12 and 31H4.
  • a purified human monoclonal antibody to human PCSK9 is provided.
  • Human antibodies avoid some of the problems associated with antibodies that possess murine or rat variable and/or constant regions.
  • the presence of such murine or rat derived proteins can lead to the rapid clearance of the antibodies or can lead to the generation of an immune response against the antibody by a patient.
  • fully human antibodies can be generated through the introduction of functional human antibody loci into a rodent, other mammal or animal so that the rodent, other mammal or animal produces fully human antibodies.
  • Humanized antibodies are those antibodies that, while initially starting off containing antibody amino acid sequences that are not human, have had at least some of these nonhuman antibody amino acid sequences replaced with human antibody sequences. This is in contrast with human antibodies, in which the antibody is encoded (or capable of being encoded) by genes possessed a human.
  • Antigen Binding Protein Variants are those antibodies that, while initially starting off containing antibody amino acid sequences that are not human, have had at least some of these nonhuman antibody amino acid sequences replaced with human antibody sequences. This is in contrast with human antibodies, in which the antibody is encoded (or capable of being encoded) by genes possessed a human.
  • such antibodies include at least one heavy chain and one light chain, whereas in other instances the variant forms contain two identical light chains and two identical heavy chains (or subparts thereof).
  • the sequence comparison in FIG. 2A-3D (and 13A-13J, other embodiments in FIGs. 31A and 3 IB) can be used in order to identify sections of the antibodies that can be modified by observing those variations that impact binding and those variations that do not appear to impact binding. For example, by comparing similar sequences, one can identify those sections (e.g., particular amino acids) that can be modified and how they can be modified while still retaining (or improving) the functionality of the ABP.
  • variants of ABPs include those consensus groups and sequences depicted in FIGs.
  • the CDRs shown in FIGs. 13 A, 13C, 13F, 13G, 31A and 3 IB were defined based upon a hybrid combination of the Chothia method (based on the location of the structural loop regions, see, e.g., "Standard conformations for the canonical structures of immunoglobulins," Bissan Al-Lazikani, Arthur M.
  • an antigen binding protein comprises a heavy chain comprising a variable region comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, 60, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, 463, 467, 471, 475, 479, and 483.
  • an antigen binding protein comprises a heavy chain comprising a variable region comprising an amino acid sequence at least 95% identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, 60, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, 463, 467, 471, 475, 479, and 483.
  • an antigen binding protein comprises a heavy chain comprising a variable region comprising an amino acid sequence at least 99% identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, 60, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, 463, 467, 471, 475, 479, and 483.
  • the antigen binding protein comprises a sequence that is at least 90%, 90-95%, and/or 95-99% identical to one or more CDRs from the CDRs in at least one of sequences of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, 60, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, 463, 467, 471, 475, 479, and 483.
  • 1, 2, 3, 4, 5, or 6 CDR (each being at least 90%, 90-95%, and/or 95-99% identical to the above sequences) is present.
  • the antigen binding protein comprises a sequence that is at least 90%, 90-95%, and/or 95-99% identical to one or more FRs from the FRs in at least one of sequences of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, 60, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, 463, 467, 471, 475, 479, and 483.
  • 1, 2, 3, or 4 FR (each being at least 90%, 90-95%, and/or 95-99% identical to the above sequences) is present.
  • an antigen binding protein comprises a light chain comprising a variable region comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 28, 30, 31, 32, 33, 35, 36, 37, 38, 39, 40, 42, 44, 46, 421, 425, 429, 433, 437, 441, 445, 49, 453, 457, 461, 465, 469, 473, 477, 481, and 485.
  • an antigen binding protein comprises a light chain comprising a variable region comprising an amino acid sequence at least 95% identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 28, 30, 31, 32, 33, 35, 36, 37, 38, 39, 40, 42, 44, and 46.
  • an antigen binding protein comprises a light chain comprising a variable region comprising an amino acid sequence at least 99% identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 28, 30, 31, 32, 33, 35, 36, 37, 38, 39, 40, 42, 44, 46, 421, 425, 429, 433, 437, 441, 445, 49, 453, 457, 461, 465, 469, 473, 477, 481, and 485.
  • the antigen binding protein comprises a sequence that is at least 90%, 90-95%, and/or 95-99% identical to one or more CDRs from the CDRs in at least one of sequences of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21,
  • 1, 2, 3, 4, 5, or 6 CDR (each being at least 90%, 90-95%, and/or 95-99% identical to the above sequences) is present.
  • the antigen binding protein comprises a sequence that is at least 90%>, 90-95%>, and/or 95-99%) identical to one or more FRs from the FRs in at least one of sequences of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21, 22,
  • 1, 2, 3, or 4 FR (each being at least 90%, 90-95%, and/or 95-99% identical to the above sequences) is present.
  • a skilled artisan will be able to determine suitable variants of the ABPs as set forth herein using well-known techniques.
  • one skilled in the art can identify suitable areas of the molecule that may be changed without destroying activity by targeting regions not believed to be important for activity.
  • One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar ABPs. In view of such information, one skilled in the art can predict the alignment of amino acid residues of an antibody with respect to its three dimensional structure. In certain embodiments, one skilled in the art can choose not to make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues can be involved in important interactions with other molecules. Moreover, one skilled in the art can generate test variants containing a single amino acid substitution at each desired amino acid residue. The variants can then be screened using activity assays known to those skilled in the art. Such variants can be used to gather information about suitable variants.
  • Additional methods of predicting secondary structure include “threading” (Jones, D., Curr. Opin. Struct. Biol, 7(3):377-87 (1997); Sippl et al, Structure, 4(1): 15-19 (1996)), “profile analysis” (Bowie et al, Science, 253: 164-170 (1991); Gribskov et al, Meth. Enzym., 183: 146-159 (1990); Gribskov et al, Proc. Nat. Acad. Sci. USA, 84(13):4355-4358 (1987)), and “evolutionary linkage” (See Holm, supra (1999), and Brenner, supra (1997)).
  • antigen binding protein variants include glycosylation variants wherein the number and/or type of glycosylation site has been altered compared to the amino acid sequences of a parent polypeptide.
  • protein variants comprise a greater or a lesser number of N-linked glycosylation sites than the native protein.
  • An N-linked glycosylation site is characterized by the sequence: Asn-X-Ser or Asn-X-Thr, wherein the amino acid residue designated as X can be any amino acid residue except proline.
  • the substitution of amino acid residues to create this sequence provides a potential new site for the addition of an N-linked carbohydrate chain. Alternatively, substitutions which eliminate this sequence will remove an existing N-linked carbohydrate chain.
  • N-linked carbohydrate chains wherein one or more N- linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N-linked sites are created.
  • Additional preferred antibody variants include cysteine variants wherein one or more cysteine residues are deleted from or substituted for another amino acid (e.g., serine) as compared to the parent amino acid sequence. Cysteine variants can be useful when antibodies must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines.
  • amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and/or (4) confer or modify other physicochemical or functional properties on such polypeptides.
  • single or multiple amino acid substitutions in certain embodiments, conservative amino acid substitutions can be made in the naturally-occurring sequence (in certain embodiments, in the portion of the polypeptide outside the domain(s) forming intermolecular contacts).
  • a conservative amino acid substitution typically may not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).
  • a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence.
  • Examples of art- recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)); Introduction to Protein Structure (C. Branden & J. Tooze, eds., Garland Publishing, New York, N.Y. (1991)); and Thornton et al, Nature, 354: 105 (1991), which are each incorporated herein by reference.
  • the variants are variants of the nucleic acid sequences of the ABPs disclosed herein.
  • the above discussion can be used for identifying, evaluating, and/creating ABP protein variants and also for nucleic acid sequences that can encode for those protein variants.
  • nucleic acid sequences encoding for those protein variants are contemplated.
  • an ABP variant can have at least 80, 80-85, 85-90, 90-95, 95-97, 97-99 or greater identity to at least one nucleic acid sequence described in SEQ ID NOs: 152, 153, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 296, 418, 420, 422, 424, 426, 428, 430, 432, 434,
  • the antibody (or nucleic acid sequence encoding it) is a variant if the nucleic acid sequence that encodes the particular ABP (or the nucleic acid sequence itself) can selectively hybridize to any of the nucleic acid sequences that encode the proteins in Table 2 (such as, but not limited to SEQ ID NO: 152, 153, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150,
  • suitable moderately stringent conditions include prewashing in a solution of 5XSSC; 0.5% SDS, 1.0 mM EDTA (pH 8:0); hybridizing at 50° C, -65° C, 5xSSC, overnight or, in the event of cross-species homology, at 45° C with 0.5xSSC; followed by washing twice at 65° C for 20 minutes with each of 2x, 0.5x and 0.2xSSC containing 0.1% SDS.
  • hybridizing DNA sequences are also within the scope of this invention, as are nucleotide sequences that, due to code degeneracy, encode an antibody polypeptide that is encoded by a hybridizing DNA sequence and the amino acid sequences that are encoded by these nucleic acid sequences.
  • variants of CDRs include nucleic acid sequences and the amino acid sequences encoded by those sequences, that hybridize to one or more of the CDRs within the sequences noted above (individual CDRs can readily be determined in light of FIGs. 2A-3D, and other embodiments in FIGs. 3CCC-3JJJ and 15A-15D).
  • the phrase "selectively hybridize” referred to in this context means to detectably and selectively bind.
  • Polynucleotides, oligonucleotides and fragments thereof in accordance with the invention selectively hybridize to nucleic acid strands under hybridization and wash conditions that minimize appreciable amounts of detectable binding to nonspecific nucleic acids.
  • nucleic acid sequence homology between the polynucleotides, oligonucleotides, and fragments of the invention and a nucleic acid sequence of interest will be at least 80%, and more typically with preferably increasing homologies of at least 85%, 90%, 95%, 99%, and 100%.
  • Two amino acid sequences are homologous if there is a partial or complete identity between their sequences. For example, 85% homology means that 85% of the amino acids are identical when the two sequences are aligned for maximum matching.
  • Gaps in either of the two sequences being matched are allowed in maximizing matching; gap lengths of 5 or less are preferred with 2 or less being more preferred.
  • two protein sequences or polypeptide sequences derived from them of at least 30 amino acids in length are homologous, as this term is used herein, if they have an alignment score of at more than 5 (in standard deviation units) using the program ALIGN with the mutation data matrix and a gap penalty of 6 or greater. See Dayhoff, M. O., in Atlas of Protein Sequence and Structure, pp. 101-1 10 (Volume 5, National Biomedical Research Foundation (1972)) and Supplement 2 to this volume, pp. 1-10.
  • the two sequences or parts thereof are more preferably homologous if their amino acids are greater than or equal to 50% identical when optimally aligned using the ALIGN program.
  • the term "corresponds to” is used herein to mean that a polynucleotide sequence is homologous (i.e., is identical, not strictly evolutionarily related) to all or a portion of a reference polynucleotide sequence, or that a polypeptide sequence is identical to a reference polypeptide sequence.
  • the term “complementary to” is used herein to mean that the complementary sequence is homologous to all or a portion of a reference polynucleotide sequence.
  • the nucleotide sequence "TATAC” corresponds to a reference sequence "TATAC” and is complementary to a reference sequence "GTATA".
  • Antigen Binding Proteins e.g. Antibodies
  • antigen binding proteins are produced by immunization with an antigen (e.g., PCSK9).
  • an antigen e.g., PCSK9
  • antibodies can be produced by immunization with full-length PCSK9, a soluble form of PCSK9, the catalytic domain alone, the mature form of PCSK9 shown in FIG. 1A, a splice variant form of PCSK9, or a fragment thereof.
  • the antibodies of the invention can be polyclonal or monoclonal, and/or can be recombinant antibodies.
  • antibodies of the invention are human antibodies prepared, for example, by immunization of transgenic animals capable of producing human antibodies (see, for example, PCT Published Application No. W0 93/12227).
  • certain strategies can be employed to manipulate inherent properties of an antibody, such as the affinity of an antibody for its target.
  • Such strategies include, but are not limited to, the use of site-specific or random mutagenesis of the polynucleotide molecule encoding an antibody to generate an antibody variant.
  • such generation is followed by screening for antibody variants that exhibit the desired change, e.g. increased or decreased affinity.
  • the amino acid residues targeted in mutagenic strategies are those in the CDRs.
  • amino acids in the framework regions of the variable domains are targeted.
  • such framework regions have been shown to contribute to the target binding properties of certain antibodies. See, e.g., Hudson, Curr. Opin. Biotech., 9:395-402 (1999) and references therein.
  • smaller and more effectively screened libraries of antibody variants are produced by restricting random or site-directed mutagenesis to hyper-mutation sites in the CDRs, which are sites that correspond to areas prone to mutation during the somatic affinity maturation process. See, e.g., Chowdhury & Pastan, Nature Biotech., 17: 568-572 (1999) and references therein.
  • certain types of DNA elements can be used to identify hyper-mutation sites including, but not limited to, certain direct and inverted repeats, certain consensus sequences, certain secondary structures, and certain palindromes.
  • DNA elements that can be used to identify hyper-mutation sites include, but are not limited to, a tetrabase sequence comprising a purine (A or G), followed by guainine (G), followed by a pyrimidine (C or T), followed by either adenosine or thymidine (A or T) (i.e., A/G-G-C/T-A/T).
  • a or T a tetrabase sequence
  • a or G guainine
  • C or T pyrimidine
  • a or T adenosine or thymidine
  • Another example of a DNA element that can be used to identify hyper-mutation sites is the serine codon, A-G-C/T.
  • a phage display technique is used to generate monoclonal antibodies. In certain embodiments, such techniques produce fully human monoclonal antibodies.
  • a polynucleotide encoding a single Fab or Fv antibody fragment is expressed on the surface of a phage particle. See, e.g., Hoogenboom et al, J. Mol. Biol, 227: 381 (1991); Marks et al, J Mol Biol 222: 581 (1991); U.S. Patent No. 5,885,793.
  • phage are "screened" to identify those antibody fragments having affinity for target.
  • antibodies of the invention are prepared through the utilization of a transgenic mouse that has a substantial portion of the human antibody producing genome inserted but that is rendered deficient in the production of endogenous, murine antibodies.
  • mice are capable of producing human immunoglobulin molecules and antibodies and are deficient in the production of murine immunoglobulin molecules and antibodies. Technologies utilized for achieving this result are disclosed in the patents, applications and references disclosed in the specification, herein. In certain embodiments, one can employ methods such as those disclosed in PCT Published Application No. WO 98/24893 or in Mendez et al, Nature Genetics, 15: 146-156 (1997), which are hereby incorporated by reference for any purpose.
  • Fully human monoclonal ABPs ⁇ e.g., antibodies) specific for PCSK9 can be produced as follows.
  • Transgenic mice containing human immunoglobulin genes are immunized with the antigen of interest, e.g. PCSK9, lymphatic cells (such as B- cells) from the mice that express antibodies are obtained.
  • lymphatic cells such as B- cells
  • Such recovered cells are fused with a myeloid-type cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to the antigen of interest.
  • the production of a hybridoma cell line that produces antibodies specific to PCSK9 is provided.
  • fully human antibodies are produced by exposing human splenocytes (B or T cells) to an antigen in vitro, and then reconstituting the exposed cells in an immunocompromised mouse, e.g. SCID or nod/SCID.
  • SCID immunocompromised mouse
  • engraftment of human fetal tissue into SCID mice results in long-term hematopoiesis and human T-cell development.
  • transplanted cells when such transplanted cells are treated either with a priming agent, such as Staphylococcal Enterotoxin A (SEA), or with anti-human CD40 monoclonal antibodies, higher levels of B cell production is detected.
  • SEA Staphylococcal Enterotoxin A
  • SEA Staphylococcal Enterotoxin A
  • Fully human antibodies can be produced by the expression of recombinant DNA in host cells or by expression in hybridoma cells.
  • antibodies can be produced using the phage display techniques described herein.
  • mice are capable of producing human immunoglobulin molecules and antibodies and are deficient in the production of murine immunoglobulin molecules and antibodies. Technologies utilized for achieving the same are disclosed in the patents, applications, and references disclosed in the background section herein. In particular, however, a preferred embodiment of transgenic production of mice and antibodies therefrom is disclosed in U.S. Patent Application Serial No. 08/759,620, filed December 3, 1996 and International Patent Application Nos. WO 98/24893, published June 11, 1998 and WO 00/76310, published December 21, 2000, the disclosures of which are hereby incorporated by reference. See also Mendez et ah, Nature Genetics, 15: 146-156 (1997), the disclosure of which is hereby incorporated by reference.
  • XenoMouse ® lines of mice are immunized with an antigen of interest ⁇ e.g. PCSK9), lymphatic cells (such as B-cells) are recovered from the hyper-immunized mice, and the recovered lymphocytes are fused with a myeloid-type cell line to prepare immortal hybridoma cell lines.
  • lymphatic cells such as B-cells
  • myeloid-type cell line to prepare immortal hybridoma cell lines.
  • These hybridoma cell lines are screened and selected to identify hybridoma cell lines that produced antibodies specific to the antigen of interest.
  • Provided herein are methods for the production of multiple hybridoma cell lines that produce antibodies specific to PCSK9. Further, provided herein are characterization of the antibodies produced by such cell lines, including nucleotide and amino acid sequence analyses of the heavy and light chains of such antibodies.
  • minilocus In an alternative approach, others, including GenPharm International, Inc., have utilized a "minilocus" approach. In the minilocus approach, an exogenous Ig locus is mimicked through the inclusion of pieces (individual genes) from the Ig locus. Thus, one or more VH genes, one or more D H genes, one or more JH genes, a mu constant region, and usually a second constant region (preferably a gamma constant region) are formed into a construct for insertion into an animal. This approach is described in U.S. Patent No. 5,545,807 to Surani et al. and U.S. Patent Nos.
  • Kirin has also demonstrated the generation of human antibodies from mice in which, through microcell fusion, large pieces of chromosomes, or entire chromosomes, have been introduced. See European Patent Application Nos. 773 288 and 843 961, the disclosures of which are hereby incorporated by reference. Additionally, KMTM mice, which are the result of cross-breeding of Kirin's Tc mice with Medarex's minilocus (Humab) mice have been generated. These mice possess the human IgH transchromosome of the Kirin mice and the kappa chain transgene of the Genpharm mice (Ishida et al, Cloning Stem Cells, (2002) 4:91-102).
  • Human antibodies can also be derived by in vitro methods. Suitable examples include but are not limited to phage display (CAT, Morphosys, Dyax, Biosite/Medarex, Xoma, Symphogen, Alexion (formerly Proliferon), Affimed) ribosome display (CAT), yeast display, and the like.
  • the antibodies described herein possess human IgG4 heavy chains as well as IgG2 heavy chains.
  • Antibodies can also be of other human isotypes, including IgGl .
  • the antibodies possessed high affinities, typically possessing a Kd of from about 10 "6 through about 10 "13 M or below, when measured by various techniques.
  • antibodies can be expressed in cell lines other than hybridoma cell lines. Sequences encoding particular antibodies can be used to transform a suitable mammalian host cell.
  • Transformation can be by any known method for introducing polynucleotides into a host cell, including, for example packaging the polynucleotide in a virus (or into a viral vector) and transducing a host cell with the virus (or vector) or by transfection procedures known in the art, as exemplified by U.S. Patent Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (which patents are hereby incorporated herein by reference). The transformation procedure used depends upon the host to be transformed.
  • Methods for introducing heterologous polynucleotides into mammalian cells include dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
  • Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), human epithelial kidney 293 cells, and a number of other cell lines. Cell lines of particular preference are selected through determining which cell lines have high expression levels and produce antibodies with constitutive PCSK9 binding properties.
  • ATCC American Type Culture Collection
  • antibodies and/or ABP are produced by at least one of the following hybridomas: 2 IB 12, 31H4, 16F12, any of the other hybridomas listed in Table 2 or disclosed in the examples.
  • antigen binding proteins bind to PCSK9 with a dissociation constant (K D ) of less than approximately 1 nM, e.g., ⁇ to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM, and/or 1 pM to 0.1 pM or less.
  • antigen binding proteins comprise an immunoglobulin molecule of at least one of the IgGl, IgG2, IgG3, IgG4, Ig E, IgA, IgD, and IgM isotype.
  • antigen binding proteins comprise a human kappa light chain and/or a human heavy chain.
  • the heavy chain is of the IgGl, IgG2, IgG3, IgG4, IgE, IgA, IgD, or IgM isotype.
  • antigen binding proteins have been cloned for expression in mammalian cells.
  • antigen binding proteins comprise a constant region other than any of the constant regions of the IgGl, IgG2, IgG3, IgG4, IgE, IgA, IgD, and IgM isotype.
  • antigen binding proteins comprise a human lambda light chain and a human IgG2 heavy chain. In certain embodiments, antigen binding proteins comprise a human lambda light chain and a human IgG4 heavy chain. In certain embodiments, antigen binding proteins comprise a human lambda light chain and a human IgGl, IgG3, IgE, IgA, IgD or IgM heavy chain. In other embodiments, antigen binding proteins comprise a human kappa light chain and a human IgG2 heavy chain. In certain embodiments, antigen binding proteins comprise a human kappa light chain and a human IgG4 heavy chain.
  • antigen binding proteins comprise a human kappa light chain and a human IgGl, IgG3, IgE, IgA, IgD or IgM heavy chain.
  • antigen binding proteins comprise variable regions of antibodies ligated to a constant region that is neither the constant region for the IgG2 isotype, nor the constant region for the IgG4 isotype.
  • antigen binding proteins have been cloned for expression in mammalian cells.
  • conservative modifications to the heavy and light chains of antibodies from at least one of the hybridoma lines: 21B12, 31H4 and 16F12 will produce antibodies to PCSK9 having functional and chemical characteristics similar to those of the antibodies from the hybridoma lines: 21B12, 31H4 and 16F12.
  • substantial modifications in the functional and/or chemical characteristics of antibodies to PCSK9 can be accomplished by selecting substitutions in the amino acid sequence of the heavy and light chains that differ significantly in their effect on maintaining (a) the structure of the molecular 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.
  • a “conservative amino acid substitution” can involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position.
  • any native residue in the polypeptide can also be substituted with alanine, as has been previously described for "alanine scanning mutagenesis.”
  • amino acid substitutions can be used to identify important residues of antibodies to PCSK9, or to increase or decrease the affinity of the antibodies to PCSK9 as described herein.
  • antibodies of the present invention can be expressed in cell lines other than hybridoma cell lines.
  • sequences encoding particular antibodies can be used for transformation of a suitable mammalian host cell.
  • transformation can be by any known method for introducing polynucleotides into a host cell, including, for example packaging the polynucleotide in a virus (or into a viral vector) and transducing a host cell with the virus (or vector) or by transfection procedures known in the art, as exemplified by U.S. Pat. Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (which patents are hereby incorporated herein by reference for any purpose).
  • the transformation procedure used can depend upon the host to be transformed.
  • Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include, but are not limited to, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
  • Mammalian cell lines available as hosts for expression are well known in the art and include, but are not limited to, many immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), and a number of other cell lines.
  • ATCC American Type Culture Collection
  • cell lines can be selected through determining which cell lines have high expression levels and produce antibodies with constitutive HGF binding properties.
  • Appropriate expression vectors for mammalian host cells are well known.
  • antigen binding proteins comprise one or more polypeptides.
  • any of a variety of expression vector/host systems can be utilized to express polynucleotide molecules encoding polypeptides comprising one or more ABP components or the ABP itself.
  • Such systems include, but are not limited to, microorganisms, such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV, tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid); or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
  • yeast transformed with yeast expression vectors insect cell systems infected with virus expression vectors (e.g., baculovirus)
  • plant cell systems transfected with virus expression vectors e.g., cauliflower mosaic virus, CaMV, tobacco mosaic virus, TMV
  • bacterial expression vectors e.g., Ti or
  • a polypeptide comprising one or more ABP components or the ABP itself is recombinantly expressed in yeast.
  • Certain such embodiments use commercially available expression systems, e.g., the Pichia Expression System (Invitrogen, San Diego, CA), following the manufacturer's instructions.
  • Pichia Expression System Invitrogen, San Diego, CA
  • transcription of the insert is driven by the alcohol oxidase (AOX1) promoter upon induction by methanol.
  • a secreted polypeptide comprising one or more ABP components or the ABP itself is purified from yeast growth medium.
  • the methods used to purify a polypeptide from yeast growth medium is the same as those used to purify the polypeptide from bacterial and mammalian cell supernatants.
  • a nucleic acid encoding a polypeptide comprising one or more ABP components or the ABP itself is cloned into a baculovirus expression vector, such as pVL1393 (PharMingen, San Diego, CA).
  • a baculovirus expression vector such as pVL1393 (PharMingen, San Diego, CA).
  • such a vector can be used according to the manufacturer's directions (PharMingen) to infect Spodoptera frugiperda cells in sF9 protein-free media and to produce recombinant polypeptide.
  • a polypeptide is purified and concentrated from such media using a heparin-Sepharose column (Pharmacia).
  • a polypeptide comprising one or more ABP components or the ABP itself is expressed in an insect system.
  • Certain insect systems for polypeptide expression are well known to those of skill in the art.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae.
  • a nucleic acid molecule encoding a polypeptide can be inserted into a nonessential gene of the virus, for example, within the polyhedrin gene, and placed under control of the promoter for that gene.
  • successful insertion of a nucleic acid molecule will render the nonessential gene inactive.
  • that inactivation results in a detectable characteristic. For example, inactivation of the polyhedrin gene results in the production of virus lacking coat protein.
  • recombinant viruses can be used to infect S. frugiperda cells or Trichoplusia larvae. See, e.g., Smith et ah, J. Virol, 46: 584 (1983); Engelhard et al, Proc. Nat. Acad. Sci. (USA), 91 : 3224-7 (1994).
  • polypeptides comprising one or more ABP components or the ABP itself made in bacterial cells are produced as insoluble inclusion bodies in the bacteria.
  • host cells comprising such inclusion bodies are collected by centrifugation; washed in 0.15 M NaCl, 10 mM Tris, pH 8, 1 mM EDTA; and treated with 0.1 mg/ml lysozyme (Sigma, St. Louis, MO) for 15 minutes at room temperature.
  • the lysate is cleared by sonication, and cell debris is pelleted by centrifugation for 10 minutes at 12,000 X g.
  • the polypeptide-containing pellet is resuspended in 50 mM Tris, pH 8, and 10 mM EDTA; layered over 50% glycerol; and centrifuged for 30 minutes at 6000 X g.
  • that pellet can be resuspended in standard phosphate buffered saline solution (PBS) free of Mg ++ and Ca ++ .
  • PBS standard phosphate buffered saline solution
  • the polypeptide is further purified by fractionating the resuspended pellet in a denaturing SDS polyacrylamide gel (See, e.g., Sambrook et al., supra).
  • such a gel can be soaked in 0.4 M KC1 to visualize the protein, which can be excised and electroeluted in gel-running buffer lacking SDS.
  • a Glutathione-S-Transferase (GST) fusion protein is produced in bacteria as a soluble protein.
  • GST fusion protein is purified using a GST Purification Module (Pharmacia).
  • polypeptides comprising one or more ABP components or the ABP itself.
  • polypeptides are produced using certain recombinant systems discussed herein.
  • polypeptides are "refolded" and/or oxidized to form desired tertiary structure and/or to generate disulfide linkages.
  • structure and/or linkages are related to certain biological activity of a polypeptide.
  • refolding is accomplished using any of a number of procedures known in the art.
  • Exemplary methods include, but are not limited to, exposing the solubilized polypeptide agent to a pH typically above 7 in the presence of a chaotropic agent.
  • An exemplary chaotropic agent is guanidine.
  • the refolding/oxidation solution also contains a reducing agent and the oxidized form of that reducing agent.
  • the reducing agent and its oxidized form are present in a ratio that will generate a particular redox potential that allows disulfide shuffling to occur. In certain embodiments, such shuffling allows the formation of cysteine bridges.
  • Exemplary redox couples include, but are not limited to, cysteine/cystamine, glutathione/dithiobisGSH, cupric chloride, dithiothreitol DTT/dithiane DTT, and 2-mercaptoethanol (bME)/dithio-bME.
  • a co-solvent is used to increase the efficiency of refolding.
  • Exemplary cosolvents include, but are not limited to, glycerol, polyethylene glycol of various molecular weights, and arginine.
  • Certain protein purification techniques are known to those of skill in the art.
  • protein purification involves crude fractionation of polypeptide fractionations from non-polypeptide fractions.
  • polypeptides are purified using chromatographic and/or electrophoretic techniques.
  • Exemplary purification methods include, but are not limited to, precipitation with ammonium sulphate; precipitation with PEG; immunoprecipitation; heat denaturation followed by centrifugation; chromatography, including, but not limited to, affinity chromatography (e.g., Protein-A-Sepharose), ion exchange chromatography, exclusion chromatography, and reverse phase chromatography; gel filtration; hydroxyapatite chromatography; isoelectric focusing; polyacrylamide gel electrophoresis; and combinations of such and other techniques.
  • a polypeptide is purified by fast protein liquid chromatography or by high pressure liquid chromotography (HPLC).
  • purification steps can be changed or certain steps can be omitted, and still result in a suitable method for the preparation of a substantially purified polypeptide.
  • Certain methods for quantifying the degree of purification are known to those of skill in the art.
  • Certain exemplary methods include, but are not limited to, determining the specific binding activity of the preparation and assessing the amount of a polypeptide within a preparation by SDS/PAGE analysis.
  • Certain exemplary methods for assessing the amount of purification of a polypeptide preparation comprise calculating the binding activity of a preparation and comparing it to the binding activity of an initial extract. In certain embodiments, the results of such a calculation are expressed as "fold purification.” The units used to represent the amount of binding activity depend upon the particular assay performed.
  • a polypeptide comprising one or more ABP components or the ABP itself is partially purified.
  • partial purification can be accomplished by using fewer purification steps or by utilizing different forms of the same general purification scheme.
  • cation-exchange column chromatography performed utilizing an HPLC apparatus will generally result in a greater "fold purification" than the same technique utilizing a low-pressure chromatography system.
  • methods resulting in a lower degree of purification can have advantages in total recovery of polypeptide, or in maintaining binding activity of a polypeptide.
  • the electrophoretic migration of a polypeptide can vary, sometimes significantly, with different conditions of SDS/PAGE.
  • epitopes to which anti-PC SK9 antibodies useful in the methods provided herein bind are described.
  • epitopes that are bound by the presently disclosed antibodies are particularly useful.
  • antigen binding proteins that bind to any of the epitopes that are bound by the antibodies described herein are useful.
  • the epitopes bound by any of the antibodies listed in Table 2 and FIGs. 2 and 3 are especially useful.
  • the epitope is on the catalytic domain PCSK9.
  • antigen binding proteins disclosed herein bind specifically to N-terminal prodomain, a subtilisin-like catalytic domain and/or a C- terminal domain. In some embodiments, the antigen binding protein binds to the substrate -binding groove of PCSK-9 (described in Cunningham et al, incorporated herein in its entirety by reference).
  • the domain(s)/region(s) containing residues that are in contact with or are buried by an antibody can be identified by mutating specific residues in PCSK9 (e.g., a wild-type antigen) and determining whether the antigen binding protein can bind the mutated or variant PCSK9 protein.
  • PCSK9 e.g., a wild-type antigen
  • residues that play a direct role in binding or that are in sufficiently close proximity to the antibody such that a mutation can affect binding between the antigen binding protein and antigen can be identified. From knowledge of these amino acids, the domain(s) or region(s) of the antigen that contain residues in contact with the antigen binding protein or covered by the antibody can be elucidated.
  • Such a domain can include the binding epitope of an antigen binding protein.
  • This general approach utilizes an arginine/glutamic acid scanning protocol (see, e.g., Nanevicz, T., et al., 1995, J. Biol. Chem., 270:37, 21619-21625 and Zupnick, A., et al, 2006, J. Biol. Chem., 281 :29, 20464-20473).
  • arginine and glutamic acids are substituted (typically individually) for an amino acid in the wild- type polypeptide because these amino acids are charged and bulky and thus have the potential to disrupt binding between an antigen binding protein and an antigen in the region of the antigen where the mutation is introduced.
  • Arginines that exist in the wild- type antigen are replaced with glutamic acid.
  • a variety of such individual mutants are obtained and the collected binding results analyzed to determine what residues affect binding.
  • An alteration (for example a reduction or increase) in binding between an antigen binding protein and a variant PCSK9 as used herein means that there is a change in binding affinity ⁇ e.g., as measured by known methods such as Biacore testing or the bead based assay described below in the examples), EC 50 , and/or a change (for example a reduction) in the total binding capacity of the antigen binding protein (for example, as evidenced by a decrease in Bmax in a plot of antigen binding protein concentration versus antigen concentration).
  • a significant alteration in binding indicates that the mutated residue is directly involved in binding to the antigen binding protein or is in close proximity to the binding protein when the binding protein is bound to antigen.
  • a significant reduction in binding means that the binding affinity, EC50, and/or capacity between an antigen binding protein and a mutant PCSK9 antigen is reduced by greater than 10%, greater than 20%, greater than 40 %, greater than 50 %, greater than 55 %, greater than 60 %, greater than 65 %, greater than 70 %, greater than75 %, greater than 80 %, greater than 85 %, greater than 90% or greater than 95% relative to binding between the antigen binding protein and a wild type PCSK9 (e.g., shown in SEQ ID NO: 1 and/or SEQ ID NO: (303).
  • binding is reduced below detectable limits.
  • a significant reduction in binding is evidenced when binding of an antigen binding protein to a variant PCSK9 protein is less than 50%> (for example, less than 40%>, 35%, 30%), 25%), 20%), 15%) or 10%>) of the binding observed between the antigen binding protein and a wild-type PCSK9 protein (for example, the protein of SEQ ID NO: 1 and/or SEQ ID NO: (303).
  • binding measurements can be made using a variety of binding assays known in the art.
  • antigen binding proteins are provided that exhibit significantly lower binding for a variant PCSK9 protein in which a residue in a wild- type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303 is substituted with arginine or glutamic acid.
  • a residue in a wild- type PCSK9 protein e.g., SEQ ID NO: 1 or SEQ ID NO: 303 is substituted with arginine or glutamic acid.
  • binding of an antigen binding protein is significantly reduced or increased for a variant PCSK9 protein having any one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 244) of the following mutations: R207E, D208R, R185E, R439E, E513R, V538R, E539R, T132R, S351R, A390R, A413R, E582R, D162R, R164E, E167R, S 123R, E129R, A31 1R, D313R, D337R, R519E, H521R, and Q554R as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • the format is: Wild type residue: Position in polypeptide: Mutant residue, with the numbering of the residues as indicated in SEQ ID NO: lor SEQ ID NO: 303.
  • binding of an antigen binding protein is significantly reduced or increased for a mutant PCSK9 protein having one or more (e.g., 1 , 2, 3, 4, 5, or more) mutations at the following positions: 207, 208, 185, 181 , 439, 513, 538, 539, 132, 351 , 390, 413, 582, 162, 164, 167, 123, 129, 31 1 , 313, 337, 519, 521 , and 554, as shown in SEQ ID NO: 1 as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • a wild-type PCSK9 protein e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • binding of an antigen binding protein is reduced or increased for a mutant PCSK9 protein having one or more (e.g., 1 , 2, 3, 4, 5, or more) mutations at the following positions: 207, 208, 185, 181 , 439, 513, 538, 539, 132, 351 , 390, 413, 582, 162, 164, 167, 123, 129, 31 1 , 313, 337, 519, 521 , and 554, as shown in SEQ ID NO: 1 as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • a wild-type PCSK9 protein e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • binding of an antigen binding protein is substantially reduced or increased for a mutant PCSK9 protein having one or more (e.g., 1 , 2, 3, 4, 5, or more) mutations at the following positions: 207, 208, 185, 181 , 439, 513, 538, 539, 132, 351 , 390, 413, 582, 162, 164, 167, 123, 129, 31 1 , 313, 337, 519, 521 , and 554, within SEQ ID NO: 1 as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • a wild-type PCSK9 protein e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • binding of an ABP is significantly reduced or increased for a mutant PCSK9 protein having one or more (e.g., 1 , 2, 3, 4, 5, etc.) of the following mutations: R207E, D208R, R185E, R439E, E513R, V538R, E539R, T132R, S351R, A390R, A413R, E582R, D162R, R164E, E167R, S 123R, E129R, A31 1R, D313R, D337R, R519E, H521R, and Q554R within SEQ ID NO: 1 or SEQ ID NO: 303, as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303).
  • a wild-type PCSK9 protein e.g., SEQ ID NO: 1 or SEQ ID NO: 303.
  • binding of an ABP is significantly reduced or increased for a mutant PCSK9 protein having one or more (e.g., 1 , 2, 3, 4, 5, etc.) of the following mutations: R207E, D208R, R185E, R439E, E513R, V538R, E539R, T132R, S351R, A390R, A413R, and E582R within SEQ ID NO: 1 or SEQ ID NO: 303, as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303).
  • the binding is reduced.
  • the reduction in binding is observed as a change in EC50.
  • the change in EC50 is an increase in the numerical value of the EC50 (and thus is a decrease in binding).
  • binding of an ABP is significantly reduced or increased for a mutant PCSK9 protein having one or more (e.g., 1 , 2, 3, 4, 5, etc.) of the following mutations: D162R, R164E, E167R, S 123R, E129R, A31 1R, D313R, D337R, R519E, H521R, and Q554R within SEQ ID NO: 1 , as compared to a wild-type PCSK9 protein (e.g., SEQ ID NO: 1 or SEQ ID NO: 303).
  • the binding is reduced.
  • the reduction in binding is observed as a change in Bmax.
  • the shift in Bmax is a reduction of the maximum signal generated by the ABP.
  • the Bmax is reduced by at least 10%, for example, reductions of at least any of the following amounts: 20, 30, 40, 50, 60, 70, 80, 90, 95, 98, 99, or 100 percent can, in some embodiments, indicate that the residue is part of the epitope.
  • any of the above embodiments can be compared to an allelic sequence, rather than purely the wild-type sequence shown in FIG. 1 A
  • binding of an antigen binding protein is significantly reduced for a variant PCSK9 protein in which the residue at a selected position in the wild-type PCSK9 protein is mutated to any other residue.
  • the herein described arginine/glutamic acid replacements are used for the identified positions.
  • alanine is used for the identified positions.
  • an antigen binding protein binds to a domain containing at least one of amino acids: 207, 208, 185, 181, 439, 513, 538, 539, 132, 351, 390, 413, 582, 162, 164, 167, 123, 129, 311, 313, 337, 519, 521, and 554 of SEQ ID NO: 1 or SEQ ID NO: 303.
  • the antigen binding protein binds to a region containing at least one of amino acids 207, 208, 185, 181, 439, 513, 538, 539, 132, 351, 390, 413, 582, 162, 164, 167, 123, 129, 311, 313, 337, 519, 521, and 554 of SEQ ID NO: 1 or SEQ ID NO: 303.
  • the antigen binding protein binds to a region containing at least one of amino acids 162, 164, 167, 207 and/or 208 of SEQ ID NO: 1 or SEQ ID NO: 303. In some embodiments, more than one (e.g., 2, 3, 4, or 5) of the identified residues are part of the region that is bound by the ABP. In some embodiments, the ABP competes with ABP 2 IB 12.
  • the antigen binding protein binds to a region containing at least one of amino acid 185 of SEQ ID NO: 1 or SEQ ID NO: 303.
  • the ABP competes with ABP 31H4.
  • the antigen binding protein binds to a region containing at least one of amino acids 439, 513, 538, and/or 539 of SEQ ID NO: 1 or SEQ ID NO: 303. In some embodiments, more than one (e.g., 2, 3, or 4) of the identified residues are part of the region that is bound by the ABP. In some embodiments, the ABP competes with ABP 31 A4. In some embodiments, the antigen binding protein binds to a region containing at least one of amino acids 123, 129, 311, 313, 337, 132, 351, 390, and/or 413 of SEQ ID NO: 1 or SEQ ID NO: 303. In some embodiments, more than one (e.g., 2, 3, 4, 5, 6, 7, 8, or 9) of the identified residues are part of the region that is bound by the ABP. In some embodiments, the ABP competes with ABP 12H11.
  • the antigen binding protein binds to a region containing at least one of amino acid 582, 519, 521, and/or 554 of SEQ ID NO: 1 or SEQ ID NO: 303. In some embodiments, more than one (e.g., 2, 3, or 4) of the identified residues are part of the region that is bound by the ABP. In some embodiments, the ABP competes with ABP 3 C4.
  • the antigen binding proteins binds to the foregoing regions within a fragment or the full length sequence of SEQ ID NO: 1 or SEQ ID NO: 303. In other embodiments, antigen binding proteins bind to polypeptides consisting of these regions.
  • the reference to "SEQ ID NO: 1 or SEQ ID NO: 303" denotes that one or both of these sequences can be employed or relevant. The phrase does not denote that only one should be employed.
  • any amino acid position in SEQ ID NO: 1 will correspond to an amino acid position 30 amino acids further into the protein in SEQ ID NO: 3.
  • position 207 of SEQ ID NO: 1 corresponds to position 237 of SEQ ID NO: 3 (the full length sequence, and the numbering system used in the present specification generally).
  • ABP 2 IB 12 binds to an epitope including residues 162- 167 (e.g., residues D162-E167 of SEQ ID NO: 1).
  • ABP 12H1 1 binds to an epitope that includes residues 123-132 (e.g., S 123-T132 of SEQ ID NO: 1).
  • ABP 12H1 1 binds to an epitope that includes residues 31 1-313 (e.g., A31 1-D313 of SEQ ID NO: 1). In some embodiments, ABPs can bind to an epitope that includes any one of these strands of sequences.
  • antigen binding proteins are provided that compete with one of the exemplified antibodies or functional fragments binding to the epitope described herein for specific binding to PCSK9. Such antigen binding proteins can also bind to the same epitope as one of the herein exemplified antigen binding proteins, or an overlapping epitope. Antigen binding proteins and fragments that compete with or bind to the same epitope as the exemplified antigen binding proteins are expected to show similar functional properties.
  • the exemplified antigen binding proteins and fragments include those described above, including those with the heavy and light chains, variable region domains and CDRs included in TABLE 2 and/or FIGs. 2-3.
  • the antigen binding proteins that are provided include those that compete with an antibody or antigen binding protein having:
  • pharmaceutical formulations containing antigen binding proteins to PCSK9 that are useful in the described methods.
  • pharmaceutical formulation is a sterile composition of a pharmaceutically active drug, namely, at least one antigen binding protein to PCSK9, that is suitable for parenteral administration (including but not limited to intravenous, intramuscular, subcutaneous, aerosolized, intrapulmonary, intranasal, or intrathecal) to a patient in need thereof and includes only pharmaceutically acceptable excipients, diluents, and other additives deemed safe by the Federal Drug Administration or other foreign national authorities.
  • compositions include liquid, e.g., aqueous, solutions that may be directly administered, and lyophilized powders which may be reconstituted into solutions by adding a diluent before administration.
  • pharmaceutical formulation are compositions for topical administration to patients, compositions for oral ingestion, and compositions for parenteral feeding.
  • the pharmaceutical formulation is a stable pharmaceutical formulation.
  • stable pharmaceutical formulation, “stable formulation” or “a pharmaceutical formulation is stable” refers to a pharmaceutical formulation of biologically active proteins that exhibit increased aggregation and/or reduced loss of biological activity of not more than 5% when stored at 2-8°C for at least 1 month, or 2 months, or 3 months, or 6 months, or 1 year or 2 years compared with a control formula sample.
  • Formulation stability can be easily determed by a person of skill in the art using any number of standard assays, including but not limited to size exclusion HPLC ("SEC-HPLC”), cation-exchange HPLC (CEX- HPLC), Subvisible Particle Detection by Light Obscuration (“HIAC”) and/or visual inspection.
  • SEC-HPLC size exclusion HPLC
  • CEX- HPLC cation-exchange HPLC
  • HIAC Subvisible Particle Detection by Light Obscuration
  • the pharmaceutical formulation comprises any of the antigen binding proteins to PCSK9 comprising: one or more heavy chain complementary determining regions (CDRHs) and one or more light chain complementary determining regions (CDRLs) depicted in Table 2 and FIGs. 2 and/or 3 and FIGS. 31A and 3 IB.
  • the pharmaceutical formulation comprises an antigen binding protein to PCSK9 comprising: a light chain variable region that comprises an amino acid sequence that is at least 90% identical the antigen binding proteins to PCSK9 depicted in Table 2 and FIGs.
  • the pharmaceutical formulation comprises any of the antigen binding proteins to PCSK9 depicted in Table 2 and FIGs. 2 and/or 3 and FIGs 31A and 3 IB.
  • the pharmaceutical formulation may comprise other antigen binding proteins to PCSK9; namely an antibody comprised of a light chain variable domain, SEQ ID NO:588 and a heavy chain variable domain, SEQ ID NO:589.
  • the pharmaceutical formulation comprises any one of 2 IB 12, 26H5, 31H4, 8A3, 11F1 or 8A1.
  • the pharmaceutical formulation comprises more than one different antigen binding protein to PCSK9.
  • pharmaceutical formulations comprise more than one antigen binding protein to PCSK9 wherein the antigen binding proteins to PCSK9 bind more than one epitope.
  • the various antigen binding proteins will not compete with one another for binding to PCSK9.
  • any of the antigen binding proteins depicted in Table 2 and FIGs. 2 and/or 3 can be combined together in a pharmaceutical formulation.
  • an antigen binding protein to PCSK9 and/or a therapeutic molecule is linked to a half-life extending vehicle known in the art.
  • vehicles include, but are not limited to, polyethylene glycol, glycogen (e.g., glycosylation of the ABP), and dextran.
  • glycogen e.g., glycosylation of the ABP
  • dextran e.g., dextran
  • acceptable formulation materials preferably are nontoxic to recipients at the dosages and concentrations employed.
  • the formulation material(s) are for s.c. and/or I.V. administration.
  • the pharmaceutical formulation comprises formulation materials for modifying, maintaining or preserving, for example, the pH, osmolality, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • suitable formulation materials include, but are not limited to, amino acids (such as proline, arginine, lysine, methionine, taurine, glycine, glutamine, or asparagine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, sodium phosphate ("NaOAC"), Tris-HCl, Tris buffer, citrates, phosphate buffer, phosphate- buffered saline (i.e., PBS buffer) or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetra acetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides; and
  • the optimal pharmaceutical formulation will be determined by one skilled in the art depending upon, for example, the intended route of administration, delivery format and desired dosage. See, for example, Remington's Pharmaceutical Sciences, supra. In certain embodiments, such formulations may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the antibodies of the invention.
  • the pharmaceutical formulation comprises high concentrations of antigen binding protein to PCSK9.
  • ABP concentration ranges from about 70 mg/ml to about 250 mg/ml, e.g., about 70 mg/ml, about 80 mg/ml, about 90 mg/ml, about 100 mg/ml, about 100 mg/ml, about 120 mg/ml, about 130 mg/ml, about 140 mg/ml, about 150 mg/ml, about 160 mg/ml, about 170 mg/ml, about 180 mg/ml, about 190 mg/ml, about 200 mg/ml, about 210 mg/ml, about 220 mg/ml, about 230 mg/ml, about 240 mg/ml, or about 250 mg/ml, and including all values in between.
  • the concentration of 2 IB 12, 26H5, or 31H4 ranges from about 100 mg/ml to about 150 mg/ml, e.g., 100 mg/ml, about 100 mg/ml, about 120 mg/ml, about 130 mg/ml, about 140 mg/ml, or about 150 mg/ml.
  • the concentration of 8 A3, 11F1 or 8A1 ranges from about 140 mg/ml to about 220 mg/ml, e.g., 140 mg/ml, about 150 mg/ml, about 160 mg/ml, about 170 mg/ml, about 180 mg/ml, about 190 mg/ml, about 200 mg/ml, about 210 mg/ml, about 220 mg/ml, or about 250 mg/ml.
  • the pharmaceutical formulation comprises at least one buffering agent such as, for example, sodium acetate, sodium chloride, phosphates, phosphate buffered saline ("PBS"), and/or Tris buffer of about pH 7.0-8.5.
  • the buffer serves to maintain a physiologically suitable pH.
  • the buffer can serve to enhance isotonicity and chemical stability of the pharmaceutical formulation.
  • the buffering agent ranges from about 0.05 mM to about 40 mM, e.g., about 0.05 mM, about 0.1 mM, about 0.5 mM, about 1.0 mM, about 5.0 mM, about 10 mM, about 15 mM, about 20 mM, about 30 mM, about 40 mM, about 50 mM, about 60 mM, about 70 mM, about 80 mM, about 90 mM, or about ⁇ buffering agent, inclusiveof all values in between.
  • the bufferning agent is NaOAC.
  • Exemplary pHs of the pharmaceutical formulation include from about 4 to about 6, or from about 4.8 to about 5.8, or from about 5.0 to about 5.2, or about 5, or about 5.2.
  • the pharmaceutical formulation is isotonic with an osmolality ranging from between about 250 to about 350 miliosmol/kg, e.g., about 250 mOsm/kg, about 260 mOsm/kg, about 270 mOsm/kg, about 280 mOsm/kg, about 290 mOsm/kg, about 300 mOsm/kg, about 310 mOsm/kg, about 320 mOsm/kg, about 330 mOsm/kg, about 340 mOsm/kg, or about 350 mOsm/kg, and including all values in between.
  • osmolality is the measure of the ratio of solutes to volume fluid. In other words, it is the number of molecules and ions (or molecules) per kilogram of a solution. Osmolality may be measured on an analytical instrument called an osmometer, such as Advanced Instruments 2020 Multi-sample Osmometer, Norwood, MA. The Advanced Instrumetns 2020 Multi-sample Osmometer measures osmolality by using the Freezing Point Depression method. The higher the osmolytes in a solution, the temperature in which it will freeze drops. Osmolality may also be measured using any other methods and in any other units known in the art such as linear extrapolation.
  • the pharmaceutical formulation comprises at least one surfactant including but not limited to Polysorbate-80, Polysorbate-60, Polysorbate-40, and Polysorbate-20.
  • the pharmaceutical formulation comprises a surfactant at a concentration that ranges from about 0.004% to about 10% weight per volume ("w/v") of the formulation, e.g., about 0.004%>, about 0.005%), about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.05%, about 0.1%), about 0.5%), about 1%, about 5%, or about 10%> surfactant w/v of the formulation.
  • the pharmaceutical formulation comprises polysorbate 80 at a concentration that ranges from about 0.004% to about 0.1% w/v of the formulation. In certain embodiments, the pharmaceutical formulation comprises polysorbate 20 at a concentration that ranges from about 0.004% to about 0.1% w/v of the formulation.
  • the pharmaceutical formulation comprises at least one stabilizing agent, such as a polyhydroxy hydrocarbon (including but not limited to sorbitol, mannitol, glycerol and dulcitol) and/or a disaccharide (including but not limited to sucrose, lactose, maltose and threhalose) and/or an amino acid (including but not limited to proline, arginine, lysine, methionine, and taurine) and or benzyl alcohol; the total of said polyhydroxy hydrocarbon and/or disaccharide and/or amino acid and/or benzyl alchol being about 0.5% to about 10% w/v of the formulation.
  • a stabilizing agent such as a polyhydroxy hydrocarbon (including but not limited to sorbitol, mannitol, glycerol and dulcitol) and/or a disaccharide (including but not limited to sucrose, lactose, maltose and threhalose) and/or an amino acid (including
  • the pharmaceutical formulation comprises a stabilizing agent at a concentration of about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%), about 8%), about 9% or about 10% sucrose.
  • the pharmaceutical formulation comprises a stabilizing agent at a concentration of about 5% sucrose.
  • the pharmaceutical formulation comprises a a stabilizing agent at a concentration of about 1%, about 2%, about 3%, about 4%, about 5%), about 6%), about 7%, about 8%, about 9% or about 10% sorbital.
  • the pharmaceutical formulation comprises a stabilizing agent at a concentration of about 9% sorbital.
  • the pharmaceutical formulation comprises a a stabilizing agent at a concentration of about 1%, about 2%, about 3%, about 4%, about 5% proline, arginine, lysine, methionine, and/or taurine. In certain embodiments, the pharmaceutical formulation comprises a stabilizing agent at a concentration of between about 2-3% proline. In certain embodiments, the pharmaceutical formulation comprises a a stabilizing agent at a concentration of about 1%, about 2%, about 3%, about 4%, about 5% benzyl alcohol. In certain embodiments, the pharmaceutical formulation comprises a stabilizing agent at a concentration of between about 1-2% benzyl alcohol.
  • the pharmaceutical formulation has a viscosity level of less than about 30 centipoise (cP) as measured at room temperature (i.e., 25C).
  • Viscosity may be measured by using a viscometer, e.g., Brookfield Engineering Dial Reading Viscometer, model LVT. Viscosity may also be measured using any other methods and in any other units known in the art (e.g., absolute, kinematic or dynamic viscosity or absolute viscosity).
  • the pharmaceutical formulation has a viscosity level of less than about 25 cP, about 20 cP, about 18 cP, about 15 cP, about 12 cP, about 10 cP; about 8 cP, about 6 cP, about 4 cP; about 2 cP; or about 1 cP.
  • the pharmaceutical formulation is stable as measured by at least one stability assay known to one of skill in the art, such as assays that examne the biophysical or biochemical characteristics of biologically active proteins over time.
  • a stable pharmaceutical formulation of the present invention is a pharmaceutical formulation of biologically active proteins that exhibits increased aggregation and/or reduced loss of biological activity of not more than 5% when stored at 2-8°C for at least 1 month, or 2 months, or 3 months, or 6 months, or 1 year or 2 years compared with a control formula sample.
  • the pharmaceutical formulation stability is measured using size exclusion HPLC ("SEC- HPLC"). SEC-HPLC separates proteins based on differences in their hydrodynamic volumes.
  • a stable pharmaceutical formulation should exhibit no more than about a 5% increase in high molecular weight species as compared to a control sample.
  • the pharmaceutical formulation should exhibit no more than about a 4%, no more than about a 3%, no more than about a 2%, no more than about a 1%, no more than about a 0.5% increase in high molecular weight speciies as compared to a control sample.
  • the pharmaceutical formulation stability is measured using cation-exchange HPLC (CEX-HPLC).
  • CEX-HPLC separates proteins based on differences in their surface charge.
  • charged isoforms of an anti-PCSK9 ABP are separated on a cation-exchange column and eluted using a salt gradient.
  • the eluent is monitored by UV absorbance.
  • the charged isoform distribution is evaluated by determining the peak area of each isoform as a percent of the total peak area.
  • a stable pharmaceutical formulation should exhibit no more than about a 5% decrease in the main isoform peak as compared to a control sample.
  • a stable pharmaceutical formulation should exhibit no more than about a 3% to about a 5% decrease in the main isoform peak as compared to a control sample. In certain embodiments, the pharmaceutical formulation should exhibit no more than about a 4% decrease, no more than about a 3% decrease, no more than about a 2% decrease, no more than about a 1% decrease, no more than about a 0.5% decrease in the main isoform peak as compared to a control sample.
  • the pharmaceutical formulation stability is measured using Subvisible Particle Detection by Light Obscuration ("HIAC").
  • HIAC Subvisible Particle Detection by Light Obscuration
  • An electronic, liquid-borne particle-counting system HIAC/Royco 9703 or equivalent
  • a light-obscuration sensor HIAC/Royco HRLD-150 or equivalent
  • a liquid sampler quantifies the number of particles and their size range in a given test sample.
  • particles in a liquid pass between the light source and the detector they diminish or "obscure” the beam of light that falls on the detector.
  • concentration of particles lies within the normal range of the sensor, these particles are detected one-by- one.
  • a stable pharmaceutical formulation should exhibit no more than 6000 ⁇ particles per container (or unit), as compared to a control sample. In certain embodiments, a stable pharmaceutical formulation should exhibit no more than 5000, no more than 4000, no more than 3000, no more than 2000, no more than 1000, ⁇ particles per container (or unit) as compared to a control sample.
  • a stable pharmaceutical formulation should exhibit no more than 600 25 ⁇ particles per container (or unit) as compared to a control sample. In certain embodiments, a stable pharmaceutical formulation should exhibit no more than 500, no more than 400, no more than 300, no more than 200, no more than 100, no more than 50 25 ⁇ particles per container (or unit) as compared to a control sample.
  • the pharmaceutical formulation stability is measured using visual assessment.
  • Visual assessment is a qualitative method used to describe the visible physical characteristics of a sample. The sample is viewed against a black and/or white background of an inspection booth, depending on the characteristic being evaluated (e.g., color, clarity, presence of particles or foreign matter). Samples are also viewed against an opalescent reference standard and color reference standards. In the case of visual assessment, a stable pharmaceutical formulation should exhibit no significant change in color, clarity, presence of particles or foreign matter as compared to a control sample.
  • One aspect of the present invention is a pharmaceutical formulation which comprises: (i) about 70 mg/ml to about 250 mg/ml of antigen binding protein to PCSK9; (ii) about 0.05 mM to about 40 mM of a buffer such as sodium acetate (“NaOAC”) serves as a buffering agent; (iii) about 1 % to about 5% proline, arginine, lysine, methionine, or taurine (also know as 2-aminoethanesulfonic acid) and/or 0.5% to about 5% benzyl alcohol which serves as a stabilizing agent; and (iv) about 0.004%> to about 10% w/v of the formulation of a non-ionic surfactant (including but not limited to Polysorbate-80, Polysorbate-60, Polysorbate-40, and Polysorbate-20); wherein said formulation has a pH in the range of about 4.0 to 6.0.
  • a buffer such as sodium acetate
  • pharmaceutical formulations of this invention comprise (i) at least about 70 mg/ml, about 100 mg/ml, about 120 mg/ml, about 140 mg/ml, about 150 mg/ml, about 160 mg/ml, about 170 mg/ml, about 180 mg/ml, about 190 mg/ml, about 200 mg/ml of an anti-PCSK9 antibody; (ii) about 10 mM NAOAC; (iii) about 0.01% polysorbate 80; and (iv) between about 2%-3% proline (or about 250 mM to about 270 mM proline), wherein the formulation has a pH of about 5.
  • pharmaceutical formulations of this invention comprise (i) at least about 70 mg/ml, about 100 mg/ml, about 120 mg/ml, about 140 mg/ml of the anti-PCSK9 antibody, 21B12, 26H5 and/or 31H4; (ii) about 10 mM NAOAC; (iii) about 0.01% polysorbate 80; and (iv) between about 2%-3% proline (or about 250 mM to about 270 mM proline), wherein the formulation has a pH of about 5.
  • pharmaceutical formulations of this invention comprise (i) at least about 150 mg/ml, about 160 mg/ml, about 170 mg/ml, about 180 mg/ml, about 190 mg/ml, about 200 mg/ml of the anti-PCSK9 antibody, 8A3, 11F1 and/or 8A1; (ii) about 10 mM NAOAC; (iii) about 0.01% polysorbate 80; and (iv) between about 2%>-3%> proline (or about 250 mM to about 270 mM proline), wherein the formulation has a pH of about 5.
  • One aspect of the present invention is a pharmaceutical formulation which comprises (i) at least about 70 mg/ml to about 250 mg/ml of an anti-PCSK9 antibody; (ii) about 5 mM to about 20 mM of a buffer, such as NAOAC; (iii) about 1% to about 10% w/v of the formulation comprises a polyhydroxy hydrocarbon such as sorbitol, or a disaccharide such as sucrose; and (iv) about 0.004%> to about 10%> w/v of the formulation of a surfactant, such as polysorbate 20 or polysorbate 80; wherein said formulation has a pH in the range of about 4.8 to 5.8; and wherein the pharmaceutical formulation optionally comprises about 80 mM to about 300 mM proline, arginine, lysine, methionine, or taurine and/or 0.5% to about 5% benzyl alcohol which serves to reduce viscosity.
  • a buffer such as NAOAC
  • pharmaceutical formulations of this invention comprise (i) at least about 70 mg/ml to about 250 mg/ml of the anti-PC SK9 antibody; (ii) about 10 mM NAOAC; (iii) about 9% sucrose; and (iv) about 0.004% polysorbate 20, wherein the formulation has a pH of about 5.2.
  • pharmaceutical formulations of this invention comprise (i) at least about 70 mg/ml, about 100 mg/ml, about 120 mg/ml, about 140 mg/ml, about 160 mg/ml, about 180 mg/ml, about 200 mg/ml of an anti-PCSK9 antibody; (ii) about 15 mM NAOAC; (iii) about 9% sucrose; and (iv) about 0.01% polysorbate 20, wherein the formulation has a pH of about 5.2.
  • pharmaceutical formulations of this invention comprise (i) at least about 70 mg/ml, about 100 mg/ml, about 120 mg/ml, about 140 mg/ml, about 160 mg/ml, about 180 mg/ml, about 200 mg/ml of an anti-PCSK9 antibody; (ii) about 20 mM NAOAC; (iii) about 9% sucrose; and (iv) about 0.01% polysorbate 20, wherein the formulation has a pH of about 5.2.
  • pharmaceutical formulations of this invention comprise (i) at least about 70 mg/ml, about 100 mg/ml, about 120 mg/ml, about 140 mg/ml, about 160 mg/ml, about 180 mg/ml, about 200 mg/ml of an anti-PCSK9 antibody; (ii) about 10 mM NAOAC; (iii) about 9% sucrose; (iv) about 0.01% polysorbate 80; and (v) about 250 mM proline, wherein the formulation has a pH of about 5.
  • compositions of the invention can be administered in combination therapy, i.e., combined with other agents.
  • the combination therapy comprises an antigen binding protein capable of binding PCSK9, in combination with at least one anti-cholesterol agent.
  • Agents include, but are not limited to, in vitro synthetically prepared chemical formulations, antibodies, antigen binding regions, and combinations and conjugates thereof.
  • an agent can act as an agonist, antagonist, allosteric modulator, or toxin.
  • an agent can act to inhibit or stimulate its target (e.g., receptor or enzyme activation or inhibition), and thereby promote increased expression of LDLR or decrease serum cholesterol levels.
  • an antigen binding protein to PCSK9 can be administered prior to, concurrent with, and subsequent to treatment with a cholesterol- lowering (serum and/or total cholesterol) agent.
  • an antigen binding protein to PCSK9 can be administered prophylacticly to prevent or mitigate the onset of hypercholesterolemia, heart disease, diabetes, and/or any of the cholesterol related disorder.
  • an antigen binding protein to PCSK9 can be administered for the treatment of an existing hypercholesterolemia condition.
  • the ABP delays the onset of the disorder and/or symptoms associated with the disorder.
  • the ABP is provided to a subject lacking any symptoms of any one of the cholesterol related disorders or a subset thereof.
  • an antigen binding protein to PCSK9 is used with particular therapeutic agents to treat homozygous familial hypercholesterolemia.
  • two, three, or more agents can be administered.
  • such agents can be provided together by inclusion in the same formulation.
  • such agent(s) and an antigen binding protein to PCSK9 can be provided together by inclusion in the same formulation.
  • such agents can be formulated separately and provided together by inclusion in a treatment kit.
  • such agents and an antigen binding protein to PCSK9 can be formulated separately and provided together by inclusion in a treatment kit.
  • such agents can be provided separately.
  • a formulation comprising an antigen binding protein to PCSK9, with or without at least one additional therapeutic agents can be prepared for storage by mixing the selected formulation having the desired degree of purity with optional formulation agents (Remington 's Pharmaceutical Sciences, supra) in the form of a lyophilized cake or an aqueous solution.
  • a formulation comprising an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent can be formulated as a lyophilizate using appropriate excipients.
  • a therapeutic formulation when parenteral administration is contemplated, can be in the form of a pyrogen-free, parenterally acceptable aqueous solution comprising a desired antigen binding protein to PCSK9, with or without additional therapeutic agents, in a pharmaceutically acceptable vehicle.
  • a vehicle for parenteral injection is sterile distilled water in which an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent, is formulated as a sterile, isotonic solution, properly preserved.
  • the preparation can involve the formulation of the desired molecule with an agent, such as injectable microspheres, bio-erodible particles, polymeric compounds (such as polylactic acid or polyglycolic acid), beads or liposomes, that can provide for the controlled or sustained release of the product which can then be delivered via a depot injection.
  • an agent such as injectable microspheres, bio-erodible particles, polymeric compounds (such as polylactic acid or polyglycolic acid), beads or liposomes, that can provide for the controlled or sustained release of the product which can then be delivered via a depot injection.
  • hyaluronic acid can also be used, and can have the effect of promoting sustained duration in the circulation.
  • implantable drug delivery devices can be used to introduce the desired molecule.
  • a pharmaceutical formulation can be formulated for inhalation.
  • an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent can be formulated as a dry powder for inhalation.
  • an inhalation solution comprising an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent can be formulated with a propellant for aerosol delivery.
  • solutions can be nebulized. Pulmonary administration is further described in PCT application no. PCT/US94/001875, which describes pulmonary delivery of chemically modified proteins.
  • a pharmaceutical formulation can involve an effective quantity of an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent, in a mixture with non-toxic excipients which are suitable for the manufacture of tablets.
  • suitable excipients include, but are not limited to, inert diluents, such as calcium carbonate, sodium carbonate or bicarbonate, lactose, or calcium phosphate; or binding agents, such as starch, gelatin, or acacia; or lubricating agents such as magnesium stearate, stearic acid, or talc.
  • sustained-release preparations can include semi permeable polymer matrices in the form of shaped articles, e.g. films, or microcapsules.
  • Sustained release matrices can include polyesters, hydrogels, polylactides (U.S. 3,773,919 and EP 058,481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al, Biopolymers, 22:547-556 (1983)), poly (2- hydroxyethyl-methacrylate) (Langer et al, J. Biomed. Mater. Res., 15:167-277 (1981) and Langer, Chem. Tech., 12:98-105 (1982)), ethylene vinyl acetate (Langer et al, supra) or poly-D(-)-3-hydroxybutyric acid (EP 133,988).
  • sustained release formulations can also include liposomes, which can be prepared by any of several methods known in the art. See, e.g., Eppstein et al, Proc. Natl. Acad. Sci. USA, 82:3688-3692 (1985); EP 036,676; EP 088,046 and EP 143,949.
  • the pharmaceutical formulation to be used for in vivo administration typically is sterile. In certain embodiments, this can be accomplished by filtration through sterile filtration membranes. In certain embodiments, where the formulation is lyophilized, sterilization using this method can be conducted either prior to or following lyophilization and reconstitution. In certain embodiments, the formulation for parenteral administration can be stored in lyophilized form or in a solution. In certain embodiments, parenteral formulations generally are 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.
  • the pharmaceutical formulation once the pharmaceutical formulation has been formulated, it can be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. In certain embodiments, such formulations can be stored either in a ready-to-use form or in a form ⁇ e.g. , lyophilized) that is reconstituted prior to administration.
  • the pharmaceutical formulation once the pharmaceutical formulation has been formulated, it can be stored in pre-filled syringes as a solution or suspension in a ready- to-use form
  • kits are provided for producing a single-dose administration unit.
  • the kit can contain both a first container having a dried protein and a second container having an aqueous formulation.
  • kits containing single and multi-chambered pre-filled syringes e.g. , liquid syringes and lyosyringes are included.
  • the effective amount of a pharmaceutical formulation comprising an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent, to be employed therapeutically will depend, for example, upon the therapeutic context and objectives.
  • the appropriate dosage levels for treatment will thus vary depending, in part, upon the molecule delivered, the indication for which an antigen binding protein to PCSK9, with or without at least one additional therapeutic agent, is being used, the route of administration, and the size (body weight, body surface or organ size) and/or condition (the age and general health) of the patient.
  • the clinician can titer the dosage and modify the route of administration to obtain the optimal therapeutic effect.
  • the formulation can be administered locally via implantation of a membrane, sponge or another appropriate material onto which the desired molecule has been absorbed or encapsulated.
  • the device can be implanted into any suitable tissue or organ, and delivery of the desired molecule can be via diffusion, timed-release bolus, or continuous administration.
  • antigen binding proteins to PCSK9 comprising: one or more heavy chain complementary determining regions (CDRHs) and one or more light chain complementary determining regions (CDRLs) depicted in Table 2 and FIGs. 2 and/or 3 and FIGS. 31A and 3 IB can be administered to a patient diagnosed with homozygous familial hypercholesterolemia according to the methods of the present invention.
  • antigen binding protein to PCSK9 comprising: a light chain variable region that comprises an amino acid sequence that is at least 90% identical the antigen binding proteins to PCSK9 depicted in Table 2 and FIGs.
  • any of the antigen binding proteins to PCSK9 depicted in Table 2 and FIGs. 2 and/or 3 and/or 13 and/or FIG.31A and 3 IB can be administered to a patient diagnosed with homozygous familial hypercholesterolemia according to the methods of the present invention.
  • any of the antigen binding proteins to PCSK9 depicted in Table 2 and FIGs. 2 and/or 3 and/or 13 and/or FIG.31A and 3 IB can be administered to a patient diagnosed with homozygous familial hypercholesterolemia according to the methods of the present invention.
  • other antigen binding proteins to PCSK9 namely an antibody comprised of a light chain variable domain, SEQ ID NO:588 and a heavy chain variable domain, SEQ ID NO:589 can be administered to a patient diagnosed with homozygous familial hypercholesterolemia.
  • an antibody comprised of a light chain variable domain, SEQ ID NO:588 and a heavy chain variable domain, SEQ ID NO:589 can be administered to a patient diagnosed with homozygous familial hypercholesterolemia.
  • any one of 21B12, 26H5, 31H4, 8A3, 11F1 or 8A1 can be administered to a patient diagnosed with homozygous familial hypercholesterolemia.
  • the amount of an antigen binding protein to PCSK9 is, generally, a therapeutically effective amount.
  • the amount of ABP may be expressed in terms of milligrams of antibody (i.e., mg) or milligrams of antibody per kilogram of patient body weight (i.e., mg/kg).
  • a typical dosage of a PCSK9 antigen binding protein can range from about 0.1 g/kg to up to about 100 mg/kg or more of antigen binding protein to PCSK9,.
  • the dosage can range from 0.1 g/kg up to about 100 mg/kg; or 1 g/kg up to about 100 mg/kg; or 5 g/kg up to about 100 mg/kg of antigen binding protein to PCSK9; or 1 mg/kg to about 50 mg/kg of antigen binding protein to PCSK9; or 2 mg/kg to about 20 mg/kg of antigen binding protein to PCSK9; or 2 mg/kg to about 10 mg/kg of antigen binding protein to PCSK9 .
  • PCSK9 can range from at least about 120 mg to about 3000 mg, of about 140 mg to about 2800 mg, of about 140 mg to about 2500 mg, of about 140 mg to about 2000 mg, of about 140 mg to about 1800 mg, of about 140 mg to about 1400 mg, of about 120 mg to about 1200 mg, of about 120 mg to about 1000 mg, of about 120 mg to about 700 mg, of about 140 mg to about 700 mg, of about 140 mg to about 600 mg, of about 140 mg to about 450 mg, of about 120 mg to about 450 mg, of about 120 mg to about 450 mg, of about 140 mg to about 450 mg, of about 210 mg to about 450 mg, or of about 280 mg to about 450 mg, of about 210 mg to about 420 mg, of about 280 mg to about 420 mg, of about 420 mg to about 3000 mg, of about 700 mg to about 3000 mg, of about 1000 mg to about 3000 mg, of about 1200 to about 3000 mg, of about 1400 mg to about 3000 mg, of about 1800 mg to about
  • the anti-PCSK9 antibody is administered to a patient at a dose of about 35 mg, of about 45 mg, of about 70 mg, of about 105 mg, of about 120 mg of about 140 mg, of about 150 mg, of about 160 mg, of about 170 mg, of about 180 mg, of about 190 mg, of about 200 mg, of about 210 mg, of about 280 mg, of about 360 mg, of about 420 mg, of about 450 mg, of about 600 mg, of about 700 mg, of about 1200 mg, of about 1400 mg, of about 1800 mg, of about 2000 mg, of about 2500 mg, of about 2800 mg, or about 3000 mg.
  • the frequency of dosing will take into account the pharmacokinetic parameters of an antigen binding protein to PCSK9 and/or any additional therapeutic agents in the formulation used.
  • a clinician will administer the formulation until a dosage is reached that achieves the desired effect.
  • the formulation can therefore be administered as a single dose, or as two, three, four or more doses (which may or may not contain the same amount of the desired molecule) over time, or as a continuous infusion via an implantation device or catheter.
  • the formulation can also be delivered subcutaneously or intravenously with a standard needle and syringe.
  • pen delivery devices as well as autoinjector delivery devices, have applications in delivering a pharmaceutical formulation of the present invention. Further refinement of the appropriate dosage is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them. In certain embodiments, appropriate dosages can be ascertained through use of appropriate dose- response data. In some embodiments, the amount and frequency of administration can take into account the desired cholesterol level (serum and/or total) to be obtained and the subject's present cholesterol level, LDL level, and/or LDLR levels, all of which can be obtained by methods that are well known to those of skill in the art.
  • a dose of at least about 120 mg; or up to about 140 mg; or up to about 210 mg; or up to about 280 mg; or up to about 350 mg, or up to about 420 mg, or up to about 450 mg of an antigen binding protein to PCSK9 is administered once a week (QW) to a patient in need thereof.
  • a dose of at least about 120 mg, or up to about 140 mg; or up to about 150 mg, or up to about 210 mg, or up to about 280 mg; or up to about 350 mg, or up to about 420 mg; or up to about 450 mg of an antigen binding protein to PCSK9 is administered once every other week, (or every two weeks)(Q2W), to a patient in need thereof.
  • 280 mg or up to about 300 mg; or up to about 350 mg; or up to about 400 mg; or up to about 420 mg; or up to about 450 mg; or up to about 600 mg; or up to about 700 mg; or up to about 1000 mg; or up to about 2000 mg; or up to about 3000 mg every four weeks of a an antigen binding protein to PCSK9 is administered once every four weeks, (or once a month)(Q4W), to a patient in need thereof.
  • a dose of at least about 400 mg; or up to about 420 mg; or up to about 450 mg; or up to about 600 mg; or up to about 700 mg; or up to about 1000 mg; or up to about 2000 mg; or up to about 3000 mg every other month of a an antigen binding protein to PCSK9 is administered once every 8 weeks, (or once every other month), to a patient in need thereof.
  • the serum LDL cholesterol level is reduced by at least about 10%, as compared to a predose serum LDL cholesterol level. In some embodiments, the serum LDL cholesterol level is reduced by at least about 15%.In some embodiments, the serum LDL cholesterol level is reduced by at least about 20%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 25%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 30%). In some embodiments, the serum LDL cholesterol level is reduced by at least about 40%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 50%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 55%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 60%>.
  • the serum LDL cholesterol level is reduced by at least about 65%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 70%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 75%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 80%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 85%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 90%.
  • the serum LDL cholesterol level is reduced by at least about 10%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 15 ), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 20%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 25%o, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 30%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 35%, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 40%, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 45%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 50%o, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 55%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 60%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 65%, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 70%, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 75%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 80%o, as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 85%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 90%), as compared to a predose serum LDL cholesterol level, and the reduction is sustained for a period of at least about 3 days, at least about 5 days, at least about 7 days, at least about 10 days, at least about 14 days, at least about 21 days, at least about 25 days, at least about 28 days, or at least about 31 days relative to a predose level.
  • the methods provided herein are for treatment of patients diagnosed with homozygous familial hypercholesterolemia using antigen binding proteins, including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9).
  • antigen binding proteins including antibodies, against proprotein convertase subtilisin/kexin type 9 (PCSK9).
  • an antigen binding protein to PCSK9 is used to modulate serum LDL cholesterol levels in a patient diagnosed with homozygous familial hypercholesterolema.
  • the antigen binding protein to PCSK9 is used to decrease the amount of serum LDL cholesterol from an abnormally high level or even a normal level.
  • the serum LDL cholesterol level is reduced by at least about 10% as compared to a predose level.
  • the serum LDL cholesterol level is reduced by at least about 15%.
  • the serum LDL cholesterol level is reduced by at least about 20%.
  • the serum LDL cholesterol level is reduced by at least about 25%.
  • the serum LDL cholesterol level is reduced by at least about 30%.
  • the serum LDL cholesterol level is reduced by at least about 35%. In certain embodiments, the serum LDL cholesterol level is reduced by at least about 40%. In certain embodiments, the serum LDL cholesterol level is reduced by at least about 45%. In certain embodiments, the serum LDL cholesterol level is reduced by at least about 50%. In certain embodiments, the serum LDL cholesterol level is reduced by at least about 55%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 60%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 65%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 70%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 75%. In some embodiments, the serum LDL cholesterol level is reduced by at least about 80%>. In some embodiments, the serum LDL cholesterol level is reduced by at least about 85%). In some embodiments, the serum LDL cholesterol level is reduced by at least about 90%.
  • an antigen binding protein to PCSK9 is used to modulate serum
  • the antigen binding protein to PCSK9 is neutralizing. In some embodiments, the antigen binding protein to PCSK9 is used to decrease PCSK9 values from an abnormally high level or even a normal level. In some embodiments, the serum PCSK9 value is reduced by at least about 20% as compared to a predose level. In some embodiments, the serum PCSK9 value is reduced by at least about 25%. In some embodiments, the serum PCSK9 value is reduced by at least about 30%. In some embodiments, the serum PCSK9 value is reduced by at least about 35%. In some embodiments, the serum PCSK9 value is reduced by at least about 40%.
  • the serum PCSK9 value is reduced by at least about 45%. In some embodiments, the serum PCSK9 value is reduced by at least about 50%. In some embodiments, the serum PCSK9 value is reduced by at least about 55%. In some embodiments, the serum PCSK9 value is reduced by at least about 60%. In some embodiments, the serum PCSK9 value is reduced by at least about 65%. In some embodiments, the serum PCSK9 value is reduced by at least about 70%. In some embodiments, the serum PCSK9 value is reduced by at least about 75%. In some embodiments, the serum PCSK9 value is reduced by at least about 80%. In some embodiments, the serum PCSK9 value is reduced by at least about 85%. In some embodiments, the serum PCSK9 value is reduced by at least about 90%>.
  • an antigen binding protein to PCSK9 is used to modulate total cholesterol level in a patient diagnosed with homozygous familial hypercholesterolemia.
  • the antigen binding protein to PCSK9 is neutralizing.
  • the antigen binding protein to PCSK9 is used to decrease the amount of total cholesterol from an abnormally high level or even a normal level.
  • the total cholesterol level is reduced by at least about 20% as compared to a predose level.
  • the total cholesterol level is reduced by at least about 25%.
  • the total cholesterol level is reduced by at least about 30%.
  • the total cholesterol level is reduced by at least about 35%.
  • the total cholesterol level is reduced by at least about 40%.
  • the total cholesterol level is reduced by at least about 45%. In some embodiments, the total cholesterol level is reduced by at least about 50%. In some embodiments, the total cholesterol level is reduced by at least about 55%. In some embodiments, the total cholesterol level is reduced by at least about 60%.
  • an antigen binding protein to PCSK9 is used to modulate the non- HDL cholesterol level in a patient diagnosed with homozygous familial hypercholesterolemia.
  • the antigen binding protein to PCSK9 is neutralizing.
  • the antigen binding protein to PCSK9 is used to decrease the non-HDL cholesterol from an abnormally high level or even a normal level.
  • the non-HDL cholesterol level is reduced by at least about 30%).
  • the non-HDL cholesterol level is reduced by at least about 35%.
  • the non-HDL cholesterol level is reduced by at least about 40%.
  • the non-HDL cholesterol level is reduced by at least about 50%.
  • the non-HDL cholesterol level is reduced by at least about 55%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 60%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 65%>. In some embodiments, the non-HDL cholesterol level is reduced by at least about 70%>. In some embodiments, the non- HDL cholesterol level is reduced by at least about 75%. In some embodiments, the non-HDL cholesterol level is reduced by at least about 80%>. In some embodiments, the non-HDL cholesterol level is reduced by at least about 85%.
  • an antigen binding protein to PCSK9 is used to modulate the
  • the antigen binding protein to PCSK9 is neutralizing. In some embodiments, the antigen binding protein to PCSK9 is used to decrease the amount of ApoB from an abnormally high level or even a normal level. In some embodiments, the ApoB level is reduced by at least about 10%> as compared to a predose level. In some embodiments, the ApoB level is reduced by at least about 15%. In some embodiments, the ApoB level is reduced by at least about 20%. In some embodiments, the ApoB level is reduced by at least about 25%. In some embodiments, the ApoB level is reduced by at least about 30%.
  • the ApoB level is reduced by at least about 35%. In some embodiments, the ApoB level is reduced by at least about 40%. In some embodiments, the ApoB level is reduced by at least about 45%. In some embodiments, the ApoB level is reduced by at least about 50%. In some embodiments, the ApoB level is reduced by at least about 55%. In some embodiments, the ApoB level is reduced by at least about 60%. In some embodiments, the ApoB level is reduced by at least about 65%. In some embodiments, the ApoB level is reduced by at least about 70%. In some embodiments, the ApoB level is reduced by at least about 75%.
  • an antigen binding protein to PCSK9 is used to modulate the Lp(a) levels in a patient diagnosed with homozygous familial hypercholesterolemia.
  • the antigen binding protein to PCSK9 is neutralizing.
  • the antigen binding protein to PCSK9 is used to decrease the amount of Lp(a) from an abnormally high level or even a normal level.
  • the Lp(a) level is reduced by at least about 10% as compared to a predose level.
  • the Lp(a) level is reduced by at least about 15%.
  • the Lp(a) level is reduced by at least about 20%.
  • the Lp(a) level is reduced by at least about 25%.
  • the Lp(a) level is reduced by at least about 30%. In some embodiments, the Lp(a) level is reduced by at least about 35%. In some embodiments, the Lp(a) level is reduced by at least about 40%). In some embodiments, the Lp(a) level is reduced by at least about 45%. In some embodiments, the Lp(a) level is reduced by at least about 50%. In some embodiments, the Lp(a) level is reduced by at least about 55%. In some embodiments, the Lp(a) level is reduced by at least about 60%. In some embodiments, the Lp(a) level is reduced by at least about 65%.
  • methods are provided of treating homozygous familial hypercholesterolemia, comprising administering a therapeutically effective amount of one or more antigen binding proteins to PCSK9 and another therapeutic agent.
  • an antigen binding protein to PCSK9 is administered prior to the administration of at least one other therapeutic agent.
  • an antigen binding protein to PCSK9 is administered concurrent with the administration of at least one other therapeutic agent.
  • an antigen binding protein to PCSK9 is administered subsequent to the administration of at least one other therapeutic agent.
  • Therapeutic agents include, but are not limited to, at least one other cholesterol-lowering (serum and/or total body cholesterol) agent.
  • the agent increases the expression of LDLR, have been observed to increase serum HDL levels, lower LDL levels or lower triglyceride levels.
  • agents include, but are not limited to, statins (atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin), Nicotinic acid (Niacin) (NIACOR, NIASPAN (slow release niacin), SLO-NIACIN (slow release niacin), CORDAPTIVE (laropiprant)), Fibric acid (LOPID (Gemfibrozil), TRICOR (fenofibrate), Bile acid sequestrants (QUESTRAN (cholestyramine), colesevelam (WELCHOL), COLESTID (colestipol)), Cholesterol absorption inhibitors (ZETIA (ezetimibe)), Combining nicotinic acid with statin (ADVICOR (LOVASTATIN and NIASPAN), Combining a statin with an absorption inhibitor (VYTORIN
  • the ABP is combined with PPAR gamma agonsits, PPAR alpha/gamma agonists, squalene synthase inhibitors, CETP inhibitors, anti-hypertensives, antidiabetic agents (such as sulphonyl ureas, insulin, GLP-1 analogs, DDPIV inhibitors, e.g., metaformin), ApoB modulators, such as mipomersan, MTP inhibitoris and /or arteriosclerosis obliterans treatments.
  • PPAR gamma agonsits PPAR alpha/gamma agonists
  • squalene synthase inhibitors CETP inhibitors
  • anti-hypertensives such as sulphonyl ureas, insulin, GLP-1 analogs, DDPIV inhibitors, e.g., metaformin
  • ApoB modulators such as mipomersan, MTP inhibitoris and /or arteriosclerosis obliterans treatments.
  • the ABP is combined with an agent that increases the level of LDLR protein in a subject, such as statins, certain cytokines like oncostatin M, estrogen, and/or certain herbal ingredients such as berberine.
  • the ABP is combined with an agent that increases serum cholesterol levels in a subject (such as certain anti-psycotic agents, certain HIV protease inhibitors, dietary factors such as high fructose, sucrose, cholesterol or certain fatty acids and certain nuclear receptor agonists and antagonists for RXR, RAR, LXR, FXR).
  • the ABP is combined with an agent that increases the level of PCSK9 in a subject, such as statins and/or insulin. The combination of the two can allow for the undesirable side-effects of other agents to be mitigated by the ABP.
  • ABP names are used generically herein (e.g., "21B12” can be used as outlined in Example 7, as in Table 2, or as in Figures 2 and/or 3 and/or 13 A, 13C, 13F-13J), unless explicitly denoted otherwise.
  • Antibodies to the mature form of PCSK9 were raised in XenoMouse® mice (Abgenix, Fremont, CA), which are mice containing human immunoglobulin genes.
  • XenoMouse® mice Two groups of XenoMouse® mice, group 1 and 2, were used to produce antibodies to PCSK9.
  • Group 1 included mice of the XenoMouse® strain XMG2-KL, which produces fully human IgG2 K and lgG2X antibodies.
  • Group 1 mice were immunized with human PCSK9.
  • PCSK9 was prepared using standard recombinant techniques using the GenBank sequence as reference (NM_174936).
  • Group 2 involved mice of the XenoMouse® strain XMG4-KL, which produce fully human IgG4 K and lgG4 antibodies.
  • Group 2 mice were also immunized with human PCSK9.
  • mice of both groups were injected with antigen eleven times, according to the schedule in Table 3.
  • each mouse was injected with a total of 10 g of antigen delivered intraperitoneally into the abdomen.
  • Subsequent boosts are 5ug doses and injection method is staggered between intraperitoneal injections into the abdomen and sub-cutaneous injections at the base of the tail.
  • antigen is prepared as an emulsion with TiterMax® Gold (Sigma, Cat # T2684) and for subcutaneous injections antigen is mixed with Alum (aluminum phosphate) and CpG oligos.
  • each mouse was injected with a total of 5 g of antigen in the adjuvant alum gel.
  • a final injection of 5 ⁇ g of antigen per mouse is delivered in Phospho buffered saline and delivered into 2 sites 50% IP into the abdomen and 50% SQ at the base of tail.
  • the immunization programs are summarized in Table 1.1, shown below.
  • the protocol used to titer the XenoMouse animals was as follows: Costar 3368 medium binding plates were coated with neutravadin @ 8ug/ml (50ul/well) and incubated at 4°C in IXPBS/O.05% azide overnight. They were washed using TiterTek 3-cycle wash with RO water. Plates were blocked using 250ul of 1XPBS/I%milk and incubated for at least 30 minutes at RT. Block was washed off using TiterTek 3-cycle wash with RO water. One then captured b-human PCSK9 @ 2ug/ml in lXPBS/l%milk/10mM Ca2+ (assay diluent) 50ul/well and incubated for lhr at RT.
  • ELISA assays were as follows: For samples comprising b-PCSK9 with no V5His tag the following protocol was employed: Costar 3368 medium binding plates (Corning Life Sciences) were employed. The plates were coated with neutravadin at 8 ⁇ g/ml in lXPBS/0.05%Azide, (50 ⁇ /well). The plates were incubated at 4°C overnight. The plates were then washed using a Titertek M384 plate washer (Titertek, Huntsville, AL). A 3-cycle wash was performed. The plates were blocked with 250 ⁇ of 1XPBS/1% milk and incubated approximately 30 minutes at room temperature. The plates were then washed using the M384 plate washer. A 3- cycle wash was performed.
  • the capture was b-hu PCSK9, without a V5 tag, and was added at 2 ⁇ g/ml in lXPBS/l%milk/10mM Ca 2+ (40 ⁇ /well). The plates were then incubated for 1 hour at room temperature. A 3-cycle wash was performed. Sera were titrated 1 :3 in duplicate from 1 : 100, and row H was blank for sera. The titration was done in assay diluent, at a volume of 50 ⁇ /well. The plates were incubated for 1 hour at room temperature. Next, a 3-cycle wash was performed.
  • Goat anti Human IgG Fc HRP at 100 ng/ml (1 :4000) in lXPBS/l%milk 10mM Ca 2+ (50 ⁇ /well) was added to the plate and was incubated 1 hour at room temperature. The plates were washed once again, using a 3-cycle wash. The plates were then patted dry with paper towel. Finally, 1 step TMB (Neogen, Lexington, Kentucky) (50 ⁇ /well) was added to the plate and was quenched with IN hydrochloric acid (50 ⁇ /well) after 30 minutes at room temperature. OD's were read immediately at 450 nm using a Titertek plate reader.
  • Positive controls to detect plate bound PCSK9 were soluble LDL receptor (R&D Systems, Cat #2148LD/CF) and a polyclonal rabbit anti-PCSK9 antibody (Caymen Chemical #10007185) titrated 1 :3 in duplicate from 3 ⁇ / ⁇ 1 in assay diluent.
  • LDLR was detected with goat anti LDLR (R&D Systems, Cat #AF2148) and rabbit anti goat IgGFc HRP at a concentration of 400 ng/ml; the rabbit polyclonal was detected with goat anti-rabbit IgG Fc at a concentration of 400 ng/ml in assay diluent.
  • Negative control was naive XMG2-KL and XMG4-KL sera titrated 1 :3 in duplicate from 1 : 100 in assay diluent.
  • the capture was b-hu PCSK9, with a V5 tag, and was added at 2 ⁇ in lXPBS/l%milk/10mM Ca 2+ (40 ⁇ /well). The plates were then incubated for 1 hour at room temperature. A 3-cycle wash was performed. Sera were titrated 1 :3 in duplicate from 1 : 100, and row H was blank for sera. The titration was done in assay diluent, at a volume of 50 ⁇ /well. The plates were incubated for 1 hour at room temperature. Next, the plates were washed using the M384 plate washer operated using a 3-cycle wash.
  • Goat anti Human IgG Fc HRP at 400 ng/ml in lXPBS/l%milk/10mM Ca 2+ was added at 50 ⁇ /well to the plate and the plate was incubated 1 hour at room temperature. The plates were washed once again, using a 3-cycle wash. The plates were then patted dry with paper towel. Finally, 1 step TMB (Neogen, Lexington, Kentucky) (50 ⁇ /well) was added to the plate and the plate was quenched with IN hydrochloric acid (50 ⁇ /well) after 30 minutes at room temperature. OD's were read immediately at 450 nm using a Titertek plate reader.
  • LDLR positive control was LDLR, rabbit anti-PCSK9 titrated 1 :3 in duplicate from 3 ⁇ g/ml in assay diluent.
  • LDLR detect with goat anti-LDLR (R&D Systems, Cat #AF2148) and rabbit anti-goat IgG Fc HRP at a concentration of 400 ng/ml; rabbit poly detected with goat anti-rabbit IgG Fc at a concentration of 400 ng/ml in assay diluent.
  • Negative control was naive XMG2-KL and XMG4-KL sera titrated 1 :3 in duplicate from 1 : 100 in assay diluent.
  • mice immunized with soluble antigen were tested by ELISA assay for mice immunized with soluble antigen as described.
  • Table 4 summarizes the ELISA data and indicates that there were some mice which appeared to be specific for PCSK9. See, e.g., Table 4. Therefore, at the end of the immunization program, 10 mice (in bold in Table 1.2) were selected for harvest, and splenocytes and lymphocytes were isolated from the spleens and lymph nodes respectively, as described herein.
  • This example outlines how the immune cells were recovered and the hybridomas were generated.
  • Selected immunized mice were sacrificed by cervical dislocation and the draining lymph nodes were harvested and pooled from each cohort.
  • the B cells were dissociated from lymphoid tissue by grinding in DMEM to release the cells from the tissues, and the cells were suspended in DMEM. The cells were counted, and 0.9 ml DMEM per 100 million lymphocytes was added to the cell pellet to resuspend the cells gently but completely.
  • Lymphocytes were mixed with nonsecretory myeloma P3X63Ag8.653 cells purchased from ATCC, cat.# CRL 1580 (Kearney et al, (1979) J. Immunol. 123, 1548- 1550) at a ratio of 1 :4.
  • the cell mixture was gently pelleted by centrifugation at 400 x g 4 min. After decanting of the supernatant, the cells were gently mixed using a 1 ml pipette.
  • Preheated PEG/DMSO solution from Sigma (cat# P7306) (1 ml per million of B-cells) was slowly added with gentle agitation over 1 min followed by 1 min of mixing.
  • Preheated ID MEM (2 ml per million of B cells) (DMEM without glutamine, L-glutamine, pen/strep, MEM non-essential amino acids (all from Invitrogen), was then added over 2 minutes with gentle agitation. Finally preheated IDMEM (8 ml per 10 6 B-cells) was added over 3 minutes.
  • the fused cells were spun down 400 x g 6 min and resuspended in 20 ml selection media (DMEM (Invitrogen), 15 % FBS (Hyclone), supplemented with L- glutamine, pen/strep, MEM Non-essential amino acids, Sodium Pyruvate, 2- Mercaptoethanol (all from Invitrogen), HA-Azaserine Hypoxanthine and OPI (oxaloacetate, pyruvate, bovine insulin) (both from Sigma) and IL-6 (Boehringer Mannheim)) per million B-cells.
  • the present example outlines how the various PCSK9 antigen binding proteins were characterized and selected.
  • the binding of secreted antibodies produced from the hybridomas produced in Examples 1 and 2) to PCSK9 was assessed. Selection of antibodies was based on binding data and inhibition of PCSK9 binding to LDLR and affinity. Binding to soluble PCSK9 was analyzed by ELISA, as described below. BIAcore ® (surface plasmon resonance) was used to quantify binding affinity.
  • a primary screen for antibodies which bind to wild-type PCSK9 was performed.
  • the primary screen was performed on two harvests.
  • the primary screen comprised an ELISA assay and was performed using the following protocol:
  • Costar 3702 medium binding 384 well plates (Corning Life Sciences) were employed. The plates were coated with neutravadin at a concentration of 4 ⁇ g/ml in lXPBS/0.05%Azide, at a volume of 40 ⁇ /well. The plates were incubated at 4°C overnight. The plates were then washed using a Titertek plate washer (Titertek, Huntsville, AL). A 3 -cycle wash was performed. The plates were blocked with 90 ⁇ of 1XPBS/I%milk and incubated approximately 30 minutes at room temperature. The plates were then washed. Again, a 3 -cycle wash was performed.
  • the capture sample was biotinylated-PCSK9, without a V5 tag, and was added at 0.9 ⁇ g/ml in lXPBS/l%milk/10mM Ca 2+ at a volume of 40 ⁇ /well.
  • the plates were then incubated for 1 hour at room temperature.
  • the plates were washed using the Titertek plate washer operated using a 3-cycle wash.
  • 10 ⁇ of supernatant was transferred into 40 ⁇ of lXPBS/l%milk/10mM Ca 2+ and incubated 1.5 hours at room temperature. Again the plates were washed using the Titertek plate washer operated using a 3-cycle wash.
  • the primary screen resulted in a total of 3104 antigen specific hybridomas being identified from the two harvests. Based on highest ELISA OD, 1500 hybridomas per harvest were advanced for a total of 3000 positives.
  • the 3000 positives were then rescreened for binding to wild-type PCSK9 to confirm stable hybridomas were established.
  • the screen was performed as follows: Costar 3702 medium binding 384 well plates (Corning Life Sciences) were employed. The plates were coated with neutravadin at 3 ⁇ g/ml in lXPBS/0.05%Azide at a volume of 40 ⁇ /well. The plates were incubated at 4°C overnight. The plates were then washed using a Titertek plate washer (Titertek, Huntsville, AL). A 3-cycle wash was performed. The plates were blocked with 90 ⁇ of 1XPBS/I%milk and incubated approximately 30 minutes at room temperature. The plates were then washed using the M384 plate washer. A 3-cycle wash was performed.
  • the capture sample was b- PCSK9, without a V5 tag, and was added at 0.9 ⁇ g/ml in lXPBS/l%milk/10mM Ca 2+ at a volume of 40 ⁇ /well.
  • the plates were then incubated for 1 hour at room temperature. Next, the plates were washed using a 3-cycle wash. 10 ⁇ of supernatant was transferred into 40 ⁇ of lXPBS/l%milk/10mM Ca 2+ and incubated 1.5 hours at room temperature. Again the plates were washed using the Titertek plate washer operated using a 3-cycle wash.
  • the panel of hybridomas was then screened for cross-reactivity to mouse PCSK9 to make certain that the antibodies could bind to both human and mouse PCSK9.
  • the following protocol was employed in the cross-reactivity screen: Costar 3702 medium binding 384 well plates (Corning Life Sciences) were employed. The plates were coated with neutravadin at 3 g/ml in lXPBS/0.05%Azide at a volume of 40 ⁇ /well. The plates were incubated at 4°C overnight. The plates were then washed using a Titertek plate washer (Titertek, Huntsville, AL). A 3-cycle wash was performed. The plates were blocked with 90 ⁇ of 1XPBS/I%milk and incubated approximately 30 minutes at room temperature.
  • the plates were then washed using the Titertek plate washer. A 3-cycle wash was performed.
  • the capture sample was biotinylated-mouse PCSK9, and was added at 1 ⁇ in lXPBS/l%milk/10mM Ca 2+ at a volume of 40 ⁇ /well.
  • the plates were then incubated for 1 hour at room temperature.
  • the plates were washed using the Titertek plate washer operated using a 3-cycle wash. 50 ⁇ of supernatant was transferred to the plates and incubated 1 hour at room temperature. Again the plates were washed using a 3-cycle wash.
  • the D374Y mutation in PCSK9 has been documented in the human population (e.g., Timms KM et al, "A mutation in PCSK9 causing autosomal-dominant hypercholesterolemia in a Utah pedigree", Hum. Genet. 1 14: 349-353, 2004).
  • the samples were then screened for binding to the mutant PCSK9 sequence comprising the mutation D374Y.
  • the protocol for the screen was as follows: Costar 3702 medium binding 384 well plates (Corning Life Sciences) were employed in the screen.
  • the plates were coated with neutravadin at 4 ⁇ g/ml in IXPBS/O.05% Azide at a volume of 40 ⁇ /well. The plates were incubated at 4°C overnight. The plates were then washed using a Titertek plate washer (Titertek, Huntsville, AL). A 3-cycle wash was performed. The plates were blocked with 90 ⁇ of 1XPBS/I%milk and incubated approximately 30 minutes at room temperature. The plates were then washed using the Titertek plate washer. A 3-cycle wash was performed. The plates were coated with biotinylated human PCSK9 D374Y at a concentration of 1 ⁇ g/ml in lXPBS/l%milk/10mMCa 2+ and incubated for 1 hour at room temperature.
  • Late exhaust hybridoma culture supernatant was diluted 1 :5 in PBS/milk/Ca 2+ (10 ml plus 40 ml) and incubated for 1 hour at room temperature.
  • 40 ⁇ /well of rabbit anti-human PCSK9 (Cayman Chemical) and human anti-His 1.2.3 1 :2 at lug/ml in lXPBS/l%milk/10mMCa 2+ was titrated onto the plates, which were then incubated for 1 hour at room temperature.
  • the plates were then washed using a Titertek plate washer. A 3-cycle wash was performed.
  • an assay was developed using the D374Y PCSK9 mutant.
  • the mutant was used for this assay because it has a higher binding affinity to LDLR allowing a more sensitive receptor ligand blocking assay to be developed.
  • the following protocol was employed in the receptor ligand blocking screen: Costar 3702 medium binding 384 well plates (Corning Life Sciences) were employed in the screen. The plates were coated with goat anti-LDLR (R&D Cat #AF2148) at 2 ⁇ in lXPBS/0.05%Azide at a volume of 40 ⁇ /well. The plates were incubated at 4°C overnight. The plates were then washed using a Titertek plate washer (Titertek, Huntsville, AL).
  • a 3-cycle wash was performed.
  • the plates were blocked with 90 ⁇ of 1XPBS/1% milk and incubated approximately 30 minutes at room temperature. The plates were then washed using the Titertek plate washer.
  • a 3-cycle wash was performed.
  • the capture sample was LDLR (R&D, Cat #2148LD/CF), and was added at 0.4 ⁇ in lXPBS/l%milk/10mM Ca 2+ at a volume of 40 ⁇ /well. The plates were then incubated for 1 hour and 10 minutes at room temperature.
  • the receptor ligand assay was then repeated using the mutant enzyme on the 384 member subset of neutralizers identified in the first large scale receptor ligand inhibition assay.
  • the same protocol was employed in the screen of the 384 member blocker subset assay as was done in the large scale receptor ligand blocking screen. This repeat screen confirmed the initial screening data.
  • This screen of the 384 member subset identified 85 antibodies that blocked interaction between the PCSK9 mutant enzyme and the LDLR greater than 90%.
  • the plates were blocked with 90 ⁇ of 1XPBS/I%milk and incubated approximately 30 minutes at room temperature. The plates were then washed using the Titertek plate washer. A 3- cycle wash was performed. LDLR (R&D Systems, #2148LD/CF or R&D Systems, #2148LD) was added at 5 ⁇ g/ml in lXPBS/l%milk/10mM Ca 2+ at a volume of 40 ⁇ /well. The plates were then incubated for 1 hour at room temperature. Next, the plates were washed using the Titertek plate washer operated using a 3-cycle wash. Contemporaneously, biotinylated human wild-type PCSK9 was pre-incubated with hybridoma exhaust supernatant in Nunc polypropylene plates.
  • the plates were pre-incubated for approximately 1 hour and 30 minutes at room temperature. 50 ⁇ /well of the preincubated mixture was transferred onto LDLR captured ELISA plates and incubated for 1 hour at room temperature. The plates were then washed using the Titertek plate washer. A 3 -cycle wash was performed.
  • hybridoma line number e.g. 21B12
  • clone number e.g. 21B12.1
  • 25A7.1 was found not to block PCSK9/LDLR but 25A7.3 (referred to herein as 25A7) was neutralizing.
  • the isolated clones were each expanded in 50-100 ml of hybridoma media and allowed to grow to exhaustion, (i.e., less than about 10% cell viability).
  • concentration and potency of the antibodies to PCSK9 in the supernatants of those cultures were determined by ELISA and by in vitro functional testing, as described herein.
  • the hybridomas with the highest titer of antibodies to PCSK9 were identified.
  • the selected hybridomas are shown in FIGS 2A-3D and Table 2.
  • This example generally describes how one of the antigen binding proteins was produced from a hybridoma line.
  • the production work used 50ml exhaust supernatant generation followed by protein A purification. Integra production was for scale up and was performed later.
  • Hybridoma line 31H4 was grown in T75 flasks in 20 ml of media (Integra Media, Table 5). When the hybridoma was nearly confluent in the T75 flasks, it was transferred to an Integra flask (Integra Biosciences, Integra CL1000, cat# 90 005).
  • the Integra flask is a cell culture flask that is divided by a membrane into two chambers, a small chamber and a large chamber.
  • a volume of 20-30 ml hybridoma cells at a minimum cell density of lxl 0 6 cells per ml from the 31H4 hybridoma line was placed into the small chamber of an Integra flask in Integra media (see Table 4.1 for components of Integra media).
  • Integra media alone (1L) was placed in the large chambers of the Integra flasks.
  • the membrane separating the two chambers is permeable to small molecular weight nutrients but is impermeable to hybridoma cells and to antibodies produced by those cells. Thus, the hybridoma cells and the antibodies produced by those hybridoma cells were retained in the small chamber.
  • Bound antibody proteins on the Protein A column were recovered by standard acidic antibody elution from Protein A columns (such as 50 mM Citrate, pH 3.0). Aggregated antibody proteins in the Protein A Sepharose pool were removed by size exclusion chromatography or binding ion exchange chromatography on anion exchanger resin such as Q Sepharose resin. The specific IEX conditions for the 31H4 proteins are Q-Sepharose HP at pH 7.8-8.0. Antibody was eluted with a NaCl gradient of 10 mM-500 mM in 25 column volumes.
  • the present example outlines how 31H4 IgG2 antibodies were produced from transfected cells.
  • 293 cells for transient expression and CHO cells for stable expression were transfected with plasmids that encode 31H4 heavy and light chains.
  • Conditioned media from transfected cells was recovered by removing cells and cell debris. Clarified conditioned media was loaded onto a Protein A-Sepharose column.
  • Clarified conditioned media was loaded onto a Protein A-Sepharose column.
  • the media can first be concentrated and then loaded onto a Protein A Sepharose column. Non-specific bindings were removed by extensive PBS wash.
  • Bound antibody proteins on the Protein A column were recovered by standard acidic antibody elution from Protein A columns (such as 50 mM citrate, pH 3.0).
  • Aggregated antibody proteins in the Protein A Sepharose pool were removed by size exclusion chromatography or binding ion exchange chromatography on anion exchanger resin such as Q Sepharose resin.
  • the specific IEX conditions for the 31H4 proteins are Q-Sepharose HP at pH 7.8-8.0.
  • the antibody was eluted with a NaCl gradient of 10 mM-500 mM in 25 column volumes.
  • Hybridoma line 2 IB 12 was grown in T75 flasks in media (Integra Media, Table 5). When the hybridomas were nearly confluent in the T75 flasks, they were transferred to Integra flasks (Integra Biosciences, Integra CL1000, cat# 90 005).
  • the Integra flask is a cell culture flask that is divided by a membrane into two chambers, a small chamber and a large chamber.
  • a volume of 20-30 ml hybridoma cells at a minimum cell density of lxl 0 6 cells per ml from the 31H4 hybridoma line was placed into the small chamber of an Integra flask in Integra media (see Table 5 for components of Integra media).
  • Integra media alone (1L) was placed in the large chambers of the Integra flasks.
  • the membrane separating the two chambers is permeable to small molecular weight nutrients but is impermeable to hybridoma cells and to antibodies produced by those cells. Thus, the hybridoma cells and the antibodies produced by those hybridoma cells were retained in the small chamber.
  • Bound antibody proteins on the Protein A column were recovered by standard acidic antibody elution from Protein A columns (such as 50 mM Citrate, pH 3.0). Aggregated antibody proteins in the Protein A Sepharose pool were removed by size exclusion chromatography or binding ion exchange chromatography on anion exchanger resin such as Q Sepharose resin. The specific IEX conditions for the 2 IB 12 proteins are Q- Sepharose HP at pH 7.8-8.0. The antibody was eluted with a NaCl gradient of 10 mM- 500 mM in 25 column volumes.
  • the present example outlines how 2 IB 12 IgG2 antibodies were produced from transfected cells.
  • Cells (293 cells for transient expression and CHO cells for stable expression) were transfected with plasmids that encode 2 IB 12 heavy and light chains.
  • Conditioned media from hybridoma cells were recovered by removing cells and cell debris. Clarified conditioned media were loaded onto a Protein A-Sepharose column.
  • Clarified conditioned media were loaded onto a Protein A-Sepharose column.
  • the media can first be concentrated and then loaded onto a Protein A Sepharose column. Non-specific bindings were removed by extensive PBS wash. Bound antibody proteins on the Protein A column were recovered by standard acidic antibody elution from Protein A columns (50 mM Citrate, pH 3.0).
  • Aggregated antibody proteins in the Protein A Sepharose pool were removed by size exclusion chromatography or binding ion exchange chromatography on cation exchanger resin such as SP-Sepharose resin.
  • the specific IEX conditions for the 2 IB 12 proteins were SP-Sepharose HP at pH 5.2.
  • Antibodies were eluted with 25 column volumes of buffer that contains a NaCl gradient of 10 mM-500 mM in 20 mM sodium acetate buffer.
  • nucleic acid and amino acid sequences for the light and heavy chains of the above antibodies were then determined by Sanger (dideoxy) nucleotide sequencing. Amino acid sequences were then deduced for the nucleic acid sequences. The nucleic acid sequences for the variable domains are depicted in FIG.s 3E-3JJ and 3LL-BBB.
  • the cDNA sequences for the lambda light chain variable regions of 31H4, 21B12, and 16F12 were determined and are disclosed as SEQ ID NOs: 153, 95, and 105 respectively.
  • the cDNA sequences for the heavy chain variable regions of 31H4, 2 IB 12, and 16F12 were determined and are disclosed as SEQ ID NOs: 152, 94, and 104 respectively.
  • the lambda light chain constant region (SEQ ID NO: 156), and the IgG2 and IgG4 heavy chain constant regions (SEQ ID NOs: 154 and 155) are shown in FIG. 3K .
  • the polypeptide sequences predicted from each of those cDNA sequences were determined.
  • the predicted polypeptide sequences for the lambda light chain variable regions of 31H4, 21B12, and 16F12 were predicted and are disclosed as SEQ ID NOs: 12, 23, and 35 respectively, the lambda light chain constant region (SEQ ID NO: 156), the heavy chain variable regions of 31H4, 2 IB 12, and 16F12 were predicted and are disclosed as (SEQ. ID NOs. 67, 49, and 79 respectively.
  • the IgG2 and IgG4 heavy chain constant regions SEQ ID NOs: 154 and 155).
  • the FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 divisions are shown in FIG 2A-3D and FIG. 3CCC-3JJJ.
  • FIGs. 2A-3D and FIG. 3CCC-3JJJ Based on the sequence data, the germline genes from which each heavy chain or light chain variable region was derived was determined. The identity of the germline genes are indicated next to the corresponding hybridoma line in FIGs. 2A-3D and FIG. 3CCC-3JJJ and each is represented by a unique SEQ ID NO. FIGs. 2A-3D and FIG. 3CCC-3JJJ also depict the determined amino acid sequences for additional antibodies that were characterized.
  • FIG. 3LLL highlights (via underlining shading, or bold) the differences between the constant regions of the 21B12, 31H4, and 31A4 Fabs (raised in E. coli) when compared to SEQ ID NOs: 156, and 155.
  • the light chain constant sequence is similar to human lambda (SEQ ID NO: 156).
  • the underlined glycine residue is an insertion between where the 2 IB 12 and 31H4 variable sequences stop and the lambda sequence starts.
  • 31A4 while it also has the same distinctions noted above, there are three additional differences. As shown in FIG. 3LLL, there are two additional amino acids at the start, which comes from incomplete processing of the signal peptide in E. coli expression. In addition, there is one additional substitution in the 31A4 heavy chain constant region when compared to SEQ ID NO: 155, which is the adjustment of a L (in SEQ ID NO: 155) to a H. Finally, 31A4 does have a glutamine as the initial amino acid of the Fab, rather than the the adjustment to glutamic acid noted above for 21B12 and 31H4.
  • the end of the heavy chain differs as well, but the amino acids are not ordered in the structure so they do not appear in the cooridnates.
  • his-tags are not a required part of the ABP and should not be considered as part of the ABP's sequence, unless explicitly called out by reference to a specific SEQ ID NO that includes a histidine tag and a statement that the ABP sequence "includes the Histidine tag.”
  • a BIAcore ® surface plasmon resonance device, Biacore, Inc., Piscataway, NJ affinity analysis of the 2 IB 12 antibodies to PCSK9 described in this Example was performed according to the manufacturer's instructions.
  • the binding affinities of antibody to human PCSK9 were also measured at pH 6.0 with the pH 6.0 HBS-P buffer (pH 6.0, 0.01 M Hepes, 0.15 M NaCl, 0.005% surfactant P-20, Biacore) supplemented with 0.01% BSA.
  • the binding signal obtained was proportional to the free PCSK9 in solution.
  • the antibodies appeared to display a tighter binding affinity at the lower pH (where the Kd was 12.5, 7.3, and 29 pM for 31H4, 21B12, and 16F12 respectively).
  • Antibody binding kinetic parameters including k a (association rate constant), k d (dissociation rate constant), and K D (dissociation equilibrium constant) were determined using the BIA evaluation 3.1 computer program (BIAcore, Inc. Piscataway, NJ). Lower dissociation equilibrium constants indicate greater affinity of the antibody for PCSK9.
  • the K D values determined by the BIAcore ® affinity analysis are presented in Table 9.2, shown below.
  • Table 9.3 depicts the k on and k 0ff rates.
  • a KinExA ® (Sapidyne Instruments, Inc., Boise, ID) affinity analysis of 16F12 and 31H4 was performed according to the manufacturer's instructions. Briefly, Reacti- GelTM (6x) (Pierce) was pre-coated with one of human, V5-tagged cyno or His-tagged mouse PCSK9 proteins and blocked with BSA. 10 or 100 pM of antibody 31H4 and one of the PCSK9 proteins was then incubated with various concentrations (0.1 pM - 25 nM) of PCSK9 proteins at room temperature for 8 hours before being passed through the PCSK9-coated beads.
  • Reacti- GelTM (6x) (Pierce) was pre-coated with one of human, V5-tagged cyno or His-tagged mouse PCSK9 proteins and blocked with BSA. 10 or 100 pM of antibody 31H4 and one of the PCSK9 proteins was then incubated with various concentrations (0.1 pM - 25 nM) of PCSK9
  • the amount of the bead-bound 31H4 was quantified by fluorescently (Cy5) labeled goat anti-human IgG (H+L) antibody (Jackson Immuno Research).
  • the binding signal is proportional to the concentration of free 31H4 at binding equilibrium.
  • Equilibrium dissociation constant (K D ) were obtained from nonlinear regression of the two sets of competition curves using a one-site homogeneous binding model.
  • the KinExA ® Pro software was employed in the analysis. Binding curves generated in this analysis are presented as FIGs. 4A-4F.
  • Both the 16F12 and 31H4 antibodies showed similar affinity to human and cyno PCSK9, but approximately 10-250 fold lower affinity to mouse PCSK9.
  • antibody 31H4 showed higher affinity to both human and cyno PCSK9 with 3 and 2 pM K D , respectively.
  • 16F12 showed slightly weaker affinity at 15pM K D to human PCSK9 and 16 pM K D to cyno PCSK9.
  • a BIAcore solution equilibrium binding assay was used to measure the Kd values for ABP 21B12. 21B12.1 showed little signal using KinExA assay, therefore, biacore solution equilibrium assay was applied. Since no significant binding was observed on binding of antibodies to immobilized PCSK9 surface, 2 IB 12 antibody was immobilized on the flow cell 4 of a CM5 chip using amine coupling with density around 7000 RU. Flow cell 3 was used as a background control. 0.3, 1, and 3 nM of human PCSK9 or cyno PCSK9 were mixed with a serial dilutions of 21B12.1 antibody samples (ranged from 0.001 ⁇ 25 nM) in PBS plus O. lmg/ml BSA, 0.005% P20.
  • Binding of the free PCSK9 in the mixed solutions were measured by injecting over the 21B12.1 antibody surface. 100% PCSK9 binding signal on 21B12.1 surface was determined in the absence of mAb in the solution. A decreased PCSK9 binding response with increasing concentrations of mAb indicated that PCSK9 binding to mAb in solution, which blocked PCSK9 from binding to the immobilized peptibody surface. Plotting the PCSK9 binding signal versus mAb concentrations, K D was calculated from three sets of curves (0.3, 1 and 3nM fixed PCSK9 concentration) using a one-site homogeneous binding model in KinExA ProTM software.
  • FIG. 5B depicts the results from the solution equilibrium assay at three different hPCSK9 concentrations for hPCSK9.
  • FIG. 5C depicts a similar set of results for mPCSK9.
  • FIG. 5D depicts the results from the above biacore capture assay.
  • This example provides the IC50 values for two of the antibodies in blocking
  • PCSK9 D374Y's ability to bind to LDLR Clear 384 well plates (Costar) were coated with 2 micrograms/ml of goat anti-LDL receptor antibody (R&D Systems) diluted in buffer A (100 mM sodium cacodylate, pH 7.4). Plates were washed thoroughly with buffer A and then blocked for 2 hours with buffer B (1% milk in buffer A). After washing, plates were incubated for 1.5 hours with 0.4 micrograms/ml of LDL receptor (R&D Systems) diluted in buffer C (buffer B supplemented with 10 mM CaC12).
  • buffer A 100 mM sodium cacodylate, pH 7.4
  • biotinylated D374Y PCSK9 20 ng/ml of biotinylated D374Y PCSK9 was incubated with various concentrations of the 31H4 IgG2, 31H4 IgG4, 2 IB 12 IgG2 or 2 IB 12 IgG4 antibody, which was diluted in buffer A, or buffer A alone (control).
  • the LDL receptor containing plates were washed and the biotinylated D374Y
  • FIGs. 6A-6D The results of this binding study are shown in FIGs. 6A-6D.
  • IC 50 values were determined for each antibody and found to be 199 pM for 31H4 IgG2 (FIG. 6A), 156 pM for 31H4 IgG4 (FIG. 6B), 170 pM for 21B12 IgG2 (FIG. 6C), and 169 pM for 21B12 IgG4 (FIG. 6D).
  • the antibodies also blocked the binding of wild-type PCSK9 to the LDLR in this assay.
  • the D374Y PCSK9/antibody mixture was transferred to the cells, followed by LDL-BODIPY (Invitrogen) diluted in uptake buffer at a final concentration of 6 ⁇ g/ml. After incubation for 3 hours at 37°C (5% C02), cells were washed thoroughly with PBS and the cell fluorescence signal was detected by SafireTM (TECAN) at 480-520nm (excitation) and 520-600nm (emission).
  • LDL-BODIPY Invitrogen
  • FIGs. 7A-7D The results of the cellular uptake assay are shown in FIGs. 7A-7D. Summarily, IC 50 values were determined for each antibody and found to be 16.7 nM for 31H4 IgG2 (FIG. 7A), 13.3 nM for 31H4 IgG4 (FIG. 7B), 13.3 nM for 21B12 IgG2 (FIG. 7C), and 18 nM for 2 IB 12 IgG4 (FIG. 7D). These results demonstrate that the applied antigen binding proteins can reduce the effect of PCSK9 (D374Y) to block LDL uptake by cells The antibodies also blocked the effect of wild-type PCSK9 in this assay.
  • mice were sacrificed with C02 asphyxiation at the pre-determined time points shown in Table 9. Blood was collected via vena cava into eppendorf tubes and was allowed to clot at room temperature for 30 minutes. The samples were then spun down in a table top centrifuge at 12,000xg for 10 minutes to separate the serum. Serum total cholesterol and HDL-C were measured using Hitachi 912 clinical analyzer and Roche/Hitachi TC and HDL-C kits.
  • mice to which antibody 31H4 was administered showed decreased serum cholesterol levels over the course of the experiment (FIG. 8A and FIG. 8B).
  • the mice also showed decreased HDL levels (FIG. 8C and FIG. 8D).
  • the percentage change is in relation to the control IgG at the same time point (*P ⁇ 0.01, # P ⁇ 0.05).
  • mice transport the majority of serum cholesterol in high density lipoprotein (HDL) particles which is different to humans who carry most serum cholesterol on LDL particles. In mice the measurement of total serum cholesterol most closely resembles the level of serum HDL-C.
  • Mouse HDL contains apolipoprotein E (apoE) which is a ligand for the LDL receptor (LDLR) and allows it to be cleared by the LDLR.
  • apoE apolipoprotein E
  • HDL in contrast, does not contain apoE and is not a ligand for the LDLR.
  • PCSK9 antibodies increase LDLR expression in mouse, the liver can clear more HDL and therefore lowers serum HDL-C levels.
  • the present example demonstrates that an antigen binding protein alters the level of LDLR in a subject, as predicted, over time.
  • a Western blot analysis was performed in order to ascertain the effect of antibody 31H4 on LDLR levels.
  • 50-100 mg of liver tissue obtained from the sacrificed mice described in Example 11 was homogenized in 0.3 ml of RIPA buffer (Santa Cruz Biotechnology Inc.) containing complete protease inhibitor (Roche). The homogenate was incubated on ice for 30 minutes and centrifuged to pellet cellular debris. Protein concentration in the supernatant was measured using BioRad protein assay reagents (BioRad laboratories).
  • the blot was washed briefly and incubated with bovine anti-goat IgG-HRP (Santa Cruz Biotechnology Inc.) 1 :2000 or goat anti-mouse IgG-HRP (Upstate) 1 :2000. After a 1 hour incubation at room temperature, the blot was washed thoroughly and immunoreactive bands were detected using ECL plus kit (Amersham biosciences). The Western blot showed an increase in LDLR protein levels in the presence of antibody 31H4, as depicted in FIG. 9.
  • FIG. 10A depicts the results of this experiment.
  • FIG. IOC depicts the results of repeating the above procedure with the lOmg/kg dose of 31H4, and with another antibody, 16F12, also at lOmg/kg. Dosing groups and time of sacrifice are shown in Table 14.2.
  • both 16F12 and 31H4 resulted in significant and substantial decreases in total serum cholesterol after just a single dose and provided benefits for over a week (10 days or more).
  • the results of the repeated 13 day study were consistent with the results of the first 13 day study, with a decrease in serum cholesterol levels of 26% on day 3 being observed.
  • the percentage change is in relation to the control IgG at the same time point (*P ⁇ 0.01).
  • the percentage change is in relation to the control IgG at the same time point (*P ⁇ 0.05).
  • Example 14 The HDL levels for the animals in Example 14 were also examined. HDL levels decreased in the mice. More specifically, animals dosed at 10 mg/kg demonstrated a 33% decrease in HDL levels on day 3, which gradually returned to pre- dosing levels by day 13.
  • FIG. 10B depicts the results of the experiment. There was a decrease in HDL levels of 34% on day 3.
  • FIG. 10B depicts the results of the repeated 13 day experiment.
  • mice will lower mouse HDL, this is not expected to occur in humans because of the differences in HDL in humans and other organisms (such as mice). Thus, the decrease in mouse HDL is not indicative of a decrease in human HDL.
  • a human patient exhibiting symptoms of hypercholesterolemia is administered a therapeutcially effective amount of PCSK9 antibody, such as 31H4 (or, for example, 2 IB 12).
  • PCSK9 antibody such as 31H4 (or, for example, 2 IB 12).
  • the human patient is monitored to determine whether the serum cholesterol level has declined.
  • the patient receiving the treatment with the PCSK9 antibodies has reduced serum cholesterol levels in comparison to arthritis patients not receiving the treatment.
  • PCSK9 Antigen Binding Protein for the Prevention of Hypercholesterolemia
  • a human patient exhibiting a risk of developing hypercholesterolemia is identified via family history analysis and/or lifestyle, and/or current cholesterol levels.
  • the subject is regularly administered (e.g., one time weekly) a therapeutically effective amount of PCSK9 antibody, 31H4 (or, for example, 21B12).
  • the patient is monitored to determine whether serum cholesterol levels have decreased.
  • subjects undergoing preventative treatment with the PCSK9 antibody have lowered serum cholesterol levels, in comparison to subjects that are not treated.
  • PCSK9 ABPs Further Upregulated LDLR in the Presence of Statins
  • HepG2 cells were seeded in DMEM with 10% fetal bovine serum (FBS) and grown to -90% confluence. The cells were treated with indicated amounts of mevinolin (a statin, Sigma) and PCSK9 ABPs (FIGs. 12A-12C) in DMEM with 3% FBS for 48 hours. Total cell lysates were prepared. 50 mg of total proteins were separated by gel electrophoresis and transferred to PVDF membrane. Immunoblots were performed using rabbit anti-human LDL receptor antibody (Fitzgerald) or rabbit anti-human b-actin antibody. The enhanced chemiluminescent results are shown in the top panels of FIGs. 12A-12C.
  • FBS fetal bovine serum
  • ABPs 21B12 and 31H4 are PCSK9 neutralizing antibodies, while 25A7.1 is a non-neutralizing antibody.
  • HepG2-PCSK9 cells were also created. These were stable HepG2 cell line transfected with human PCSK9. The cells were seeded in DMEM with 10% fetal bovine serum (FBS) and grew to -90% confluence. The cells were treated with indicated amounts of mevinolin (Sigma) and PCSK9 ABPs (FIGs. 12D-12F) in DMEM with 3% FBS for 48 hours. Total cell lysates were prepared. 50 mg of total proteins were separated by gel electrophoresis and transferred to PVDF membrane. Immunoblots were performed using rabbit anti-human LDL receptor antibody (Fitzgerald) or rabbit anti-human b-actin antibody. The enhanced chemiluminescent results are shown in the top panels. The intensity of the bands were quantified by ImageJ software and normalized by b-actin.
  • FBS fetal bovine serum
  • Consensus sequences were determined using standard phylogenic analyses of the CDRs corresponding to the V H and V L of anti-PCSK9 ABPs. The consensus sequences were determined by keeping the CDRs contiguous within the same sequence corresponding to a V H or V L . Briefly, amino acid sequences corresponding to the entire variable domains of either V H or V L were converted to FASTA formatting for ease in processing comparative alignments and inferring phylogenies.
  • framework regions of these sequences were replaced with an artificial linker sequence ("bbbbbbbbbbbbbbbb” placeholders, non-specific nucleic acid construct) so that examination of the CDRs alone could be performed without introducing any amino acid position weighting bias due to coincident events (e.g., such as unrelated antibodies that serendipitously share a common germline framework heritage) while still keeping CDRs contiguous within the same sequence corresponding to a V H or V L .
  • V H or V L sequences of this format were then subjected to sequence similarity alignment interrogation using a program that employs a standard ClutalW-like algorithm (see, Thompson et ah, 1994, Nucleic Acids Res. 22:4673-4680).
  • a gap creation penalty of 8.0 was employed along with a gap extension penalty of 2.0.
  • This program likewise generated phylograms (phylogenic tree illustrations) based on sequence similarity alignments using either UPGMA (unweighted pair group method using arithmetic averages) or Neighbor- Joining methods (see, Saitou and Nei, 1987, Molecular Biology and Evolution 4:406-425) to construct and illustrate similarity and distinction of sequence groups via branch length comparison and grouping. Both methods produced similar results but UPGMA-derived trees were ultimately used as the method employs a simpler and more conservative set of assumptions.
  • UPGMA-derived trees were generated where similar groups of sequences were defined as having fewer than 15 substitutions per 100 residues (see, legend in tree illustrations for scale) amongst individual sequences within the group and were used to define consensus sequence collections. The results of the comparisons are depicted in FIGs. 13A-13J and FIGs. 31A and 3 IB. In FIG. 13E, the groups were chosen so that sequences in the light chain that clade are also a clade in the heavy chain and have fewer than 15 substitutions.
  • Results from this assay demonstrate that 1 IFl binds to PCSK9 and not to PCSKl, PCSK2, PCSK7, or furin, demonstrating the specificity of 1 IFl for PCSK9.
  • Biotinylated PCSK9 diluted in buffer A (25 mM Tris, 150 mM NaCl, 0.1% BSA, 0.05%) tween, pH 7.5) was bound to neutravidin coated 96 well plates at a concentration of 0.2 ⁇ g/mL, for one hour incubation at room temperature.
  • This assay relied on the ability of proprotein convertase in solution to compete for the binding of 1 IFl to plate-captured PCSK9.
  • Pre-incubation of 1 IFl and PCSK9 in solution dose dependently and robustly reduced the amount of 1 IF 1 binding to plate- captured PCSK9 detected as reduced OD450 (FIG. 14). All results were expressed as the mean OD450 value ⁇ standard deviation versus concentration of the proprotein convertase.
  • Pre-incubation of 1 IFl with PCSKl, PCSK2, PCSK7, or furin, in solution did not significantly impact the binding of 1 IFl to plate-captured PCSK9. Therefore, at the protein concentrations studied, 1 IFl binds only to PCSK9 and not to the other proprotein convertase family members tested.
  • the example demonstrates that nanomolar concentrations of 1 IF 1 can inhibit binding of both D374Y and wild-type PCSK9 to the LDLR under the conditions of this assay.
  • Binding of the biotinylated PCSK9 to the LDLR was detected by incubation with streptavidin-HRP (500 ng/mL in buffer C) followed by TMB substrate. The reaction was stopped with IN HCl and the absorbance was read at a wavelength of 450 nm on a SpectraMax Plus 384 Spectrophotometer (Molecular Devices Inc., Sunnyvale, CA). GraphPad Prism (v 4.01) software was used to plot log of antibody concentration versus OD450 to determine IC50 values by nonlinear regression.
  • IC50 values are dependent on the amount of recombinant D374Y PCSK9 or WT PCSK9 used in the binding assay.
  • a representative dose response curve for both the D374Y and wild-type assays are presented in FIG. 15 and FIG. 16, respectively.
  • Efficacy of 1 IF 1 in Blocking Cell LDL Uptake 11F1 blocks the interaction between PCSK9 and LDLR in vitro and can prevent the PCSK9-mediated reduction of LDL uptake in HepG2 cells.
  • HepG2 cells were seeded in black, clear bottom 96-well plates (Fisher Scientific CO LLC, Santa Clara, CA) at a density of 5x104 cells per well in DMEM (Mediatech Inc., Herndon, VA) supplemented with 10% FBS and 1% of antibiotic-antimycotic solution (Mediatech Inc., Herndon, VA). Cells were incubated at 37°C (5% C02) overnight.
  • serial dilutions (1 :2) of 11F1 from 666.7 nM to 0.7 nM (for blocking D374Y PCSK9) or from 3.3 ⁇ to 3.3 nM (for blocking WT PCSK9), were prepared in formulation buffer (25 mM HEPES, pH 7.5, 0.15 M NaCL).
  • Either D374Y PCSK9 (2 ⁇ g/mL) or WT PCSK9 (25 ⁇ g/mL) were diluted in uptake buffer (DMEM containing 1% FBS) and incubated with the various concentrations of 1 IF 1 or uptake buffer alone (negative control) for 1 hour at room temperature with shaking.
  • BODIPY-LDL (Invitrogen, Carlsbad, CA) was diluted in uptake buffer to a concentration of 12 ⁇ g/mL. Following overnight incubation, HepG2 cells were rinsed twice with DPBS (Mediatech Inc., Herndon, VA). Twenty-five microliters of the D374Y PCSK9 or WT PCSK9 complex with 11F1 and 25 ⁇ , of diluted BODIPY-LDL (Invitrogen, Carlsbad, CA) were added to the cells and incubated at 37°C (5% C02) for 3 hours. Cells were washed with DPBS 5 times and resuspended in 100 ⁇ , DPBS. Fluorescent signals were detected using a Satire plate reader (Tecan Systems Inc., San Jose, CA) at 480-520 nm (excitation) and 520-600 nm (emission) and expressed as relative fluorescence unit (RFU).
  • GraphPad Prism (Version 4.02, GraphPad Software Inc., San Diego, CA) software was used to plot log of antibody concentration versus RFU and to determine EC50 values by nonlinear regression using the sigmoidal dose-response (variable slope) curve fitting program.
  • the EC50 values reported here are representative for mean values derived from 3 to 6 separate measurements for 1 IF 1.
  • a single intravenous bolus administration of the anti-PCSK9 antibodies 11F1 or 8A3 leads to a significant decrease in serum non-HDL-C and TC in mice expressing human PCSK9 by AAV.
  • This example demonstrates the effectiveness of both anti- PCSK9 antibodies in blocking the function of human PCSK9 in vivo.
  • mice expressing human PCSK9 were generated by infection with an engineered adeno associated virus (AAV) coding for human PCSK9, resulting in elevated levels of circulating low density lipoprotein cholesterol (LDL-C).
  • AAV engineered adeno associated virus
  • mice were then administered 11F1, 8 A3 or anti-keyhole limpethemocyanin (KLH) IgG2 control antibody at 30 mg/kg via tail vein injection.
  • KLH anti-keyhole limpethemocyanin
  • mice were euthanized and whole blood was collected from the vena cava and allowed to coagulate for 30 minutes at room temperature. Following centrifugation at 12,000 rpm with a bench top centrifuge for 10 minutes, serum was collected. Serum cholesterol analysis was performed using the Cobas Integra 400 plus chemistry analyzer.
  • Serum concentrations of PCSK9 were determined using a sandwich ELISA assay. Clear 96 well plates were coated overnight with 2 ⁇ g/ml of monoclonal anti- PCSK9 antibody (31H4) diluted in IX PBS. Plates were washed thoroughly with IX PBS/.05% tween and then blocked for 2 hours with 3% BSA/1XPBS. After washing, plates were incubated for 2 hours with serum diluted in general assay diluents
  • Serum concentrations of antibody were determined using a sandwich ELISA assay.
  • Polyclonal goat anti-human Fc IgG and an HRP-labeled goat anti-human IgG Fey polyclonal reagent both from Jackson ImmunoResearch Laboratories Inc, West Grove, PA were used as the capture and the detection antibody, respectively.
  • a 3,3 ',5,5 'tetramethylbenzidine (TMB) substrate solution reacted with peroxide, and in the presence of horse radish peroxidase (HRP), created a colorimetric signal that was proportional to the amount of the respective anti-PCSK9 antibody bound by the capture reagent.
  • TMB horse radish peroxidase
  • the intensity of the color was measured at 450 nm minus 650 nm using a microplate reader (Spectra Max Plus 384). Data was analyzed using Watson version 7.0.0.01 (Thermo Scientific, Waltham, MA) data reduction package with a Logistic (auto-estimate) regression of separately prepared standard curves. The lower limit of quantification (LLOQ) for the assay was ng/mL. 34.4.
  • NCA Non-compartmental analysis
  • mice expressing human PCSK9 were euthanized prior to injection of antibodies and blood was collected.
  • Non-HDL-C, HDL-C and TC levels in these animals were 33 ⁇ 4, 117 ⁇ 4 and 183 ⁇ 9 mg/dL, respectively (mean ⁇ SEM).
  • Levels of PCSK9 in naive animals were determined to be 4921 ng/niL ⁇ 2044 ng/niL.
  • injection of 8 A3 produced significant lowering of non-HDL-C at days 1, 2, and 4 post-injection (with a maximum of 65%), while TC was significantly lowered at day 2 post-injection (with a maximum of 24%) (FIG. 19, FIG. 20). No significant lowering of HDL-C was observed at any time point (FIG. 21).
  • Anti-KLH IgG2 control antibody CLO-t was similar to that of 11F1 and 8 A3, when calculated using the corrected dose, and the apparent half-life of the anti-KLH IgG2 control antibody was estimated at >120 hours. These data suggested that affects of the PCSK9 ligand on antibody disposition are less pronounced for 1 IF 1 and 8 A3 when compared to other antibodies dosed in the AAV model because 11F1 and 8 A3 CLO-t values are more similar to anti-KLH IgG2 control antibody.
  • Animals were then injected SC with either anti-KLH IgG2 control antibody, 11F1, 21B12, or 8A3 (all in 10 mM NaOAc pH 5.2, 9% sucrose) at 0.5 mg/kg (all at 0.4 mL/kg body weight). Fasting blood samples were then collected from animals at designated time points over a 45 day period.
  • the reagents for the assay were obtained from DiaSorin (Stillwater, MN).
  • Antibody concentrations in serum were determined using an enzyme-linked immunosorbent assay (ELISA) with an assay range of 34.4 to 3000 ng/mL (34.4 ng/mL being the lower limit of quantitation [LLOQ]).
  • ELISA enzyme-linked immunosorbent assay
  • NCA Non-compartmental analysis
  • Cmax concentration
  • AUCO-inf area under the serum concentration-time curve from time zero to infinity
  • CL/F apparent serum clearance
  • 2 IB 12 did induce a statistically significant change in HDL-C at a single time point (day 18 following injection) (FIG. 26 and FIG. 27).
  • Serum ApoB levels were measured at days 3, 6, 15, 24 and 33, post-injection. 11F1 and 8 A3 were associated with ApoB lowering at days 3 to 24, as compared to anti-KLH IgG2 control antibody-treated monkeys (FIG. 28). 2 IB 12 was associated with statistically significant lower ApoB levels at day 3 only.
  • Pharmacokinetic Profiles of 11F1, 21B12, and 8A3 A summary plot of the mean concentration-time profiles by treatment is shown in Figure 29. The estimated mean pharmacokinetic parameters for animals receiving 11F1, 21B12, 8 A3, and anti-KLH IgG2 control antibody are displayed in Table of Figure 30.
  • IF 1 achieved a maximal lowering of LDL-C (vs anti-KLH IgG2 control antibody at the same time point) 9 days following injection (-57%).
  • 21B12 achieved a maximal lowering of LDL-C (vs anti-KLH IgG2 control antibody at the same time point) 3 days following injection (-64%).
  • 8A3 achieved a maximal lowering of LDL-C (vs anti-KLH IgG2 control antibody at the same time point) 4 days following injection (-54%).
  • 21B12 lowered HDL-C at a single time point, 18 days after injection. No statistically significant changes were observed in HDL-C levels following 11F1 or 8 A3 administration. No statistically significant changes were observed in triglycerides levels following 11F1, 21B12, or 8 A3 administration.
  • Hyperchoesterolemia Study Design This is a 2 part study.
  • Part A is an open label, single arm, multicenter pilot study.
  • Part B is a double-blind, randomized, placebo-controlled, multicenter, study of antibody, 21B12, (heavy chain, SEQ ID NO:592 and light chain, SEQ ID NO:591) with expanded enrollment but otherwise identical design to Part A. Both inclusion/exclusion criteria and the Schedule of Assessments is the same for Parts A and B.
  • Inclusion Criteria includes: • Males and females > 12 to ⁇ 65 years of age
  • Exclusion Criteria includes:
  • NHA New York Heart Failure Association
  • Schedule of Assessments include, but are not limited to, collection of adverse event (AE) and significant adverse event (SAE) data, vital signs, concomitant medication, laboratory tests, etc.
  • AE adverse event
  • SAE significant adverse event
  • the 2 IB 12 formulation is presented as a sterile, clear, colorless frozen liquid.
  • Each sterile vial is filled with a 1-mL deliverable volume of 70 mg/mL 2 IB 12 formulated with 10 mM sodium acetate, 9%> (w/v) sucrose, 0.004%> (w/v) polysorbate 20, pH 5.2.
  • Each vial is for single use only.
  • Placebo is presented in identical containers as a clear, colorless, sterile, protein-free frozen liquid and is formulated as 10 mM sodium acetate, 9%> (w/v) sucrose, 0.004%> (w/v) polysorbate 20, pH 5.2.
  • Part A eight subjects with genetically confirmed homozygous familial hypercholesterolemia on stable lipid-lowering drug therapy for greater than (or equal to) 4 weeks are enrolled and received open label 2 IB 12 formulation.
  • Table 26.1 shows the genotypes of the patients in the study.
  • LDL-r Low density lipoprotein receptor.
  • the 2 IB 12 formulation (420 mg) is administered subcutaneous ly every 4 weeks for 12 weeks, followed by an additional 12 weeks of treatment at 4 week intervals, and then 12 weeks with AMG 145 420 mg administered every 2 weeks. Study visits occurr at least every 4 weeks. During these visits adverse event (AE) and significant adverse event (SAE) data, vital signs, concomitant medication, laboratory tests, etc. are collected.
  • AE adverse event
  • SAE significant adverse event
  • Q4W every 4 weeks; Q2W: every 2 weeks; SE: standard error; LDL: low-density lipoprotein; HDL: high-density lipoprotein; PCSK9: proprotein convertase subtilisin/kexin type 9.
  • Approximately 51 new subjects are enrolled into Part B. Subjects enrolled are randomized to a 2: 1 allocation into 2 treatment groups: 420 mg 2 IB 12 Q4W SC or placebo Q4W SC. Randomization is stratified by baseline LDL-C levels. Study visits occur every 4 weeks, with two optional visits occurring at week 2 and week 10. Visits entail collection of AE and SAE data, vital signs, concomitant medication, laboratory tests, etc. A fasting lipid panel is collected at week 6 to assess the nadir LDL-C level in response to 2 IB 12 treatment. 2 IB 12 formulation is administered at day 1, week 4, and week 8. The end-of-study (EOS) visit and the last estimation of lipids occurs at week 12 for all subjects.
  • EOS end-of-study

Abstract

La présente invention concerne des procédés de traitement d'une hypercholestérolémie familiale homozygote au moyen d'anticorps dirigés contre la proprotéine convertase subtilisine/kexine de type 9 (PCSK9).
EP13737090.4A 2013-06-28 2013-06-28 Procédés de traitement d'une hypercholestérolémie familiale homozygote Pending EP3013422A1 (fr)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2013/048714 WO2014209384A1 (fr) 2013-06-28 2013-06-28 Procédés de traitement d'une hypercholestérolémie familiale homozygote

Publications (1)

Publication Number Publication Date
EP3013422A1 true EP3013422A1 (fr) 2016-05-04

Family

ID=48790655

Family Applications (1)

Application Number Title Priority Date Filing Date
EP13737090.4A Pending EP3013422A1 (fr) 2013-06-28 2013-06-28 Procédés de traitement d'une hypercholestérolémie familiale homozygote

Country Status (5)

Country Link
EP (1) EP3013422A1 (fr)
JP (1) JP6267792B2 (fr)
AU (2) AU2013396206B2 (fr)
CA (1) CA2916259C (fr)
WO (1) WO2014209384A1 (fr)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG174053A1 (en) 2006-09-01 2011-09-29 Therapeutic Human Polyclonals Inc Enhanced expression of human or humanized immunoglobulin in non-human transgenic animals
CA2954767A1 (fr) * 2014-07-14 2016-01-21 Amgen Inc. Formulations d'anticorps cristallins
CN106589127A (zh) * 2015-10-16 2017-04-26 钜川生物医药 一种pcsk9抗体及其制备方法和应用
CN108347906A (zh) * 2015-10-29 2018-07-31 豪夫迈·罗氏有限公司 具有共同轻链的转基因兔
WO2017118307A1 (fr) * 2016-01-05 2017-07-13 江苏恒瑞医药股份有限公司 Anticorps anti-pcsk9, fragment de liaison à l'antigène associé et application médicale associée
EP3609532A1 (fr) 2017-04-13 2020-02-19 Cadila Healthcare Limited Vaccin pcsk9 à base de nouveaux peptides
JP6639463B2 (ja) * 2017-12-21 2020-02-05 アムジエン・インコーポレーテツド ホモ接合性家族性高コレステロール血症の治療方法
EA202191892A1 (ru) 2019-01-18 2022-02-24 Астразенека Аб Ингибиторы pcsk9 и способы их применения
KR20210095781A (ko) 2020-01-24 2021-08-03 주식회사 에이프릴바이오 항원결합 단편 및 생리활성 이펙터 모이어티로 구성된 융합 컨스트럭트를 포함하는 다중결합항체 및 이를 포함하는 약학조성물

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3180193A (en) 1963-02-25 1965-04-27 Benedict David Machines for cutting lengths of strip material
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4263428A (en) 1978-03-24 1981-04-21 The Regents Of The University Of California Bis-anthracycline nucleic acid function inhibitors and improved method for administering the same
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
EP0088046B1 (fr) 1982-02-17 1987-12-09 Ciba-Geigy Ag Lipides en phase aqueuse
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
DE3474511D1 (en) 1983-11-01 1988-11-17 Terumo Corp Pharmaceutical composition containing urokinase
US4740461A (en) 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
DE3675588D1 (de) 1985-06-19 1990-12-20 Ajinomoto Kk Haemoglobin, das an ein poly(alkenylenoxid) gebunden ist.
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4959455A (en) 1986-07-14 1990-09-25 Genetics Institute, Inc. Primate hematopoietic growth factors IL-3 and pharmaceutical compositions
DE3785186T2 (de) 1986-09-02 1993-07-15 Enzon Lab Inc Bindungsmolekuele mit einzelpolypeptidkette.
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4912040A (en) 1986-11-14 1990-03-27 Genetics Institute, Inc. Eucaryotic expression system
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
EP1690935A3 (fr) 1990-01-12 2008-07-30 Abgenix, Inc. Génération d'anticorps xenogéniques
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
FR2664073A1 (fr) 1990-06-29 1992-01-03 Thomson Csf Moyens de marquage d'objets, procede de realisation et dispositif de lecture.
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
CA2090473A1 (fr) 1990-08-29 1992-03-01 Robert M. Kay Recombinaison homologue dans des cellules mammaliennes
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 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
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
KR100272077B1 (ko) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 이종 항체를 생산할 수 있는 전이유전자를 가진 인간이외의 동물
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
WO1993012227A1 (fr) 1991-12-17 1993-06-24 Genpharm International, Inc. Animaux transgeniques non humains capables de produire des anticorps heterologues
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
WO1992022670A1 (fr) 1991-06-12 1992-12-23 Genpharm International, Inc. Detection precoce d'embryons transgeniques
WO1992022645A1 (fr) 1991-06-14 1992-12-23 Genpharm International, Inc. Animaux transgeniques non humains presentant une deficience immunitaire
WO1994004679A1 (fr) 1991-06-14 1994-03-03 Genentech, Inc. Procede pour fabriquer des anticorps humanises
WO1993004169A1 (fr) 1991-08-20 1993-03-04 Genpharm International, Inc. Ciblage de genes dans des cellules animales au moyen de produits de synthese d'adn isogeniques
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
PT1696031E (pt) 1991-12-02 2010-06-25 Medical Res Council Produção de auto-anticorpos a partir de reportórios de segmentos de anticorpo e exibidos em fagos
ES2202310T3 (es) 1991-12-13 2004-04-01 Xoma Corporation Metodos y materiales para la preparacion de dominios variables de anticuerpos modificados y sus usos terapeuticos.
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
JPH07508410A (ja) 1992-06-18 1995-09-21 ジェンファーム インターナショナル インコーポレイテッド 酵母人工染色体を有するトランスジェニック非ヒト動物の製造方法
JPH07509137A (ja) 1992-07-24 1995-10-12 セル ジェネシス,インク. 異種抗体の生産
US6066718A (en) 1992-09-25 2000-05-23 Novartis Corporation Reshaped monoclonal antibodies against an immunoglobulin isotype
US5981175A (en) 1993-01-07 1999-11-09 Genpharm Internation, Inc. Methods for producing recombinant mammalian cells harboring a yeast artificial chromosome
CA2161351C (fr) 1993-04-26 2010-12-21 Nils Lonberg Animaux transgeniques, pouvant produire des anticorps heterologues
US5625825A (en) 1993-10-21 1997-04-29 Lsi Logic Corporation Random number generating apparatus for an interface unit of a carrier sense with multiple access and collision detect (CSMA/CD) ethernet data network
US5643763A (en) 1994-11-04 1997-07-01 Genpharm International, Inc. Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
CA2219486A1 (fr) 1995-04-28 1996-10-31 Abgenix, Inc. Anticorps humains derives de xeno-souris immunisees
US6127977A (en) 1996-11-08 2000-10-03 Cohen; Nathan Microstrip patch antenna with fractal structure
AU718138B2 (en) 1995-08-29 2000-04-06 Kyowa Hakko Kirin Co., Ltd. Chimeric animal and method for constructing the same
EP2314625B1 (fr) 1996-12-03 2014-05-07 Amgen Fremont Inc. Mammifères transgèniques obtenus par génie génétique contenant des loci des immunoglobulines humaines qui comprennent plusieurs régions de VH et de Vkappa, et anticorps aussi obtenus
US6133426A (en) 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
US6833268B1 (en) 1999-06-10 2004-12-21 Abgenix, Inc. Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
WO2001083525A2 (fr) 2000-05-03 2001-11-08 Amgen Inc. Peptides modifies utilises comme agents therapeutiques
US8680534B2 (en) * 2005-01-11 2014-03-25 Semileds Corporation Vertical light emitting diodes (LED) having metal substrate and spin coated phosphor layer for producing white light
WO2008006332A1 (fr) 2006-07-13 2008-01-17 Luk Lamellen Und Kupplungsbau Beteiligungs Kg Chaîne articulée, en particulier pour un entraînement de véhicule
CN101636179B (zh) * 2006-11-07 2012-10-10 默沙东公司 Pcsk9拮抗剂
US20100233177A1 (en) * 2007-04-13 2010-09-16 David Langdon Yowe Molecules and methods for modulating proprotein convertase subtilisin/kexin type 9 (pcsk9)
JOP20080381B1 (ar) * 2007-08-23 2023-03-28 Amgen Inc بروتينات مرتبطة بمولدات مضادات تتفاعل مع بروبروتين كونفيرتاز سيتيليزين ككسين من النوع 9 (pcsk9)
JP2011501952A (ja) 2007-10-26 2011-01-20 シェーリング コーポレイション 脂質障害およびコレステロール障害を治療するための抗pcsk9および方法
AR070315A1 (es) 2008-02-07 2010-03-31 Merck & Co Inc Anticuerpos 1b20 antagonistas de pcsk9
AR070316A1 (es) 2008-02-07 2010-03-31 Merck & Co Inc Antagonistas de pcsk9 (proproteina subtilisina-kexina tipo 9)
TWI516501B (zh) 2008-09-12 2016-01-11 禮納特神經系統科學公司 Pcsk9拮抗劑類
JO3672B1 (ar) 2008-12-15 2020-08-27 Regeneron Pharma أجسام مضادة بشرية عالية التفاعل الكيماوي بالنسبة لإنزيم سبتيليسين كنفرتيز بروبروتين / كيكسين نوع 9 (pcsk9).
CA2777698A1 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes de la pcsk9 avec anticorps fab ax189 et ax1, et variantes afferentes
WO2012054438A1 (fr) 2010-10-22 2012-04-26 Schering Corporation Anti-pcsk9
US20120195910A1 (en) * 2010-12-22 2012-08-02 Genentech, Inc. Anti-pcsk9 antibodies and methods of use
DK2668212T3 (en) * 2011-01-28 2018-07-02 Saonofi Biotechnology HUMAN ANTIBODIES AGAINST PCSK9 FOR USE IN PROCEDURES FOR TREATING PARTICULAR GROUPS OF PERSONS
BR112013020402A2 (pt) * 2011-02-11 2018-09-25 Irm Llc antagonistas pcsk9
JOP20200043A1 (ar) * 2011-05-10 2017-06-16 Amgen Inc طرق معالجة أو منع الاضطرابات المختصة بالكوليسترول
AR087715A1 (es) * 2011-09-16 2014-04-09 Lilly Co Eli Anticuerpos anti pcsk9 y usos de los mismos
US9401875B2 (en) 2012-06-01 2016-07-26 Nippon Telegraph And Telephone Corporation Packet transfer processing method and packet transfer processing device
JP6071725B2 (ja) 2013-04-23 2017-02-01 カルソニックカンセイ株式会社 電気自動車の駆動力制御装置
US9300829B2 (en) 2014-04-04 2016-03-29 Canon Kabushiki Kaisha Image reading apparatus and correction method thereof
US11284893B2 (en) 2019-04-02 2022-03-29 Covidien Lp Stapling device with articulating tool assembly

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None *
See also references of WO2014209384A1 *

Also Published As

Publication number Publication date
AU2020201012C1 (en) 2022-06-30
WO2014209384A1 (fr) 2014-12-31
JP6267792B2 (ja) 2018-01-24
JP2016528193A (ja) 2016-09-15
CA2916259C (fr) 2024-02-20
AU2013396206B2 (en) 2019-11-14
AU2020201012B2 (en) 2021-12-23
AU2013396206A1 (en) 2016-01-21
CA2916259A1 (fr) 2014-12-31
AU2020201012A1 (en) 2020-03-05

Similar Documents

Publication Publication Date Title
US20210047434A1 (en) Methods of treating or preventing cholesterol related disorders
EP2844285B1 (fr) Formulations stables contenant des anticorps anti-pcsk9
US20150004174A1 (en) Methods for treating homozygous familial hypercholesterolemia
AU2020201012C1 (en) Methods for treating homozygous familial hypercholesterolemia
US20140004122A1 (en) Methods for treating or preventing cholesterol related disorders
EP2615114B1 (fr) Protéines de liaison à un antigène pour proprotéine convertase subtilisine kexine de type 9 (PCSK9)
AU2018247211A1 (en) Antigen Binding Proteins to Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9)
JP6639463B2 (ja) ホモ接合性家族性高コレステロール血症の治療方法
NZ734570B2 (en) Methods of treating or preventing cholesterol related disorders
NZ618300B2 (en) Methods of treating or preventing cholesterol related disorders

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20160128

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20190319

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS