WO2016071701A1 - Traitement de maladie par liaison de ligand à des cibles d'intérêt - Google Patents

Traitement de maladie par liaison de ligand à des cibles d'intérêt Download PDF

Info

Publication number
WO2016071701A1
WO2016071701A1 PCT/GB2015/053361 GB2015053361W WO2016071701A1 WO 2016071701 A1 WO2016071701 A1 WO 2016071701A1 GB 2015053361 W GB2015053361 W GB 2015053361W WO 2016071701 A1 WO2016071701 A1 WO 2016071701A1
Authority
WO
WIPO (PCT)
Prior art keywords
human
toi
ligand
nucleotide sequence
amino acid
Prior art date
Application number
PCT/GB2015/053361
Other languages
English (en)
Inventor
Jasper Clube
Original Assignee
Kymab Limited
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
Priority claimed from US14/536,129 external-priority patent/US9062105B1/en
Priority claimed from US14/536,049 external-priority patent/US9045545B1/en
Priority claimed from US14/537,403 external-priority patent/US9067998B1/en
Priority claimed from PCT/GB2014/053729 external-priority patent/WO2015092393A2/fr
Priority claimed from DE202014010421.2U external-priority patent/DE202014010421U1/de
Priority claimed from US14/665,579 external-priority patent/US9139648B1/en
Priority claimed from US14/665,532 external-priority patent/US9150660B1/en
Priority claimed from PCT/EP2015/068491 external-priority patent/WO2016023916A1/fr
Application filed by Kymab Limited filed Critical Kymab Limited
Publication of WO2016071701A1 publication Critical patent/WO2016071701A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1

Definitions

  • the technology described herein relates to ligands, e.g., antibodies for the treatment of disease.
  • the 1000 Genomes Project has the objective of cataloguing sequences in the human genome, involving sequencing the genomes of a very large sampling of individuals from diverse art -recognized human ethnic populations.
  • the present invention provides for improved human patient diagnosis and therapy. Importantly, the invention enables tailored medicines that address individual human patient genotypes or phenotypes.
  • TOI genomic human target of interest
  • the inventor realised that there is significant industrial and medical application for the invention in terms of guiding the choice of an anti-TOI ligand for administration to human patients for therapy and/or prophylaxis of TOI-mediated or associated diseases and conditions.
  • the patient receives drugs and ligands that are tailored to their needs - as determined by the patient's genetic or phenotypic makeup.
  • the invention provides for the genotyping and/or phenotyping of patients in connection with such treatment, thereby allowing a proper match of drug to patient.
  • the TOI is an iron homeostasis target, VEGF, PD-Ll, PD-1, PDGF-B, PDGFR-B, Navl.8 or Nav 1.9.
  • iron homeostasis targets include BMP6, hemojuvelin, TMPRSS6, ferroportin, hepcidin, HFE, transferrin and CUBN.
  • the invention provides :- [0008] In a First Configuration
  • step (a) said human has been or is genotyped as positive for said nucleotide sequence or phenotyped as positive for said TOI variant.
  • step (a) the ligand has been or is determined as capable of binding to said TOI variant.
  • step (a) said human has been or is genotyped as negative for a variant nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%; or phenotyped as negative for a TOI variant encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • An anti-human TOI ligand for use in a method of treating and/or preventing a TOI- mediated disease or condition in a human, wherein the TOI is present in humans as different polymorphic variants and wherein the genome of said human comprises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, the method comprising administering the ligand to the human.
  • a ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, for use in a method comprising the step of using the ligand to target said TOI in a human to treat and/or prevent a disease or condition mediated by TOI, the method comprising administering the ligand to the human.
  • a pharmaceutical composition or kit for treating or preventing a condition or disease mediated by a TOI is provided.
  • a method of producing an anti-human TOI antibody binding site comprising obtaining a plurality of anti-TOI antibody binding sites, screening the antibody binding sites for binding to a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody binding site that binds in the screening step.
  • a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest
  • a method of producing an anti-human TOI antibody comprising immunising a non-human vertebrate (eg, a mouse or a rat) with a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody that binds a TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, and optionally producing a TOI-binding fragment or derivative of the isolated antibody.
  • a non-human vertebrate eg, a mouse or a rat
  • TOI comprising an amino acid sequence encoded
  • kits for TOI genotyping a human comprising a nucleic acid comprising a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the nucleic acid comprises a nucleotide sequence that comprises at least 10 contiguous nucleotides of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • an anti-TOI ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for treating and/or preventing a TOI-mediated disease or condition in a human whose genome comprises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • an anti-TOI ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for targeting said TOI in a human to treat and/or prevent a disease or condition mediated by TOI.
  • a method of targeting a TOI for treating and/or preventing a TOI-mediated disease or condition in a human comprising administering an anti-TOI ligand to a human comprising a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, whereby a TOI encoded by said nucleotide sequence is targeted.
  • a method of TOI genotyping a nucleic acid sample of a human comprising identifying in the sample the presence of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • a method of TOI typing a protein sample of a human comprising identifying in the sample the presence of a TOI amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the TOI is a human TOI selected from the group consisting of PCSK9, VEGF-A and IL6 receptor.
  • the TOI is human IL4Ra, PDGF-B, PDGFR-B or Ang-2.
  • a Fifteenth Configuration provides a ligand, method, use, kit or composition of the invention, wherein
  • the ligand eg, antibody or fragment
  • variable domain that is encoded by a human V region nucleotide sequence, wherein the V nucleotide sequence is derived from recombination of human VH, D and JH gene segments or human VL and JL gene segments; or
  • a first gene segment of said gene segments of (a), or said C region gene segment of (b) comprises a first single nucleotide polymorphism (SNP) encoding a first amino acid polymorphism
  • the genome of said human comprises said first SNP or wherein said human expresses (a') an antibody variable domain comprising said first amino acid polymorphism or (b') an antibody constant domain comprising said first amino acid polymorphism.
  • a Sixteenth Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand comprises or consists of an antibody or fragment that comprises a human antibody variable domain derived from the recombination of a human V gene segment and a human J gene segment (and optionally a human D gene segment when the variable domains are VH domains); and wherein the genome of the human comprises said human V gene segment and/or the human expresses antibodies comprising antibody variable domains derived from the recombination of said human V gene segment and a human J gene segment (and optionally a human D gene segment).
  • a Sixteenth Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc-fused human PCSK9 receptor) comprises a human heavy chain constant domain encoded by a first constant region nucleotide sequence; and wherein the genome of the human comprises a heavy chain constant region nucleotide sequence that is identical to said first constant region nucleotide sequence and/or the human expresses antibodies comprising said human constant domain.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc-fused human PCSK9 receptor
  • the genome of the human comprises a heavy chain constant region nucleotide sequence that is identical to said first constant region nucleotide sequence and/or the human expresses antibodies comprising said human constant domain.
  • a Seventeenth Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc-fused TOI receptor) comprises a human gamma- 1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 42 or a Leu corresponding to position 206 of SEQ ID NO: 42 and wherein the genome of the human comprises a gamma- 1 heavy chain constant region nucleotide sequence that encodes such an Asp or Leu or the human expresses antibodies comprising human gamma- 1 constant regions comprising such an Asp or Leu.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc-fused TOI receptor
  • the genome of the human comprises a gamma- 1 heavy chain constant region nucleotide sequence that encodes such an Asp or Leu or the human expresses antibodies comprising human gamma- 1 constant regions compris
  • a Eighteenth Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc-fused TOI receptor) comprises a human gamma-2 heavy chain constant region that comprises an amino acid selected from the group consisting of a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44, a Phe corresponding to position 76 of SEQ ID NO: 44, a Val corresponding to position 161 of SEQ ID NO: 44 and an Ala corresponding to position 257 of SEQ ID NO: 44; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such a selected amino acid or the human expresses antibodies comprising human gamma-2 constant regions comprising such a selected amino acid.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc-fused TOI
  • a Ninteenth Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc-fused TOI receptor) comprises a human kappa light chain constant region that comprises a Val
  • the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val or Cys or the human expresses antibodies comprising human kappa light chain constant regions comprising such a Val or Cys.
  • a Twentieth Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand comprises or consists of an antibody or fragment, wherein the antibody or fragment comprises a VH domain that is derived from the recombination of a human VH gene segment, a human D gene segment and a human JH gene segment, wherein the VH gene segment is selected from the group consisting of (i) IGHV1- 18*01 and the genome of the human comprises a human IGHV1- 18*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV1-18*01 ; or (ii) IGVH1-46*01 and the genome of the human comprises a human IGHV1-46*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV1-46*01.
  • a Twenty-First Configuration provides the ligand, method, use, kit or composition of the invention, wherein the ligand comprises or consists of an antibody or fragment, wherein the antibody or fragment comprises a VL domain that is derived from the recombination of a human VL gene segment and a human JL gene segment, wherein the VL gene segment is selected from the group consisting of (i) IGKV4-1*01 and the genome of the human comprises a human IGKV4-1*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV4-1*01 ; (ii) IGLV2- 14*01 and the genome of the human comprises a human IGLV2- 14*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGLV2- 14*01 ; or (iii) IGKV1 -13*02 and the genome of the human comprises
  • a Twenty-Second Configuration provides a method of treating or reducing the risk of an IL4Ra-mediated disease or condition in a human in need thereof, the method comprising administering to said human a ligand (eg, an antibody or antibody fragment) that specifically binds a human IL4RA protein that comprises a mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • a ligand eg, an antibody or antibody fragment
  • Said human comprises a nucleotide sequence encoding said IL4RA protein comprising said mutation selected from the group consisting of 175 V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • a Twenty-Third Configuration provides a ligand (eg, an antibody or antibody fragment) for treating or reducing the risk of an IL4Ra-mediated disease or condition in a human in need thereof, the method comprising administering to said human said ligand, wherein the ligand specifically binds a human IL4RA protein that comprises a mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • a ligand eg, an antibody or antibody fragment
  • Said human comprises a nucleotide sequence encoding said IL4RA protein comprising said mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • a Twenty-Fourth Configuration provides a method of targeting IL4Ra in a human, the method comprising administering to said human a ligand (eg, an antibody or antibody fragment) that specifically binds a human IL4RA protein that comprises a mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • Said human comprises a nucleotide sequence encoding said IL4RA protein comprising said mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • a Twenty-Fifth Configuration provides a ligand (eg, an antibody or antibody fragment) for targeting IL4Ra in a human, the method comprising administering to said human said ligand, wherein the ligand specifically binds a human IL4RA protein that comprises a mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • Said human comprises a nucleotide sequence encoding said IL4RA protein comprising said mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • the human is suffering from or at riskof an IL4Ra-mediated disease or condition.
  • the method treats or reduces the risk of an IL4Ra-mediated disease or condition in the human.
  • the antibody or fragment comprises a VH domain derived from the recombination of a human VH segment, a human D gene segment and a human JH segment, the human VH segment encoding the framework 1 of SEQ ID NO: 40 and wherein said human comprises a VH gene segment encoding the framework 1 of SEQ ID NO: 40 , or the human expresses VH domains that comprise the framework 1 of SEQ ID NO: 40 ; and wherein (ii) said human comprises a nucleotide sequence encoding said IL4RA protein comprising said mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • the antibody or fragment comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises an IGHG4*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-4 heavy chain constant regions comprising a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73; and wherein (ii) said human comprises a nucleotide sequence encoding said IL4RA protein comprising said mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • a ligand eg, an antibody or antibody fragment
  • a huma Navl.7 protein that comprises an amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I),
  • a ligand eg, an antibody or antibody fragment
  • said amino acid is selected from the group consisting of any one of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P and 1449V; optionally wherein said disease or condition is a pain disease or condition.
  • said amino acid is selected from the group consisting of 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641 and 1627K; optionally wherein said disease or condition is a pain disease or condition.
  • said amino acid is selected from the group consisting of 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein X is an amino acid other than R), 1659X (wherein X is an amino acid other than K) and 1689X (wherein X is an amino acid other than W); optionally wherein said disease or condition is a pain disease or condition.
  • the constant region gene segment comprised by said human is a germline gene segment.
  • the method further comprises, before said administering, selecting a human comprising said nucleotide sequence of (ii).
  • the human has been determined to comprise the nucleotide sequence that encodes a Navl.7 protein comprising said amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X
  • X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein X is an amino acid other than R), 1659X (wherein X is an amino acid other than K), 1689X (wherein X is an amino acid other than W), 422D, 490N, 943L, 1002L, 1161W and 1919G and/or a Navl.7 protein comprising said amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T,
  • the method further comprises the step of determining that the human comprises (a) the nucleotide sequence that encodes a Navl.7 protein comprising said amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein
  • X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein X is an amino acid other than R), 1659X (wherein X is an amino acid other than K), 1689X (wherein X is an amino acid other than W), 422D, 490N, 943L, 1002L, 1161W and 1919G, optionally, wherein the determining step is performed before administration of the antibody to the human.
  • the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a Navl.7 protein comprising said amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein X is
  • the assaying comprises contacting the biological sample with at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding a Navl.7 protein comprising said amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other
  • the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid
  • said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human.
  • said human is indicated as heterozygous for a nucleotide sequence encoding the Navl.7 protein comprising said amino acid selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein X is an amino acid other
  • said human is or has been further determined to be substantially resistant to a pain or itching treatment. In some embodiments, said human is receiving or has received a pain or anti-itching treatment or has reduced responsiveness to a pain or itching treatment.
  • said disease or condition is a pain or itching disease or condition.
  • said disease or condition is a channelopathy or associated with a channelopathy; or is selected from the group consisting of primary erythermalgia (PE), paroxysmal extreme pain disorder (PEPD) and channelopathy-associated insensitivity to pain (CIP).
  • PE primary erythermalgia
  • PEPD paroxysmal extreme pain disorder
  • CIP channelopathy-associated insensitivity to pain
  • said human has been diagnosed with a pain or itching disease or condition.
  • said ligand fragment treats or reduces the risk in said human of a pain or itching disease or condition.
  • the nucleotide sequence comprises one or more SNPs selected from the group consisting of rs6746030, rs3750904, rs58022607, rs4369876, rsl3402180 and rsl2478318.
  • a method of treating or reducing the risk of a disease or condition mediated by VEGF-A in a human comprising administering to said human an anti -VEGF-A ligand (eg, an anti- VEGF-A trap, antibody or antibody fragment) that specifically binds to a human VEGF-A that is expressed by a VEGF-A nucleotide sequence comprising a SNP selected from the group consisting of rs699947, rs833061, rs2010963, rs3025039, rs699946, rs2146323, rsl413711, rs833068, rs833069, rs3025000 and rsl570360, wherein said human comprises a VEGF-A nucleotide sequence comprising said selected SNP.
  • the invention also provides a corresponding ligand, eg, antibody or antibody fragment, for
  • a method of treating or reducing the risk of a disease or condition mediated by VEGF-A in a human comprising administering to said human an anti -VEGF-A ligand (eg, an anti- VEGF-A trap, antibody or antibody fragment) that specifically binds to a human VEGF-A
  • the human comprises (i) an ARMS2 nucleotide sequence comprising a G at the position of SNP rs 10490924; (ii) a CFH nucleotide sequence comprising a T at the position of SNP rsl061170 or an T at the position of rs3766404; or (iii) a VEGFR2 nucleotide sequence comprising SNP rs4576072 or rs6828477, wherein the ligand comprises a human gamma- 1 heavy chain constant region that comprises an amino acid selected from the group consisting
  • a method of treating or reducing the risk of a VEGF-A mediated disease or condition in a human comprising administering an anti-human VEGF-A ligand to a human, wherein the human comprises (i) a PDGF-B nucleotide sequence comprising a SNP selected from the group consisting of rsl42404523 (ie, a C corresponding to position -776) and a C at the position of rsl800818 (ie, a C corresponding to position -735); or (ii) a PDGFR-B nucleotide sequence comprising a SNP selected from the group consisting of rs246395 (ie, a G corresponding to position 2601) and rs74943037 (ie, a T corresponding to position 1391).
  • the invention also provides a corresponding ligand, eg, antibody or antibody fragment, for use in such
  • the ligand comprises an antibody constant region (eg, an antibody Fc region).
  • the ligand comprises a human gamma- 1 heavy chain constant region that comprises an amino acid selected from the group consisting of an Asp corresponding to position 204 of SEQ ID NO: 42 and a Leu corresponding to position 206 of SEQ ID NO: 42 and wherein said human comprises an IGHG1*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma- 1 heavy chain constant regions comprising said selected amino acid.
  • the disease or condition is an ocular condition or a cancer; or angiogenesis or neovascularisation.
  • the method further comprises antagonising PDGF-B in said human.
  • the method further comprises antagonising angiopoietin-2 (Ang2) in said human.
  • Ang2 angiopoietin-2
  • the invention provides: -
  • a method of treating or reducing the risk of a disease or condition mediated by PD-Ll in a human comprising administering to said human an anti-PD-Ll ligand (eg, an anti-PD- Ll trap, antibody or antibody fragment) that specifically binds to a human PD-Ll that is expressed by a PD-Ll nucleotide sequence comprising a variation selected from the variations listed in Table 21.
  • an anti-PD-Ll ligand eg, an anti-PD- Ll trap, antibody or antibody fragment
  • a method of treating or reducing the risk of a disease or condition mediated by PD-Ll in a human comprising administering to said human an anti-PD-Ll ligand (eg, an anti-PD-Ll trap, antibody or antibody fragment) that specifically binds to a human PD-Ll that is expressed by a PD-Ll nucleotide sequence comprising a variation selected from the variations listed in Table 21, wherein said human comprises a PD-Ll nucleotide sequence comprising said selected variation.
  • an anti-PD-Ll ligand eg, an anti-PD-Ll trap, antibody or antibody fragment
  • a method of treating or reducing the risk of a cancer in a human comprising administering to said human an anti-PD-Ll ligand (eg, an anti-PD-Ll trap, antibody or antibody fragment) that specifically binds to a human PD-Ll that is expressed by a PD-Ll nucleotide sequence comprising a variation selected from the variations listed in Table 21, wherein said human comprises a PD-Ll nucleotide sequence comprising said selected variation.
  • an anti-PD-Ll ligand eg, an anti-PD-Ll trap, antibody or antibody fragment
  • a method of treating or reducing the risk of an autoimmune disease or condition in a human comprising administering to said human an anti-PD-Ll ligand (eg, an anti-PD-Ll trap, antibody or antibody fragment) that specifically binds to a human PD-Ll that is expressed by a PD-Ll nucleotide sequence comprising a variation selected from the variations listed in Table 21, wherein said human comprises a PD-Ll nucleotide sequence comprising said selected variation.
  • an anti-PD-Ll ligand eg, an anti-PD-Ll trap, antibody or antibody fragment
  • a method of treating or reducing the risk inflamatory disease or condition in a human comprising administering to said human an anti-PD-Ll ligand (eg, an anti-PD-Ll trap, antibody or antibody fragment) that specifically binds to a human PD- Ll that is expressed by a PD-Ll nucleotide sequence comprising a variation selected from the variations listed in Table 21, wherein said human comprises a PD-Ll nucleotide sequence comprising said selected variation.
  • an anti-human PD-L1 ligand eg, an antibody, antibody fragment or human PD-L1 trap
  • the invention provides: A method of cancer immunotherapy in a human by targeting an immune cell TOI (eg, PD-1) in the human, the TOI being present in humans as a plurality of variants differing by one or more amino acid polymorphisms, the method comprising administering a ligand (eg, an antibody or antibody fragment) to the human, the ligand comprising first and second protein domains, wherein the first domain specifically binds a TOI variant comprising a first amino acid polymorphism, wherein the second domain comprises a second polymorphism, and wherein the human expresses (i) TOI comprising said first amino acid polymorphism; and (ii) protein domains comprising said second polymorphism, wherein the ligand comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises
  • This configuration also provides the following aspects :-
  • a method of cancer immunotherapy in a human by targeting PD-1 in the human comprising administering an antibody or antibody fragment to the human, wherein the antibody or antibody fragment specifically binds a PD-1 encoded by a PD-1 nucleotide sequence comprising a SNP selected from the group consisting of the variations set out in Table 23, wherein the antibody or antibody fragment comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises (i) an IGHG4*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-4 heavy chain constant regions comprising a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73; and (ii) a PD-1 nucleotide sequence comprising said selected SNP.
  • a method of cancer immunotherapy in a human by targeting PD-1 in the human comprising administering an antibody or antibody fragment to the human, wherein the antibody or antibody fragment specifically binds a PD-1 encoded by a PD-1 nucleotide sequence comprising a SNP (first polymorphism) selected from the group consisting of the variations set out in Table 23 (eg, selected from the group consisting of rs36084323, rsl0204225, rsl l568821, rs2227981 and rs2227982), wherein the antibody or antibody fragment comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises (i) an IGHG4*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-4 heavy chain constant regions comprising a Leu at position 189 shown
  • a method of treating cancer in a human comprising administering an antibody or antibody fragment to the human, wherein the antibody or antibody fragment inhibits the binding of PD-L1 or PD-L2 to a PD-1 comprising a first amino acid polymorphism, wherein the antibody or antibody fragment comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises (i) an IGHG4*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-4 heavy chain constant regions comprising a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73; and (ii) a PD-1 nucleotide sequence encoding a PD-1 comprising said first amino acid polymorphism or the human expresses a PD-1 comprising said first amino acid polymorphism
  • the invention instead of “A method of cancer immunotherapy” or “A method of treating cancer”, the invention instead provides "A method of treating or reducing the risk of an autoimmune disease or condition” or “A method of treating or reducing the risk inflammatory disease or condition” and the TOI polymorphism is associated with an autoimmune or inflammatory disease or condition.
  • the invention also provides a ligand (eg, a trap, an antibody or antibody fragment) for use in any of the methods.
  • a ligand eg, a trap, an antibody or antibody fragment
  • the invention provides: A method of treating or reducing the risk of a disease or condition (eg, anaemia) in a human, wherein the disease or condition is mediated by a TOI, the TOI being present in humans as a plurality of variants differing by one or more amino acid polymorphisms, the method comprising administering a ligand (eg, an antibody or antibody fragment) to the human, the ligand comprising first and second protein domains, wherein the first domain specifically binds a TOI variant comprising a first amino acid polymorphism, wherein the second domain comprises a second polymorphism, and wherein the human expresses (i) TOI comprising said first amino acid polymorphism; and (ii) protein domains comprising said second polymorphism, optionally wherein the ligand comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 2
  • a ligand eg, an
  • Examples of any configuration or relating to any TOI are as follows :- (i) wherein the ligand comprises a VH domain derived from the recombination of a human VH segment (eg, human VH3 -23*04), a human D gene segment and a human JH segment, the human VH segment encoding the framework 1 of SEQ ID NO: 40 and wherein said human comprises a VH gene segment encoding the framework 1 of SEQ ID NO: 40, or the human expresses VH domains that comprise the framework 1 of SEQ ID NO: 40.
  • a human VH segment eg, human VH3 -23*04
  • a human D gene segment e.g, a human D gene segment and a human JH segment
  • the human VH segment encoding the framework 1 of SEQ ID NO: 40 e.g, human VH3 -23*04
  • said human comprises a VH gene segment encoding the framework 1 of SEQ ID NO: 40
  • the human expresse
  • the ligand comprises a VH domain derived from the recombination of human VH segment IGHV3-7*01, a human D gene segment and a human JH segment, and wherein said human comprises a IGHV3-7*01 VH gene segment or the human expresses VH domains derived from the recombination of human VH segment IGHV3-7*01, a human D gene segment and a human JH segment.
  • the ligand comprises a VK domain derived from the recombination of human VK segment IGKV1-12*01 and a human JK segment
  • said human comprises a IGKV1-12*01 Vtc gene segment or the human expresses VK domains derived from the recombination of human VK segment IGKV1-12*01 and a human JK segment.
  • the ligand comprises a VK domain derived from the recombination of a human VK segment and a human JK segment, the human VK segment encoding (i) a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 36 and wherein said human comprises a VK gene segment encoding a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 36, or the human expresses VK domains that comprise a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 36; or (ii) a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 38 and wherein said human comprises a VK gene segment encoding a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 38 or the human expresses VK domains that comprise a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 38.
  • the ligand comprises a human gamma- 1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 4 or a Leu at position 206 shown in SEQ ID NO: 4 and wherein said human comprises (i) an IGHG1*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma- 1 heavy chain constant regions comprising an Asp at position 204 shown in SEQ ID NO: 4 or a Leu at position 206 shown in SEQ ID NO: 4.
  • the ligand comprises a human gamma-2 heavy chain constant region that comprises an amino acid selected from the group consisting of a Pro at position 72 shown in SEQ ID NO: 6, an Asn at position 75 shown in SEQ ID NO: 6, a Phe at position 76 shown in SEQ ID NO: 6, a Val at position 161 shown in SEQ ID NO: 6 and an Ala at position 257 shown in SEQ ID NO: 6 and wherein said human comprises (i) an IGHG2*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-2 heavy chain constant regions comprising said selected Pro at position 72 shown in SEQ ID NO: 6, Asn at position 75 shown in SEQ ID NO: 6, Phe at position 76 shown in SEQ ID NO: 6, Val at position 161 shown in SEQ ID NO: 6 or Ala at position 257 shown in SEQ ID NO: 6.
  • the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 16 or a Cys at position 87 shown in SEQ ID NO: 16 and wherein said human comprises (i) an IGKC1*01 human kappa chain constant region gene segment, or the human expresses antibodies comprising human kappa chain constant regions comprising a Val corresponding to position 84 shown in SEQ ID NO: 16 or a Cys at position 87 shown in SEQ ID NO: 16.
  • the ligand comprises a human IGLC1*01 lambda chain constant region and wherein said human comprises (i) a human IGLC1*01 lambda chain constant region gene segment, or the human expresses antibodies comprising human IGLC1*01 lambda chain constant regions.
  • the ligand comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises (i) an IGHG4*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-4 heavy chain constant regions comprising a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73.
  • the ligand comprises a human gamma-3 heavy chain constant region encoded by a first human IGHG3 (eg, IGHG3*01) constant region gene segment and wherein said human comprises (i) said first constant region gene segment (eg, an IGHG3*01), or the human expresses antibodies comprising human gamma-3 heavy chain constant regions encoded by said first human IGHG3 (eg, IGHG3*01) constant region gene segment.
  • the ligand comprises a human epsilon heavy chain constant region encoded by a first human epsilon heavy chain constant region gene segment and wherein said human comprises (i) said first constant region gene segment, or the human expresses antibodies comprising human epsilon heavy chain constant regions encoded by said first constant region gene segment.
  • the ligand comprises a human mu heavy chain constant region encoded by a first human mu heavy chain constant region gene segment and wherein said human comprises (i) said first constant region gene segment, or the human expresses antibodies comprising human mu heavy chain constant regions encoded by said first constant region gene segment.
  • the ligand comprises a human alpha heavy chain constant region encoded by a first human alpha heavy chain constant region gene segment and wherein said human comprises (i) said first constant region gene segment, or the human expresses antibodies comprising human alpha heavy chain constant regions encoded by said first constant region gene segment.
  • the ligand comprises a human delta heavy chain constant region encoded by a first human delta heavy chain constant region gene segment and wherein said human comprises (i) said first constant region gene segment, or the human expresses antibodies comprising human delta heavy chain constant regions encoded by said first constant region gene segment.
  • the ligand comprises a human kappa light chain constant region encoded by a first human kappa light chain constant region gene segment and wherein said human comprises (i) said first constant region gene segment, or the human expresses antibodies comprising human kappa light chain constant regions encoded by said first constant region gene segment.
  • the ligand comprises a human lambda light chain constant region encoded by a first human lambda light chain constant region gene segment and wherein said human comprises (i) said first constant region gene segment, or the human expresses antibodies comprising human lambda light chain constant regions encoded by said first constant region gene segment.
  • said ligand eg, antibody or antibody fragment
  • said ligand is administered by inhaled, intravenous or subcutaneous administration and/or is comprised in an inhalable or injectable preparation.
  • the human gamma-4 heavy chain constant region of the ligand comprises the amino acid sequence of SEQ ID NO: 73 or an ADCC inactivated version thereof.
  • the human gamma-4 heavy chain constant region comprises 228P and 235E.
  • the TOI is selected from the group consisting of human TOIs: PCSK9, IL6R, IL4Ra, VEGF-A, Placental growth factor (PGF), PDGF-B, PDGFR-
  • TMPRSS6, transferrin, human hemochromatosis protein (HFE) and sclerostin TMPRSS6, transferrin, human hemochromatosis protein (HFE) and sclerostin.
  • the TOI is PCSK9.
  • the TOI is IL6R.
  • the TOI is IL4Ra.
  • the TOI is VEGF-A.
  • the TOI is Placental growth factor (PGF).
  • PPF Placental growth factor
  • the TOI is PDGF-B.
  • the TOI is PDGFR-B .
  • the TOI is Ang-2.
  • the TOI is Navl .7.
  • the TOI is Navl.8.
  • the TOI is Navl .9.
  • the TOI is PD- 1.
  • the TOI is PD-L1.
  • the TOI is ICOS.
  • the TOI is BMP6.
  • the TOI is hemojuvelin.
  • the TOI is ferroportin.
  • the TOI is TMPRSS6.
  • the TOI is transferrin.
  • the TOI is human hemochromatosis protein (HFE).
  • HFE hemochromatosis protein
  • the TOI is sclerostin.
  • a method of treating or reducing the risk of a Nav 1.8 -mediated disease or condition in a human in need thereof, the method comprising administering to said human a ligand (eg, an antibody or antibody fragment) that specifically binds a human Navl.8 protein that comprises an amino acid selected from the group consisting of L554P, A1304T, I1706V, A1073V, G1662S, L1092P, I962V, S509P, I206M and R90W or an amino acid encoded by a SNP selected from the group consisting of rs6801957, rs6795970, rsl0428132, rsl2632942, rs57326399, rs7630989, rs74717885 and rsl44270136;
  • a ligand eg, an antibody or antibody fragment
  • the ligand comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises (i) an IGHG4*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-4 heavy chain constant regions comprising a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73; and wherein (ii) said human comprises a nucleotide sequence encoding said Navl.8 protein comprising said selected amino acid.
  • the invention also provides a corresponding ligand, eg, antibody or antibody fragment, for use in such a method.
  • VH or VL domain of the ligand eg, antibody
  • a different constant region is matched according to the invention.
  • a method of treating or reducing the risk of a Navl.9-mediated disease or condition in a human in need thereof, the method comprising administering to said human a ligand (eg, an antibody or antibody fragment) that specifically binds a human Navl.8 protein that comprises an amino acid selected from the group consisting of I381T, K419N, A582T, A681D, A842P, L1158P, F1689L, L811P, R225C, A808G, V909I, R86G, T1609I and G481E; wherein (i) the ligand comprises a human gamma-4 heavy chain constant region that comprises a Leu at position 189 shown in SEQ ID NO: 73 or an Arg at position 289 shown in SEQ ID NO: 73 and wherein said human comprises (i) an IGHG4*01 human heavy chain
  • a VH or VL domain of the ligand (eg, antibody) and/or a different constant region is matched according to the invention.
  • Figure 1 shows in silico modeling of PCSK9 surface variant residues.
  • Figure 2 depicts the cumulative allele frequency distribution across the 1000 Genomes Project databse of human VH3-23 alleles comprising SNP rs56069819 (such alleles denonted “C” and the most frequent allele (which does not comprise this SNP) denoted "A").
  • the figure shows that VH3-23 alleles comprising SNP rs56069819 are present at a cumulative frequency of 11% across all human ethnic populations taken as a whole, whereas in certain specific human ethnic sub -populations (ASW, LWK, YRI, CEU and GBR) such alleles are present at an above-average cumulative frequency.
  • ASW, LWK, YRI, CEU and GBR certain specific human ethnic sub -populations
  • Indicated in the figure are those human PCSK9 variant forms (marked "Variants") that are found in the various sub -populations with above-average occurrence of human VH3-23 alleles comprising SNP rs56069819.
  • Figure 3 depicts frameworks and CDRs encoded by VH3 -23*04 as obtained from the IMGT database (available on the World Wide Web at www.IMGT.org).
  • Figure 3 discloses the nucleotide sequences as SEQ ID NOS 117, 117, 117, 119, 119, 120, 122, 124, 39 and 125, respectively, in order of appearance.
  • Figure 3 discloses the coded amino acid sequences as SEQ ID NOS 118, 118, 118, 118, 118, 118, 118, 121, 123, 123, 38 and 126, respectively, in order of appearance.
  • Figure 4 depicts sequences of VH3-23*04.
  • the portion of VH3-23*04 comprising the FW1 residue change of rs56069819 (SEQ ID NO: 38).
  • the portion of the nucleic acid sequence encoding rs56069819 is depicted (SEQ ID NO: 39).
  • the FWl encoded by VH3-23*04 is depicted (SEQ ID NO: 40).
  • the skilled person will know that SNPs or other changes that translate into amino acid variation can cause variability in conformation or activity of human targets to be addressed. This has spawned great interest in personalized medicine where genotyping and knowledge of protein and nucleotide variability is used to tailor medicines and diagnosis of patients more effectively.
  • the present invention provides for tailored pharmaceuticals and testing that specifically addresses rarer variant forms of a human target of interest (TOI).
  • TOI human target of interest
  • the present invention harnesses the power of human genetic variation analysis and rationally-designed sequence selection.
  • the technical applications of these approaches, as per the present invention contribute to better treatment, prophylaxis and diagnosis in humans and provides for patient benefit by providing choice and enabling personalized medicines and therapies. This provides advantages of better prescribing, less wastage of medications and improved chances of drug efficacy and better diagnosis in patients.
  • HapMap The International HapMap Consortium. 2003; available on the World Wide Web at hapmap.ncbi.nlm.nih.gov/index.html.en).
  • the HapMap Project is an international project that aims to compare the genetic sequences of different individuals to identify chromosomal regions containing shared genetic variants.
  • the HapMap www site provides tools to identify
  • chromosomal regions and the variant therein with options to drill down to population level frequency data.
  • the present invention involves the identification and cataloguing of naturally-occurring human genomic target sequence variants, including those found to be relatively low-frequency or rare variants that segregate with specific human ethnic populations and in many individual humans.
  • An aspect of the invention is based on rational design of sequence selection addressing the desirability to tailor medicaments and diagnostics to rarer, but yet still significant groups of human individuals that suffer from, or have the potential to suffer from (ie, who are at risk of), a disease or condition mediated or associated with the target of interest.
  • the inventor included considerations of the spread of prevalence of naturally-occurring target variant sequences across multiple, diverse human ethnic populations, as well as the importance of addressing such populations where many individuals are likely to display a genotype and/or phenotype of one or more of the variants being analysed.
  • the inventor saw the importance of adopting the art -recognised classifications of human ethnic populations, and in this respect the inventor based the analysis and design on the recognised human ethnic populations adopted by the 1000 Genomes Project, since this is a resource that is, and will continue to be, widely adopted by the scientific and medical community.
  • the inventor designed the following variant sequence selection criteria, these being criteria that the inventor realised would provide for useful medical drugs and diagnostics to tailored need in the human population.
  • the inventor's selection included, as a consideration, selection for nucleotide variation that produced amino acid variation in corresponding TOI forms (ie, non-synonymous variations), as opposed to silent variations that do not alter amino acid residues in the target protein.
  • the cumulative human allele frequency is 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 20% or 1 to 15% or 1 to 10%.
  • the total human genotype frequency is 35, 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 25%, 1 to 20%, 1 to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%.
  • the naturally -occurring human target variant sequences are found in at least 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 different human ethnic populations (using the standard categorisation of the 1000 Genomes Project).
  • the naturally -occurring human target variant sequences are found in at least 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 130, 140 or 150 individuals distributed across such many different ethnic populations.
  • the criteria are applied with reference to one or more human genomic sequence databases as described herein.
  • the criteria are those as applied to the 1000 Genomes database.
  • the 1000 Genomes database release 13 For example, the 1000 Genomes database in its most recent version as at 1 October 2013.
  • further sequence analysis and 3D in silico modelling can also be used as an additional selection criterion: variants whose variant amino acid residues (versus the most common form of human TOI) are surface-exposed on the target are desirable for selection, since the inventor saw these as contributing to determining the topography of the target and potentially contributing to how and where ligand binding on the target occurs.
  • the ethnic populations are selected from those identified in the 1000 Genomes Project database.
  • Table 4 which provides details of the ethnic populations on which the 1000 Genomes Project database is based.
  • N A Rosenberg et al (Science 20 December 2002: vol. 298 no. 5602 2342-2343) studied the genetic structure of human populations of differing geographical ancestry. In total, 52 populations were sampled, these being populations with: [00137] African ancestry
  • the International HapMap Project Nature, 2003 Dec 18;426(6968):789-96, discloses that goal of the HapMap Project: to determine the common patterns of DNA sequence variation in the human genome by determining the genotypes of one million or more sequence variants, their frequencies and the degree of association between them in DNA samples from populations with ancestry from parts of Africa, Asia and Europe.
  • the relevant human populations of differing geographical ancestry include Yoruba, Japanese, Chinese, Northern European and Western European populations. More specifically: -
  • a suitable sample of human populations used in the present invention is as follows :-
  • each human population is selected from a population marked "(a)" above.
  • each human population is selected from a population marked "(b)" above.
  • each human population is selected from a population marked "(c)" above.
  • the ethnic populations are selected from the group consisting of an ethnic population with European ancestry, an ethnic population with East Asian, an ethnic population with West African ancestry, an ethnic population with Americas ancestry and an ethnic population with South Asian ancestry.
  • the ethnic populations are selected from the group consisting of an ethnic population with Northern European ancestry; or an ethnic population with Western European ancestry; or an ethnic population with Toscani ancestry; or an ethnic population with British ancestry; or an ethnic population with Icelandic ancestry; or an ethnic population with Finnish ancestry; or an ethnic population with Iberian ancestry; or an ethnic population with Japanese ancestry; or an ethnic population with Chinese ancestry; or an ethnic population Vietnamese ancestry; or an ethnic population with Yoruba ancestry; or an ethnic population with Luhya ancestry; or an ethnic population with Gambian ancestry; or an ethnic population with Malawian ancestry; or an ethnic population with Native American ancestry; or an ethnic population with Afro-Caribbean ancestry; or an ethnic population with Mexican ancestry; or an ethnic population with Puerto Rican ancestry; or an ethnic population with Columbian ancestry; or an ethnic population of
  • the invention provides useful anti-target ligands for addressing humans suffering from or likely to suffer from a disease or condition mediated or associated with the TOI.
  • the ligand specifically binds to the TOI variant as per the invention.
  • the ligand may inhibit or antagonise the activity of the target, eg, the ligand neutralises the target.
  • the skilled person will be familiar with neutralising ligands in general, such as antibodies or antibody fragments, and can readily test suitable ligands for specific binding and/or neutralisation of a target in vitro or in an in vivo assay.
  • An antibody "fragment” comprises a portion intact antibody, preferably the antigen binding and/or the variable region of the intact antibody.
  • antibody fragments include dAb, Fab, Fab', F(ab')2 and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules and multispecific antibodies formed from antibody fragments.
  • the ligand of the invention is or comprises an antibody or antibody fragment, for example an antibody or fragment comprising human variable regions (and optionally also human constant regions).
  • Anti-TOI or TOI-binding or targeting antibodies and fragments can be prepared according to any known method, eg, using transgenic mice (eg, the KymouseTM or
  • VelocimouseTM or OmnimouseTM , XenomouseTM, HuMab MouseTM or MeMo MouseTM), rats (eg, the OmniratTM), camelids, sharks, rabbits, chickens or other non-human animals immunised with the TOI followed optionally by humanisation of the constant regions and/or variable regions to produce human or humanised antibodies.
  • display technologies can be used, such as yeast, phage or ribosome display, as will be apparent to the skilled person.
  • Standard affinity maturation eg, using a display technology, can be performed in a further step after isolation of an antibody lead from a transgenic animal, phage display library or other library.
  • a VELOCIMMU ETM or other mouse or rat can be challenged with the antigen of interest, and lymphatic cells (such as B -cells) are recovered from the mice that express antibodies.
  • lymphatic cells such as B -cells
  • the lymphatic cells may be fused with a myeloma 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.
  • DNA encoding the variable regions of the heavy chain and light chain may be isolated and linked to desirable isotypic constant regions of the heavy chain and light chain.
  • Such an antibody protein may be produced in a cell, such as a CHO cell.
  • DNA encoding the antigen-specific chimaeric antibodies or the variable domains of the light and heavy chains may be isolated directly from antigen-specific lymphocytes.
  • high affinity chimaeric antibodies are isolated having a human variable region and a mouse constant region.
  • the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc.
  • the mouse constant regions are replaced with a desired human constant region to generate the fully human antibody of the invention, for example wild-type or modified IgGl or IgG4 (for example, SEQ ID NO: 751, 752,753 in
  • the ligand of the invention is or comprises a nucleic acid, eg, RNA, eg, siRNA that hybridises under stringent condition to the TOI variant sequence, eg, hybridises a nucleotide sequence comprising one or more nucleotides that are variant (versus the most common TOI sequence, eg, with reference to the 1000 Genomes Project database).
  • RNA eg, siRNA that hybridises under stringent condition to the TOI variant sequence
  • hybridises a nucleotide sequence comprising one or more nucleotides that are variant (versus the most common TOI sequence, eg, with reference to the 1000 Genomes Project database).
  • Target binding ability, specificity and affinity can be determined by any routine method in the art, eg, by surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • Kd is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction.
  • the surface plasmon resonance (SPR) is carried out at 25°C. In another embodiment, the SPR is carried out at 37°C.
  • the SPR is carried out at physiological pH, such as about pH7 or at pH7.6 (eg, using Hepes buffered saline at pH7.6 (also referred to as HBS-EP)).
  • physiological pH such as about pH7 or at pH7.6 (eg, using Hepes buffered saline at pH7.6 (also referred to as HBS-EP)).
  • the SPR is carried out at a physiological salt level, eg, 150mM NaCl.
  • the SPR is carried out at a detergent level of no greater than 0.05% by volume, eg, in the presence of P20 (polysorbate 20; eg, Tween-20TM) at 0.05% and EDTA at 3mM.
  • P20 polysorbate 20; eg, Tween-20TM
  • EDTA EDTA
  • the SPR is carried out at 25°C or 37°C in a buffer at pH7.6, 150mM NaCl, 0.05% detergent (eg, P20) and 3mM EDTA.
  • the buffer can contain lOmM Hepes.
  • the SPR is carried out at 25°C or 37°C in HBS-EP.
  • HBS-EP is available from Teknova Inc (California; catalogue number H8022).
  • the affinity of the ligand is determined using SPR by 1. Coupling anti-mouse (or other relevant human, rat or non-human vertebrate antibody constant region species -matched) IgG (eg, BiacoreTM BR-1008-38) to a biosensor chip (eg, GLM chip) such as by primary amine coupling; 2. Exposing the anti-mouse IgG (or other matched species antibody) to a test IgG antibody to capture test antibody on the chip;
  • SPR surface plasmon resonance
  • Regeneration of the capture surface can be carried out with lOmM glycine at pH1.7. This removes the captured antibody and allows the surface to be used for another interaction.
  • the binding data can be fitted to 1 : 1 model inherent using standard techniques, eg, using a model inherent to the ProteOn XPR36TM analysis software.
  • the ligand of the invention is contained in a medical container, eg, a vial, syringe, IV container or an injection device (eg, an intraocular or intravitreal injection device).
  • the ligand is in vitro, eg, in a sterile container.
  • the invention provides a kit comprising the ligand of the invention, packaging and instructions for use in treating or preventing or diagnosing in a human a disease or condition mediated by the TOI.
  • the instructions indicate that the human should be genotyped for a TOI variant sequence of the invention before administering the ligand to the human.
  • the instructions indicate that the human should be phenotyped for a TOI variant of the invention before administering the ligand to the human.
  • the human is of Chinese (eg, Han) ethnicity and the instructions are in Chinese (eg, Mandarin).
  • the instructions comprise directions to administer alirocumab or evolocumab to said human.
  • Clause 1 A method of treating or preventing a disease or condition in a human, wherein the disease or condition is mediated by a Target of Interest (TOI), wherein the TOI is present in humans as different polymorphic variants, the method comprising
  • step (a) said human has been or is genotyped as positive for said nucleotide sequence or phenotyped as positive for said TOI variant, or the method comprises before step (a) genotyping the human as positive for said nucleotide sequence or phenotyping the human as positive for said TOI variant.
  • frequencies may be determined using bioinformatics.
  • frequencies may be determined by reference to a database comprising at least 1000 or 2000 human sequences.
  • heterozygous human genotype frequency means the cumulative frequency of all genotypes in the sample or database or in humans having one occurrence of the rare variant allele and one occurrence of another allele (heterozygous state), eg, genotype in 1000 Genomes database.
  • homozygous human genotype frequency means the cumulative frequency of two occurrences of the variant allele (homozygous state), eg, genotype in 1000 Genomes Project database.
  • total human genotype frequency means the total of heterozygous plus homozygous human genotype frequencies.
  • cumulative human allele frequency refers to the total of all occurrences of the variant allele (eg, SNP) in the sample or database or in humans, eg, in the 1000 Genomes Project database.
  • Clause 2 The method of clause 1, wherein before step (a) the ligand has been or is determined as being capable of binding to said TOI variant, eg, with an affinity (Kd) disclosed below.
  • the ligand is (or has been determined as) a neutraliser of the TOI.
  • determination is carried out in a human (eg, in a clinical trial).
  • determination is carried out in a non-human, eg, in a mouse, rat, rabbit, pig, dog, sheep or non-human primate (eg, Cynomolgous monkey, rhesus monkey or baboon).
  • Clause 3 A method of treating or preventing a disease or condition in a human, wherein the disease or condition is mediated by a Target of Interest (TOI), wherein the TOI is present in humans as different polymorphic variants, the method comprising
  • the ligand Before step (a) the ligand has been or is determined as capable of binding to said TOI variant, eg, with an affinity (Kd) disclosed below.
  • the ligand is (or has been determined as) a neutraliser of the TOI.
  • determination is carried out in a human (eg, in a clinical trial).
  • determination is carried out in a non-human, eg, in a mouse, rat, rabbit, pig, dog, sheep or non-human primate (eg, Cynomolgous monkey, rhesus monkey or baboon).
  • Clause 4 The method of clause 3, wherein the genome of said human comprises a nucleotide sequence encoding said TOI variant; and before step (a) said nucleotide sequence has been or is determined as having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the TOI variant is not the most frequent.
  • Clause 5 The method of clause 3 or 4, wherein said human has been or is genotyped as positive for said variant nucleotide sequence before step (a), or the method comprises genotyping the human as positive for said variant nucleotide sequence before step (a).
  • Clause 6 The method of any preceding clause, wherein the human has been or is phenotyped as positive for said TOI variant before step (a), or the method comprises phenotyping the human as positive for said variant nucleotide sequence before step (a).
  • Clause 7 The method of any preceding clause, wherein said frequency is less than 10 or 15% (eg, from 1 to 10%).
  • the cumulative human allele frequency is 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 20% or 1 to 15% or 1 to 10%.
  • the total human genotype frequency is 35, 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 25%, 1 to 20%, 1 to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%.
  • Clause 8 The method of any preceding clause, wherein the ligand is capable of binding to two or more different TOI variants, each being encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% (eg, from 1 to 10%) and/or having a total human genotype frequency of less than 50% (eg, from 1 to 20%).
  • the cumulative human allele frequency of each TOI variant is 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 20% or 1 to 15% or 1 to 10%.
  • the total human genotype frequency of each TOI variant is 35, 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 25%, 1 to 20%, 1 to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%.
  • Clause 9 A method of treating or preventing a disease or condition in a human, wherein the disease or condition is mediated by a Target of Interest (TOI), wherein the TOI is present in humans as different polymorphic variants, the method comprising
  • a. Administering to the human an anti-TOI ligand to target a TOI variant in the human and treat or prevent (eg, by at least 40, 50, 60, 70, 80, 90 or 95%) said disease or condition, wherein the TOI in said human is a variant encoded by a nucleotide sequence having a cumulative human allele frequency of more than 50% (eg, the highest frequency) and/or having a total human genotype frequency of more than 50% (eg, the highest frequency);
  • step (a) said human has been or is genotyped as negative for a variant
  • the method comprises genotyping the human as negative for a variant nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%; or phenotyping the human as negative for a TOI variant encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the cumulative human allele frequency is 55, 60, 65, 70, 75, 80, 85 or 90 or more but less than 95, 96, 97, 98, 99 or 100% (eg, in the range from 51 to 80%).
  • the total human genotype frequency is 55, 60, 65, 70, 75, 80, 85 or 90 or more but less than 95, 96, 97, 98, 99 or 100% (eg, in the range from 51 to 80%).
  • the cumulative human allele frequency is 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 20% or 1 to 15% or 1 to 10%.
  • the total human genotype frequency is 35, 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 25%, 1 to 20%, 1 to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%.
  • Clause 10 The method of clause 9, wherein before step (a), the human has been or is phenotyped as positive for the most frequent TOI variant or genotyped for the nucleotide sequence thereof.
  • the human before step (a) the human has been or is genotyped as positive for TOI variant nucleotide sequence having a cumulative human allele frequency of 55, 60, 65, 70, 75, 80, 85 or 90 or more but less than 95, 96, 97, 98, 99 or 100% (eg, in the range from 51 to 80%) or phenotyped for the TOI variant thereof.
  • step (a) the human has been or is genotyped as positive for TOI variant nucleotide sequence having a total human genotype frequency of 55, 60, 65, 70, 75, 80, 85 or 90 or more but less than 95, 96, 97, 98, 99 or 100% (eg, in the range from 51 to 80%) or phenotyped for the TOI variant thereof.
  • Clause 11 The method of clause 9 or 10, wherein before step (a) the ligand has been or is determined as being capable of binding to the most frequent TOI variant.
  • Clause 12 The method of clause 9, 10 or 11, wherein before step (a) the ligand has been or is determined as being substantially incapable of neutralising or inhibiting said TOI variant recited in step (b).
  • substantially incapable or neutralising or inhibiting is meant: Neutralisation or inhibition less than 50, 25, 10, 5 or 0.5% inhibition or neutralisation of the most frequent TOI variant.
  • Clause 13 The method of any one of clauses 9 to 12, wherein the ligand is capable of binding to the most frequent TOI variant.
  • Clause 14 The method of any one of clauses 9 to 13, wherein the ligand is capable of binding to two or more different TOI variants, each being encoded by a nucleotide sequence having a cumulative human allele frequency of more than 50%.
  • each TOI variant is encoded by a nucleotide sequence having a cumulative human allele frequency of 55, 60, 65, 70, 75, 80, 85 or 90 or more but less than 95, 96, 97, 98, 99 or 100% (eg, in the range from 51 to 80%).
  • each TOI variant is encoded by a nucleotide sequence having a total human genotype frequency of 55, 60, 65, 70, 75, 80, 85 or 90 or more but less than 95, 96, 97, 98, 99 or 100% (eg, in the range from 51 to 80%).
  • Clause 15 The method of any preceding clause, wherein said variant nucleotide sequence recited in step (a) has been or is determined as being present in at least 2 different human ethnic populations, eg, at least 2, 3, 4, 5, 6, 7, 8 or 9 different human ethnic populations in Table 4.
  • Clause 16 The method of any preceding clause, wherein said human frequency is the frequency in a database of naturally-occurring sequences sampled from at least 15, 20 or 25 different human ethnic populations and comprising at least 1000 sequences.
  • the database is the 1000 Genomes Project database as described herein.
  • Clause 17 An anti-human TOI ligand for use in a method of treating and/or preventing a TOI-mediated disease or condition in a human, wherein the TOI is present in humans as different polymorphic variants and wherein the genome of said human comprises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, the method comprising administering the ligand to the human.
  • clause 17 provides an anti-human TOI ligand for use in a method according to any one of clauses 1 to 16, the method comprising administering the ligand to the human.
  • the cumulative human allele frequency is 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 20% or 1 to 15% or 1 to 10%.
  • the total human genotype frequency is 35, 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 25%, 1 to 20%, 1 to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%.
  • Clause 18 The ligand of clause 17, wherein the ligand has been or is determined as capable of binding the human TOI encoded by said nucleotide sequence.
  • clause 18 provides a ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, for use in a method comprising the step of using the ligand to target said TOI in a human to treat and/or prevent a disease or condition mediated by TOI, the method comprising administering the ligand to the human.
  • Clause 19 A ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, for use in a method according to any one of clauses 1 to 16, the method comprising administering the ligand to the human.
  • Clause 20 The ligand of any one of clauses 17 to 19, wherein the human has been or is genotyped as positive for said TOI nucleotide sequence having a cumulative human allele frequency of less than 50%.
  • Clause 21 The ligand of any one of clauses 17 to 20, wherein the human has been or is phenotyped as positive for a TOI encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • Clause 22 The ligand of any one of clauses 17 to 21, wherein the human has been or is genotyped as heterozygous for a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%;
  • the human has been or is genotyped as comprising a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and a TOI nucleotide sequence having a cumulative human allele frequency of more than 50% (eg, having the highest cumulative human allele frequency) and/or having a total human genotype frequency of more than 50% (eg, having the highest total human genotype frequency).
  • Clause 23 The ligand of any one of clauses 17 to 22, wherein the genome of the human has been or is genotyped as homozygous for a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • Clause 24 The ligand of any one of clauses 17 to 23, wherein the ligand comprises an antibody binding site that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% ; and optionally has been or is determined as capable of such binding.
  • Clause 25 The ligand of clause 24, wherein the ligand is an antibody or antibody fragment.
  • Clause 26 The ligand of any one of clauses 17 to 23, wherein the ligand comprises a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the ligand comprises a nucleotide sequence that comprises at least 10 contiguous nucleotides of a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • the ligand comprises a nucleotide sequence that comprises at least 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50 or 100 contiguous nucleotides of a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • Clause 27 The ligand of any one of clauses 17 to 26, wherein the genome of said human comprises a nucleotide sequence having a cumulative human allele frequency of less than 50% and the sequence is found in at least 2 different ethnic populations (eg, found in at least 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 different human ethnic populations (for example as per the populations in Table 4)).
  • numbers are with reference to the 1000 Genomes Project database.
  • numbers are with reference to the 1000 Genomes Project database.
  • Clause 28 A pharmaceutical composition or kit for treating or preventing a condition or disease mediated by a TOI as recited in any preceding clause, the composition or kit comprising a ligand of any one of clauses 17 to 27; and optionally in combination with a label or instructions for use to treat and/or prevent said disease or condition in a human; optionally wherein the label or instructions comprise a marketing authorisation number (eg, an FDA or EMA authorisation number); optionally wherein the kit comprises an injection pen or IV container that comprises the ligand.
  • a marketing authorisation number eg, an FDA or EMA authorisation number
  • the label or instructions cover or describe use for a human comprising a TOI variant encoded by a nucleotide sequence as recited in clause 17.
  • Clause 29 A method of producing an anti-human TOI antibody binding site, the method comprising obtaining a plurality of anti-TOI antibody binding sites, screening the antibody binding sites for binding to a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody binding site that binds in the screening step.
  • the antibody, fragment or binding site is recombinant.
  • clause 29 provides: A method of producing an anti-human TOI antibody, the method comprising immunising a non-human vertebrate (eg, a mouse or a rat) with a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody that binds a TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, and optionally producing a TOI -binding fragment or derivative of the isolated antibody.
  • a non-human vertebrate eg, a mouse or a rat
  • isolated with reference to a ligand, antibody or protein, for example in any aspect, configuration, example or emodiment, means that a subject ligand, antibody, protein etc (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" ligand, antibody, protein etc 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 ligand, antibody protein etc.
  • the isolated ligand, antibody protein etc 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.
  • an "isolated" antibody is one that has been identified, separated and/or recovered from a component of its production environment (eg, naturally or recombinantly).
  • the isolated polypeptide is free of association with all other components from its production environment, eg, so that the antibody has been isolated to an FDA-approvable or approved standard.
  • Contaminant components of its production environment such as that resulting from recombinant transfected cells, are materials that would typically interfere with research, diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the polypeptide will be purified: (1) to greater than 95% by weight of antibody as determined by, for example, the Lowry method, and in some embodiments, to greater than 99% by weight; (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under non -reducing or reducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, an isolated polypeptide or antibody will be prepared by at least one purification step.
  • the invention encompasses the ligand (eg, antibody) conjugated to a therapeutic moiety (“immunoconjugate”), such as a cytotoxin, a chemotherapeutic drug, an immunosuppressant or a radioisotope.
  • a therapeutic moiety such as a cytotoxin, a chemotherapeutic drug, an immunosuppressant or a radioisotope.
  • Cytotoxin agents include any agent that is detrimental to cells. Examples of suitable cytotoxin agents and chemotherapeutic agents for forming immunoconjugates are known in the art, see for example, WO 05/103081.
  • the antibodies of the present invention may be monospecific, bispecific, or multispecific. Multispecific mAbs may be specific for different epitopes of one target polypeptide or may contain antigen-binding domains specific for more than one target polypeptide. See, e.g., Tutt et al. (1991) J. Immunol. 147:60-69.
  • the human anti-TOI (eg, anti-PCSK9) mAbs can be linked to or co-expressed with another functional molecule, e.g., another peptide or protein.
  • an antibody or fragment thereof can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody or antibody fragment, to produce a bispecific or a multispecific antibody with a second binding specificity.
  • An exemplary bi-specific antibody format that can be used in the context of the present invention involves the use of a first immunoglobulin (Ig) CH3 domain and a second Ig CH3 domain, wherein the first and second Ig CH3 domains differ from one another by at least one amino acid, and wherein at least one amino acid difference reduces binding of the bispecific antibody to Protein A as compared to a bi-specific antibody lacking the amino acid difference.
  • the first Ig CH3 domain binds Protein A and the second Ig CH3 domain contains a mutation that reduces or abolishes Protein A binding such as an H95R modification (by IMGT exon numbering; H435R by EU numbering).
  • the second CH3 may further comprise a Y96F modification (by IMGT; Y436F by EU). Further modifications that may be found within the second CH3 include: D16E, L18M, N44S, K52N, V57M, and V821 (by IMGT; D356E, L358M, N384S, K392N, V397M, and V422I by EU) in the case of IgGl antibodies; N44S, K52N, and V82I (IMGT; N384S, K392N, and V422I by EU) in the case of IgG2 antibodies; and Q15R, N44S, K52N, V57M, R69K, E79Q, and V82I (by IMGT; Q355R, N3845, K392N, V397M, R409K, E419Q, and V422I by EU) in the case of IgG4 antibodies.
  • Clause 30 The method of clause 29, comprising the step of obtaining a nucleic acid encoding the antibody, fragment, derivative or binding site and optionally inserting the nucleic acid in an expression vector.
  • kits for TOI genotyping a human comprising a nucleic acid comprising a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the nucleic acid comprises a nucleotide sequence that comprises at least 10 (eg, at least 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50 or 100) contiguous nucleotides of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • the kit comprises a nucleic acid comprising a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the nu
  • the nucleic acid hybridises to a region immediately flanking a nucleotide that is variant compared to the corresponding nucleotide of the TOI nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency.
  • the nucleic acid hybridises to at two or more such variant nucleotides.
  • Specific hybridisation is under stringent conditions, as will be apparent to the skilled person, eg, conditions of 5xSSC, 5xDenhardt's reagent, and 0.5% SDS at 65° C.
  • Clause 32 A kit for TOI genotyping or phenotyping a human, wherein the kit comprises a ligand according to any one of clauses 17 to 27 or an antibody, fragment or derivative produced by the method of any one of clauses 29 to 31.
  • the ligand specifically binds to an epitope comprising an amino acid that is variant compared to the corresponding amino acid of the TOI encoded by a nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency.
  • the ligand specifically binds to an epitope comprising two or more such variant amino acids.
  • specific binding means binding with an affinity (Kd) of lmM, ⁇ , ⁇ or InM or less, eg, as determined by SPR.
  • epitopes is a region of an antigen that is bound by an antibody.
  • Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may also be conformational, that is, composed of non-linear amino acids.
  • epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • Clause 33 Use of an anti-TOI ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for treating and/or preventing a TOI-mediated disease or condition in a human whose genome comprises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • Clause 34 Use of an anti-TOI ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for targeting said TOI in a human to treat and/or prevent a disease or condition mediated by TOI.
  • Clause 35 A method of targeting a TOI for treating and/or preventing a TOI-mediated disease or condition in a human, the method comprising administering an anti-TOI ligand to a human comprising a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, whereby a TOI encoded by said nucleotide sequence is targeted.
  • Clause 36 The method of clause 35, wherein the method comprises targeting a human TOI comprising an amino acid sequence with said ligand to treat and/or prevent said disease or condition in said human, wherein said amino acid sequence is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%.
  • Clause 37 A method of TOI genotyping a nucleic acid sample of a human, the method comprising identifying in the sample the presence of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the method comprises obtaining a TOI nucleic acid sample from the human and then carrying out the identifying step.
  • Clause 38 A method of TOI typing a protein sample of a human, the method comprising identifying in the sample the presence of a TOI amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the method comprises obtaining a TOI protein sample from the human and then carrying out the identifying step.
  • Clause 39 The method of clause 37 or 38, comprising obtaining a sample of serum, blood, faeces, hair, urine or saliva from a human, whereby the nucleic acid or protein sample is obtained for use in the step of identifying said sequence.
  • Clause 40 The method of any one of clauses 37 to 39, comprising using a ligand according to any one of clauses 17 to 27 to carry out said identifying step.
  • Clause 41 A diagnostic kit comprising a ligand that is capable of binding a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% and instructions for carrying out the method of clause 38 or 39.
  • Clause 42 A diagnostic kit comprising a nucleic acid probe comprising a nucleotide sequence that specifically hybridises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof and instructions for carrying out the method of clause 38 or 39.
  • Clause 43 The method, ligand, composition, kit or use of any preceding clause, wherein the TOI is encoded by a nucleotide sequence having a cumulative human allele frequency from 1 to
  • the TOI is human PCSK9, eg, a mature, cleaved, autocatalysed or active PCSK9.
  • the disease is a cardiovascular disease such as hyperlipidaemia.
  • Ligands of the invention are useful, for instance, in specific binding assays, for genotyping or phenotyping humans, affinity purification of the TOI and in screening assays to identify other antagonists of TOI activity. Some of the ligands of the invention are useful for inhibiting binding of TOI to a congnate human receptor or protein, or inhibiting TOI-mediated activities.
  • the invention encompasses anti-TOI (eg, PCSK9) antibody ligands having a modified glycosylation pattern.
  • modification to remove undesirable glycosylation sites may be useful, or e.g., removal of a fucose moiety to increase antibody dependent cellular cytotoxicity (ADCC) function (see Shield et al. (2002) JBC 277:26733).
  • ADCC antibody dependent cellular cytotoxicity
  • modification of galactosylation can be made in order to modify complement dependent cytotoxicity (CDC).
  • the invention features a pharmaceutical composition
  • a pharmaceutical composition comprising a ligand of the invention, wherein the ligand is or comprises a recombinant human antibody or fragment thereof which specifically binds the TOI (eg, a rare variant as described herein) and a
  • the invention features a composition which is a combination of an antibody ligand or antigen-binding fragment of an antibody of the invention, and a second therapeutic agent.
  • the second therapeutic agent may be any of an anti-inflammatory agent, an anti-angiogenesis agent, a painkiller, a diuretic, a chemotherapeutic agent, an anti -neoplastic agent, a vasodilator, a vasoconstrictor, a statin, a beta blocker, a nutrient, an adjuvant, an anti-obesity agent and an anti -diabetes agent.
  • “Pharmaceutically acceptable” refers to approved or approvable by a regulatory agency of the USA Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
  • a “pharmaceutically acceptable carrier, excipient, or adjuvant” refers to an carrier, excipient, or adjuvant that can be administered to a subject, together with an agent, e.g., any antibody or antibody chain described herein, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the agent.
  • the invention features a method for inhibiting TOI activity using the anti- TOI ligand of the invention (eg, an antibody or antigen-binding portion of the antibody of the invention), wherein the therapeutic method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising the ligand.
  • the disorder treated is any disease or condition which is improved, ameliorated, inhibited or prevented by removal, inhibition or reduction of TOI activity.
  • terapéuticaally effective amount is meant an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • the term "specifically binds,” or the like, means that a ligand, eg, an antibody or antigen- binding fragment thereof, forms a complex with an antigen that is relatively stable under physiologic conditions. Specific binding can be characterized by an equilibrium dissociation constant of at least about 1x10 6 M or less (e.g., a smaller KD denotes a tighter binding). Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. An isolated antibody that specifically binds a human TOI may, however, exhibit cross-reactivity to other antigens such as a TOI molecule from another species. Moreover, multi-specific antibodies (e.g., bispecifics) that bind to human TOI and one or more additional antigens are nonetheless considered antibodies that "specifically bind" TOI, as used herein.
  • SNPlex is a proprietary genotyping platform sold by Applied Biosystems.
  • Miniaturized assays such as microarrays with oligonucleotide reagents immobilized on small surfaces, are frequently proposed for large-scale mutation analysis and high-throughput genotyping (Large-scale identification, mapping, and genotyping of single-nucleotide polymorphisms in the human genome (Wang DG, Fan JB, Siao CJ, Berno A, Young P, Sapolsky R, Ghandour G, Perkins N, Winchester E, Spencer J, Kruglyak L, Stein L, Hsie L, Topaloglou T, Hubbell E, Robinson E, Mittmann M, Morris MS, Shen N, Kilburn D, Rioux J, Nusbaum C, Rozen S, Hudson TJ, Lipshutz R, Chee M, Lander ES, Science.
  • Cleavage separates a 5'-fluorophore from a 3 '-quencher leading to detectable fluorescent signal.
  • the use of two allele-specific probes carrying different fluorophores permits SNP determination in the same tube without any post-PCR processing. Genotype is determined from the ratio of intensities of the two fluorescent probes at the end of amplification. Thus, rather than taking advantage of the full set of real-time PCR data as in quantitative studies, only end-point data are used.
  • TaqMan SNP genotyping in a high-throughput, automated manner is facilitated by the use of validated Pre-made TaqMan ® Genotyping assays, but Custom TaqMan ® Assays may also be used (High-throughput genotyping with single nucleotide polymorphisms, Ranade K, Chang MS, Ting CT, Pei D, Hsiao CF, Olivier M, Pesich R, Hebert J, Chen YD, Dzau VJ, Curb D, Olshen R, Risch N, Cox DR, Botstein D, Genome Res.
  • Single nucleotide polymorphisms can be determined using TaqManTM real-time PCR assays (Applied Biosystems) and commercial software that assigns genotypes based on reporter probe signals at the end of amplification.
  • An algorithm for automatic genotype caling of SNPs using the full course of TaqMan real-time data is available for use (A. Callegaro et al, Nucleic Acids Res. 2006; 34(7): e56, Published online 2006 April 14. doi: 10.1093/nar/gkll85, PMCID: PMC1440877).
  • the algorithm is unique in that it classifies samples according to the behavior of blanks (no DNA samples), which cluster with heterozygous samples. This method of classification eliminates the need for positive controls and permits accurate genotyping even in the absence of a genotype class, for example when one allele is rare.
  • the invention provides therapeutic compositions comprising the anti-TOI ligand, eg, antibodies or antigen-binding fragments thereof, of the present invention.
  • the administration of therapeutic compositions in accordance with the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • a multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. "Compendium of excipients for parenteral formulations" PDA (1998) J Pharm Sci Technol 52:238-311.
  • the dose may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like.
  • the ligand, eg, antibody, of the present invention is used for treating various conditions and diseases associated with the TOI in an adult patient, it is advantageous to intravenously administer the antibody of the present invention normally at a single dose of about 0.01 to about 20 mg/kg body weight, more preferably about 0.02 to about 7, about 0.03 to about 5, or about 0.05 to about 3 mg/kg body weight.
  • the frequency and the duration of the treatment can be adjusted.
  • ligand or pharmaceutical composition of the invention for example a ligand provided by e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al. (1987) J. Biol. Chem. 262:4429-4432).
  • Methods of introduction include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the ligand or composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • epithelial or mucocutaneous linings e.g., oral mucosa, rectal and intestinal mucosa, etc.
  • Administration can be systemic or local.
  • the ligand or pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249: 1527-1533; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365; Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).
  • a liposome see Langer (1990) Science 249: 1527-1533; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365; Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).
  • the ligand or pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201).
  • polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974).
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138, 1984).
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe.
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition.
  • the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition.
  • the pen delivery device can then be reused.
  • a disposable pen delivery device there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a ligand or pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK), DISETRONICTM pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTMI, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTM pen (Becton Dickinson, Franklin Lakes, N.J.),
  • OPTIPENTTM OPTIPENTTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTTM (sanofi-aventis, Frankfurt, Germany), to name only a few.
  • disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTARTM pen (sanofi-aventis), the FLEXPENTM (Novo Nordisk), and the KWIKPENTM (Eli Lilly).
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the ligand(s).
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of the aforesaid antibody contained is generally about 5 to about 500 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the aforesaid antibody is contained in about 5 to about 100 mg and in about 10 to about 250 mg for the other dosage forms.
  • Exemplary TOIs For example in any configuration, aspect, concept, example or configuration of the invention, the or each TOI is selected from the group consisting of ABCF1 ; ACVR1 ; ACVR1B; ACVR2; ACVR2B; ACVRL1 ; ADORA2A; Aggrecan; AGR2; AICDA; AW1; AIG1 ; AKAP1 ;
  • CCL22 MDC / STC-1
  • CCL23 MPIF-1
  • CCL24 MPIF-2 I eotaxin-2
  • CCL25 TECK
  • CCL26 eotaxin-3
  • CCL27 CCL28; CCL3 (MIP-la); CCL4 (MIP-lb); CCL5 (RANTES); CCL7 (MCP-3); CCL8 (mcp-2); CCNA1 ; CCNA2; CCND1 ; CCNE1 ; CCNE2; CCR1 (CKR1 / HM145); CCR2 (mcp-lRB / RA);CCR3 (CKR3 / CMKBR3); CCR4; CCR5 (CMKBR5 / ChemR13); CCR6 (CMKBR6 / CKR-L3 / STRL22 / DRY6); CCR7 (CKR7 / EBI1); CCR8
  • CDKBR8 / TER1 / CKR-L1 CCR9
  • CCR9 GPR-9-6
  • CCRL1 VSHK1
  • CCRL2 L-CCR
  • FCER1A FCER2; FCGR3A; FCRL4; FGF; FGF1 (aFGF); FGFIO; FGF11; FGF12; FGF12B;
  • IL1RAPL1 IL1RAPL2; IL1RL1 ;IL1RL2 IL1RN; 1L2; 1L20; IL20RA; IL21R; 1L22; 1L22R;
  • KRTHB6 hair-specific type II keratin
  • LAG3 LAMA5; LEP (leptin); LIGHT; Lingo-p75; Lingo- Troy; LPS; LRP5; LTA (TNF-b); LTB; LTB4R (GPR16); LTB4R2; LTBR; MACMARCKS; MAG or Omgp; MAP2K7 (c-Jun); MDK; MIB1 ; midkine; MIF; MIP-2; MK167 (Ki-67); MMP2; MMP9; MS4A1 ; MS MB; MT3 (metallothionectin-ifi); MTSS 1 ; MUC 1 (mucin); MYC; MYD88; NCK2; neurocan; Na v 1.7; Na v 1.8; NFKB 1 ; NFKB2; NGFB (NGF); NGFR; NgR-Lingo; NgR-Nogo66 (Nogo); NgR-p
  • NT5E NTN4; ODZl ; OPG; OPRDl ; OX40L; OX40; P2RX7; PAP; PARTI ; PATE; PAWR; PCA3; PCNA; PCSK9, PD-1, PD-L1 ; PDGFA; PDGFB; PECAM1 ; PF4 (CXCL4); PGF; PGR; phosphacan; PIAS2; Placental Growth Factor (PIGF); PIK3CG; PLAU (uPA); PLG; PLXDC1 ; PPBP (CXCL7); PPID; PR1 ; PRKCQ; PRKD1 ; PRL; PROC; PROK2; PSAP; PSCA; PTAFR; PTEN; PTGS2 (COX- 2); PTN; RAC2 (p21Rac2); RARB; RGS1 ; RGS13; RGS3; RNF110 (ZNF144); ROB02; ROR1 ; S
  • the TOI is a human TOI selected from Table 5
  • the TOI is OX40 ligand.
  • the TOI is OX40.
  • the TOI is PCSK9.
  • the TOI is IL6 receptor (IL-6R).
  • the TOI is LIGHT.
  • the TOI is VEGF-A.
  • the TOI is TNF alpha.
  • the TOI is PIGF.
  • the TOI is IGF1R.
  • the TOI is OPG.
  • the TOI is ICOS
  • the TOI is NGF.
  • the TOI is BMP6.
  • the TOI is ferroportin.
  • the TOI is TMPRSS6.
  • the TOI is hemojuvelin.
  • the TOI is VEGF receptor.
  • the TOI is PDGF receptor.
  • the TOI is stem cell factor receptor.
  • the TOI is hepcidin.
  • the TOI is IL-4 receptor alpha.
  • the TOI is sclerostin.
  • the TOI is IL-13 receptor.
  • the TOI is CD7.
  • the TOI is delta-like ligand-4 (D114).
  • the TOI is HGF.
  • the TOI is angiopoietin-2 (Ang2).
  • the TOI is GDF8.
  • the TOI is ERBB3.
  • the TOI is IL-17 receptor.
  • the TOI is CD40.
  • the TOI is CD40 ligand.
  • the TOI is EGFR.
  • the TOI comprises a mutation as described herein for that TOI or is encoded by a nucleotide sequence comprising a SNP as described herein for that TOI.
  • the human comprises said TOI comprising said mutation; or the human comprises a nucleotide sequence encoding said TOI comprising said mutation.
  • the TOI is human PCSK9 and comprises a mutation 1474, E670G, N425S or Q619P in SEQ ID NO: 1.
  • the human comprises said PCSK9 comprising said mutation; or the human comprises a nucleotide sequence encoding said PCSK9 comprising said mutation.
  • a PCSK9 is stated to be of a specific "form", eg, form f, c, r, p, e, h, aj or q
  • the PCSK9 is a human PCSK9 comprising a mutation R46L, A53V, N425S, A443T, I474V, Q619P and E670G (eg, comprises a mutation 1474, E670G, N425S or Q619P) in SEQ ID NO: 1 ;
  • the PCSK9 comprises I474V in SEQ ID NO: 1 and optionally the human comprises such a PCSK9 or a nucleotide sequence encoding such a PCSK9 (eg, wherein the method is for treating or preventing dislipidemia, eg reducing cholesterol or maintaining reduced cholesterol in the human);
  • the PCSK9 comprises E670G in SEQ ID NO: 1 and optionally the human comprises such a PCSK9 or
  • the TOI is human IL6R and comprises a mutation D358A or V385I in SEQ ID NO: 78.
  • the human comprises said IL6R comprising said mutation; or the human comprises a nucleotide sequence encoding said IL6R comprising said mutation.
  • the TOI is human IL4Ra and comprises a mutation selected from the group consisting of I75V, E400A, C431R, S503P, Q576R and S752A in SEQ ID NO: 67.
  • the human comprises said IL4Ra comprising said mutation; or the human comprises a nucleotide sequence encoding said IL4Ra comprising said mutation.
  • the TOI is huma Navl.7 and comprises a mutation selected from the group consisting of 1361, 216S, 241T, 395K, 848T, 858H, 858F, 863P, 1449V, 996C, 1298F, 1298D, 1299F, 1461T, 1462V, 14641, 1627K, 277X (wherein X is an amino acid other than R), 328X (wherein X is an amino acid other than Y), 395K, 459X (wherein X is an amino acid other than S), 693 X (wherein X is an amino acid other than E), 767X (wherein X is an amino acid other than I), 830X (wherein X is an amino acid other than R), 897X (wherein X is an amino acid other than W), 1200L, 1235L, 1488X (wherein X is an amino acid other than R), 1659X (wherein X is an amino acid other than R), 1659X (where
  • the TOI is human VEGF-A and is encoded by a VEGF-A nucleotide sequence comprising a SNP selected from the group consisting of rs699947, rs833061, rs2010963, rs3025039, rs699946, rs2146323, rsl413711, rs833068, rs833069, rs3025000 and rsl570360.
  • a SNP selected from the group consisting of rs699947, rs833061, rs2010963, rs3025039, rs699946, rs2146323, rsl413711, rs833068, rs833069, rs3025000 and rsl570360.
  • the human comprises said VEGF-A comprising said mutation; or the human comprises a nucleotide sequence encoding said VEGF-A comprising said mutation.
  • the TOI is human PDGF-B and is encoded by a PDGF-B nucleotide sequence comprising a SNP selected from the group consisting of rsl42404523 (ie, a C corresponding to position -776) and a C at the position of rs 1800818 (ie, a C corresponding to position -735).
  • the human comprises said PDGF-B comprising said mutation; or the human comprises a nucleotide sequence encoding said PDGF-B comprising said mutation.
  • the TOI is human PDGFR-B and is encoded by a PDGFR-B nucleotide sequence comprising a SNP selected from the group consisting of rs246395 (ie, a G corresponding to position 2601) and rs74943037 (ie, a T corresponding to position 1391).
  • the human comprises said PDGFR-B comprising said mutation; or the human comprises a nucleotide sequence encoding said PDGFR-B comprising said mutation.
  • “reduction” does not encompass a complete reduction as compared to a reference level.
  • a decrease can be preferably down to a level accepted as within the range of normal for an individual without a given disorder. However, for example, for the purposes of lowering or reducing cholesterol level, for example, a reduction by about 5-10 points can be considered a "decrease" or "reduction.”
  • Inhibition refers and refers to decrease by at least 10% as compared to a reference level (e.g. the absence of a given treatment) and can include, for example, a decrease by at least about 10%, 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%, at least about 90%, at least about 95%, at least about 98%, at least about 99% , or more including 100% inhibition as compared to a reference level.
  • “Complete inhibition” refers to a 100% inhibition as compared to a reference level.
  • the terms “increased”, “increase”, “enhance”, or “activate” are all used herein to mean an increase by a statically significant amount.
  • the terms “increased”, “increase”, “enhance”, or “activate” can mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3 -fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • an "increase” is a statistically significant increase in such level.
  • the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest.
  • substantially is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena. For the removal of doubt, “substantially” can refer to at least a 90% extent or degree of a characteristic or property of interest, e.g. at least 90%, at least 92%, at least 95%, at least 98%, at least 99% or greater.
  • a "subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. In some embodiments, the subject is a mammal, e.g., a primate, e.g., a human. The terms, "individual,” “patient” and “subject” are used interchangeably herein.
  • the subject can be a non-human vertebrate, e.g. a primate, a rodent, a mouse, a rat, a pig, a sheep, a zebrafish, a frog, etc.
  • a non-human vertebrate e.g. a primate, a rodent, a mouse, a rat, a pig, a sheep, a zebrafish, a frog, etc.
  • the subject is a mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but is not limited to these examples.
  • Mammals other than humans can be advantageously used as subjects that represent animal models of a disease or condition, e.g., a cardiovascular condition.
  • a subject can be male or female.
  • a subject can be one who has been previously diagnosed with or identified as suffering from or having a condition in need of treatment or one or more complications related to such a condition, and optionally, have already undergone treatment for the condition or the one or more complications related to the condition.
  • a subject can also be one who has not been previously diagnosed as having the condition or one or more complications related to the condition.
  • a subject can be one who exhibits one or more risk factors for the condition or one or more complications related to the condition or a subject who does not exhibit risk factors.
  • a "subject in need” or “human in need” of treatment for a particular condition can be a subject having that condition, such as increased cholesterol levels, diagnosed as having that condition, or at risk of developing that condition.
  • protein and “polypeptide” are used interchangeably herein to designate a series of amino acid residues, connected to each other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues.
  • protein and “polypeptide” refer to a polymer of amino acids with natural amino acids.
  • modified polypeptides one refers to polypeptides that include modified amino acids (e.g., phosphorylated, glycated, glycosylated, etc.) and amino acid analogs, regardless of its size or function.
  • Protein and polypeptide are often used in reference to relatively large polypeptides, whereas the term “peptide” is often used in reference to small polypeptides, but usage of these terms in the art overlaps.
  • the terms “protein” and “polypeptide” are used interchangeably herein when referring to a gene product and fragments thereof.
  • exemplary polypeptides or proteins include gene products, naturally occurring proteins with the specified sequence.
  • One can also use peptide homologs, peptide orthologs, peptide paralogs, peptide fragments and other equivalents, variants, fragments, and analogs of the peptides as these terms are understood by one of ordinary skill in the art.
  • nucleic acid or “nucleic acid sequence” refers to any molecule, preferably a polymeric molecule, incorporating units of ribonucleic acid, deoxyribonucleic acid.
  • the nucleic acid can be either single-stranded or double-stranded.
  • a single-stranded nucleic acid can be one nucleic acid strand of a denatured double- stranded DNA. Alternatively, it can be a single- stranded nucleic acid not derived from any double-stranded DNA.
  • the nucleic acid can be DNA.
  • nucleic acid can be RNA.
  • Suitable nucleic acid molecules are DNA, including genomic DNA or cDNA. Other suitable nucleic acid molecules are RNA, including mRNA. In some aspects one can also use analogs of nucleic acids.
  • nucleic acid probe refers to an isolated oligonucleotide molecule having a nucleic acid sequence which can hybridize to a target nucleic acid sequence, e.g. specifically hybridize to the target sequence.
  • a nucleic acid probe can further comprise a detectable label.
  • a nucleic acid probe can be attached to a solid surface.
  • a nucleic acid from is from about 5 nt to about 100 nt in length.
  • siRNA refers to a nucleic acid that forms an RNA molecule comprising two individual strands of RNA which are substantially complementary to each other.
  • the siRNA is at least about 15-40 nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is about 15-40 nucleotides in length, and the double stranded siRNA is about 15-40 base pairs in length, preferably about 19-25 base nucleotides, e.g., 19, 20, 21, 22, 23, 24, or 25 nucleotides in length).
  • a siRNA can be blunt-ended.
  • a siRNA can comprise a 3' and/or 5' overhang on each strand having a length of about 0, 1, 2, 3, 4, or 5 nucleotides.
  • the length of the overhang is independent between the two strands, i.e., the length of the overhang on one strand is not dependent on the length of the overhang on the second strand.
  • the siRNA molecules can also comprise a 3' hydroxyl group.
  • the siRNA can comprise a 5' phosphate group.
  • a siRNA has the ability to reduce or inhibit expression of a gene or target RNA when the siRNA is present or expressed in the same cell as the target gene, e.g. the target RNA.
  • siRNA -dependent post-transcriptional silencing of gene expression involves cutting the target RNA molecule at a site guided by the siRNA.
  • PCSK9 or "proprotein convertase subtilisin/kexin type 9” refers to a serine protease involved in regulating the levels of the low density lipoprotein receptor (LDLR) protein (Horton et al., 2007; Seidah and Prat, 2007). PCSK9 has been shown to directly interact with the LDLR protein, be endocytosed along with the LDLR, and co-immunofluoresce with the LDLR throughout the endosomal pathway (Lagace et al., 2006).
  • LDLR low density lipoprotein receptor
  • PCSK9 is a prohormone -proprotein convertase in the subtilisin (S8) family of serine proteases (Seidah et al., 2003).
  • S8 subtilisin family of serine proteases
  • the sequence of PCSK9 for a variety of species is known, e.g., human PCSK9 (NCBI Gene ID No: 255738).
  • Nucleotide and polypeptide sequences for a number of PCSK9 isoforms are provided herein, e.g., SEQ ID NOs: 1-37.
  • PCSK9 exists as both a pro-form and a mature form.
  • Autocatalysis of the PCSK9 proform occurs between Glnl52 and Serl53 (VFAQISIP (SEQ ID NO: 116)) (Naureckiene et al., 2003), and has been shown to be required for its secretion from cells (Seidah et al., 2003).
  • the inactive form prior to this cleavage can be referred to herein as the "inactive”, “pro-form”, or "unprocessed” form of PCSK9.
  • the C-terminal fragment generated by the autocatalysis event can be referred to herein as the "mature,” “cleaved”, “processed” or “active” PCSK9.
  • Examples of pro-form and mature PCSK9 isoforms are provided herein, see, e.g. SEQ ID NOs: 1-27.
  • the "catalytic domain” of PCSK9 refers to the portion of a PCSK9 polypeptide corresponding to positions 153 to 449 of PCSK9, e.g. of SEQ ID NO: 1.
  • the "C-terminal domain” of PCSK9 refers to the portion of a PCSK9 polypeptide corresponding to positions 450-692 of PCSK9, e.g., of SEQ ID NO: 1.
  • a disease or condition "mediated by PCSK9” refers to a disease or condition which is caused by or characterized by a change in PCSK9, e.g. a change in expression level, a change in activity, and/or the presence of a variant or mutation of PCSK9.
  • diseases or conditions can include, for example, a lipid disorder,
  • hyperlipoproteinemia hyper lipidemia
  • dyslipidemia hypercholesterolemia
  • a heart attack a stroke
  • coronary heart disease atherosclerosis
  • peripheral vascular disease claudication (eg, claudication associated with elevated cholesterol)
  • type II diabetes high blood pressure
  • a cardiovascular disease or condition a cardiovascular disease or condition.
  • Another example is acute coronary syndrome.
  • the disease or condition is an inflammatory or autoimmune disease or condition.
  • a subject at risk of having or developing a disease or condition mediated by PCSK9 can be a subject exhibiting one or more signs or symptoms of such a disease or condition or having one or more risk factors for such a disease or condition, e.g. being overweight, having elevated cholesterol level, comprising one or more genetic polymorphisms known to predispose to the disease or condition, e.g., elevated cholesterol level, such as having a mutation in the LDLR (encoding low- density lipoprotein receptor) or APOB (encoding apolipoprotein B) or in the PCSK9 gene and/or having a family history of such a disease or condition.
  • LDLR encoding low- density lipoprotein receptor
  • APOB encoding apolipoprotein B
  • ligand refers to a molecule which can bind, e.g., specifically bind, to a second molecule or receptor.
  • a ligand can be, e.g., an antibody, antibody fragment, antibody portion, and/or affibody.
  • variant refers to a peptide or nucleic acid that differs from the polypeptide or nucleic acid (eg, the most common one in humans, eg, most frequent in a database as disclosed herein, such as the 1000 Genomes Project database) by one or more amino acid or nucleic acid deletions, additions, yet retains one or more specific functions or biological activities of the naturally occurring molecule.
  • Amino acid substitutions include alterations in which an amino acid is replaced with a different naturally-occurring amino acid residue. Such substitutions may be classified as "conservative", in which case an amino acid residue contained in a polypeptide is replaced with another naturally occurring amino acid of similar character either in relation to polarity, side chain functionality or size.
  • substitutions encompassed by the present invention may also be "non-conservative", in which an amino acid residue which is present in a peptide is substituted with an amino acid having different properties, such as naturally -occurring amino acid from a different group (e.g., substituting a charged or hydrophobic amino; acid with alanine), or alternatively, in which a naturally-occurring amino acid is substituted with a non- conventional amino acid.
  • amino acid substitutions are conservative.
  • polynucleotide or polypeptide refers to a polynucleotide or polypeptide that can vary in primary, secondary, or tertiary structure, as compared to a reference polynucleotide or polypeptide, respectively (e.g., as compared to a wild- type polynucleotide or polypeptide).
  • Variants of PCSK9 are provided elsewhere herein. Variants of PCSK9 can include the forms described herein as a, f, c, r, p, m, e h, aj, and q. Sequences of these variants are provided herein, see, e.g, SEQ ID NOs: l-27 and in Table 1, 2 or 6.
  • “Modified variants” can include conservative or non-conservative amino acid changes, as described below. Polynucleotide changes can result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence.
  • Some aspects use include insertion variants, deletion variants or substituted variants with substitutions of amino acids, including insertions and substitutions of amino acids and other molecules) that do not normally occur in the peptide sequence that is the basis of the variant, for example but not limited to insertion of ornithine which do not normally occur in human proteins.
  • the term "conservative substitution,” when describing a polypeptide, refers to a change in the amino acid composition of the polypeptide that does not substantially alter the polypeptide's activity. For example, a conservative substitution refers to substituting an amino acid residue for a different amino acid residue that has similar chemical properties. Conservative amino acid substitutions include replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
  • Constant amino acid substitutions result from replacing one amino acid with another having similar structural and/or chemical properties, such as the replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
  • “conservative substitution” of a particular amino acid sequence refers to substitution of those amino acids that are not critical for polypeptide activity or substitution of amino acids with other amino acids having similar properties (e.g., acidic, basic, positively or negatively charged, polar or non -polar, etc.) such that the substitution of even critical amino acids does not reduce the activity of the peptide, (i.e. the ability of the peptide to penetrate the blood brain barrier (BBB)).
  • BBB blood brain barrier
  • the following six groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W). (See also Creighton, Proteins, W. H.
  • substitutions suitable for amino acids on the exterior of a protein or peptide for example, but not limited to, the following substitutions can be used: substitution of Y with F, T with S or K, P with A, E with D or Q, N with D or G, R with K, G with N or A, T with S or K, D with N or E, I with L or V, F with Y, S with T or A, R with K, G with N or A, K with R, A with S, K or P.
  • an "antibody” refers to IgG, IgM, IgA, IgD or IgE molecules or antigen-specific antibody fragments thereof (including, but not limited to, a Fab, F(ab') 2 , Fv, disulphide linked Fv, scFv, single domain antibody, closed conformation multispecific antibody, disulphide -linked scfv, diabody), whether derived from any species that naturally produces an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria. Antibodies can be humanized using routine technology.
  • an "antigen” is a molecule that is bound by a binding site on an antibody agent.
  • antigens are bound by antibody ligands and are capable of raising an antibody response in vivo.
  • An antigen can be a polypeptide, protein, nucleic acid or other molecule or portion thereof.
  • antigenic determinant refers to an epitope on the antigen recognized by an antigen-binding molecule, and more particularly, by the antigen-binding site of said molecule.
  • an antibody fragment refers to a polypeptide that includes at least one immunoglobulin variable domain or immunoglobulin variable domain sequence and which specifically binds a given antigen.
  • An antibody fragment can comprise an antibody or a polypeptide comprising an antigen-binding domain of an antibody.
  • an antibody fragment can comprise a monoclonal antibody or a polypeptide comprising an antigen-binding domain of a monoclonal antibody.
  • an antibody can include a heavy (H) chain variable region (abbreviated herein as VH), and a light (L) chain variable region (abbreviated herein as VL).
  • an antibody in another example, includes two heavy (H) chain variable regions and two light (L) chain variable regions.
  • antibody fragment encompasses antigen-binding fragments of antibodies (e.g., single chain antibodies, Fab and sFab fragments, F(ab')2, Fd fragments, Fv fragments, scFv, and domain antibodies (dAb) fragments (see, e.g. de Wildt et al., Eur J. Immunol. 1996; 26(3):629-39; which is incorporated by reference herein in its entirety)) as well as complete antibodies.
  • An antibody can have the structural features of IgA, IgG, IgE, IgD, IgM (as well as subtypes and combinations thereof).
  • Antibodies can be from any source, including mouse, rabbit, pig, rat, and primate (human and non-human primate) and primatized antibodies. Antibodies also include midibodies, humanized antibodies, chimeric antibodies, and the like.
  • antibody variable domain refers to the portions of the light and heavy chains of antibody molecules that include amino acid sequences of Complementarity Determining
  • CDRs CDR1, CDR2, and CDR3
  • FRs Framework Regions
  • VH refers to the variable domain of the heavy chain.
  • VL refers to the variable domain of the light chain.
  • amino acid positions assigned to CDRs and FRs may be defined according to Kabat (Sequences of Proteins of Immunological Interest (National Institutes of
  • D domain or region refers to the diversity domain or region of an antibody chain.
  • J domain or region refers to the joining domain or region of an antibody chain.
  • An antibody “gene segment”, e.g. a VH gene segment, D gene segment, or JH gene segment refers to oligonucleotide having a nucleic acid sequence that encodes that portion of an antibody, e.g. a VH gene segment is an oligonucleotide comprising a nucleic acid sequence that encodes a polypeptide VH domain.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed “complementarity determining regions” ("CDR"), interspersed with regions that are more conserved, termed “framework regions” ("FR").
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • antigen-binding fragment or "antigen -binding domain”, which are used interchangeably herein are used to refer to one or more fragments of a full length antibody that retain the ability to specifically bind to a target of interest.
  • binding fragments encompassed within the term "antigen -binding fragment” of a full length antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment including two Fab fragments linked by a disulfide bridge at the hinge region; (iii) an Fd fragment consisting of the VH and CHI domains; (iv) an Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341 :544- 546; which is incorporated by reference herein in its entirety), which
  • the term "antibody binding site” refers to a polypeptide or domain that comprises one or more CDRs of an antibody and is capable of binding an antigen.
  • the polypeptide comprises a CDR3 (eg, HCDR3).
  • the polypeptide comprises CDRs 1 and 2 (eg, HCDR1 and 2) or CDRs 1-3 of a variable domain of an antibody (eg, HCDRsl-3).
  • the antibody binding site is provided by a single variable domain (eg, a VH or VL domain).
  • the binding site comprises a VH/VL pair or two or more of such pairs.
  • the term "specific binding” refers to a chemical interaction between two molecules, compounds, cells and/or particles wherein the first entity binds to the second, target entity with greater specificity and affinity than it binds to a third entity which is a non-target.
  • specific binding can refer to an affinity of the first entity for the second target entity which is at least 10 times, at least 50 times, at least 100 times, at least 500 times, at least 1000 times or greater than the affinity for the third nontarget entity.
  • specific binding can be "specific hybridization.”
  • a recombinant human(ized) antibody can be further optimized to decrease potential immunogenicity, while maintaining functional activity, for therapy in humans.
  • functional activity means a polypeptide capable of displaying one or more known functional activities associated with a recombinant antibody or antibody reagent thereof as described herein. Such functional activities include, e.g. the ability to bind to a target molecule.
  • immunizing refers to the step or steps of administering one or more antigens to an animal so that antibodies can be raised in the animal Generally, immunizing comprises injecting the antigen or antigens into the animal. Immunization can involve one or more administrations of the antigen or antigens. Suitable methods are prime -boost and RIMMS procedures as known to the skilled person in the art.
  • an "affibody” refers to a relatively small synthetic protein molecule that has high binding affinity for a target protein (e.g. for PCSK9 or a variant therefo).
  • Affibodies are composed of a three -helix bundle domain derived from the IgG-binding domain of staphylococcal protein A.
  • the protein domain consists of a 58 amino acid sequence, with 13 randomized amino acids affording a range of affibody variants. Despite being significantly smaller than an antibody
  • an affibody molecule works like an antibody since its binding site is approximately equivalent in surface area to the binding site of an antibody.
  • VH3-23*04 refers to a human VH domain variant comprising the polypeptide sequence of SEQ ID NO: 38.
  • VH3-23*04 has a valine residue instead of a leucine residue (see Figures 3 and 4; L24V, numbering including signal sequence; valine at position 5 shown in Figure 4) as a result of the presence of the rs56069819 SNP in the nucleic acid sequence encoding the VH domain.
  • rs56069819 refers to a mutation or variant in a VH gene segment from adenosine to cytosine (or thymine to guanine, depending upon the strand of DNA which is being read), resulting in the VH domain encoding VH3- 23*04.
  • Rs56069819 is depicted in Figure 4 and SEQ ID NO: 39, which demonstrate the T->G mutation (it is noted that the dbSNP entry for RS5606819 depicts the other strand, which comprises the A->C mutation). Further description of VH3-23*04 can be found, e.g., in US Patent Publication 2013/0071405; which is incorporated by reference herein in its entirety.
  • determine refers to ascertaining, e.g., by a quantitative or qualitative analysis.
  • has been determined can refer to ascertaining on the basis of previously obtained information or simultaneously obtained information.
  • selecting can include automation such as a computer implemented software program that upon input of the relevant data such as ethnicity or a panel of SNP data can make the determination based on the instructions set forth herein.
  • assaying refers to assessing, evaluating, quantifying, measuring, or characterizing an analyte, e.g., measuring the level of an analyte in a sample, identifying an analyte, or detecting the presence or absence of an analyte in a sample. In some embodiments, assaying refers to detecting a presence or absence of the analyte of interest. In some
  • assaying refers to quantifying an amount of an analyte, e.g., providing a measure of concentration or degree of analyte abundance. In some embodiments, assaying refers to enumerating the number of molecules of analyte present in a sample and/or specimen, e.g., to determine an analyte copy number.
  • multiplex refers to the carrying out of a method or process simultaneously and in the same reaction vessel on two or more, typically three or more, different target sequences, e.g. on two or more isoforms of PCSK9, or PCSK9 and an additional target.
  • a multiplex analysis typically includes analysis of 10-50; 10-100; 10-1000, 10-5000, 10-10000 reactions in a multiplex format, such as a multiwall, an array, or a multichannel reaction.
  • the analysis or multiplex analysis is also automated using robotics and typically software executed by a computer and may include a robotic handling of samples, automatic or robotic selection of positive or negative results, assaying for presence of absence of a target, such as a nucleic acid polymorphism or a protein variant.
  • a target such as a nucleic acid polymorphism or a protein variant.
  • biological sample or "test sample” as used herein denotes a sample taken or isolated from a biological organism, e.g., a sample from a subject.
  • exemplary biological samples include, but are not limited to, a biofluid sample; serum; plasma; urine; saliva; hair, epithelial cells, skin, a tumor biopsy and/or tissue sample etc.
  • the term also includes a mixture of the above-mentioned samples.
  • test sample or “biological sample” also includes untreated or pretreated (or pre-processed) biological samples.
  • the biological sample should typically comprise at least one cell comprising nucleic acids.
  • the test sample can be obtained by removing a sample of cells from a subject, but can also be accomplished by using previously isolated cells (e.g. isolated at a prior time point and isolated by the same or another person).
  • the test sample can be freshly collected or a previously collected, refrigerated, frozen or otherwise preserved sample.
  • the test sample can be an untreated test sample.
  • untreated test sample refers to a test sample that has not had any prior sample pre-treatment except for dilution and/or suspension in a solution.
  • Exemplary methods for treating a test sample include, but are not limited to, centrifugation, filtration, sonication, homogenization, heating, freezing and thawing, and combinations thereof.
  • the test sample can be a frozen test sample, e.g., a frozen tissue. The frozen sample can be thawed before employing methods, assays and systems described herein.
  • a frozen sample can be centrifuged before being subjected to methods, assays and systems described herein.
  • the test sample is a clarified test sample, for example, by centrifugation and collection of a supernatant comprising the clarified test sample.
  • a test sample can be a pre-processed test sample, for example, supernatant or filtrate resulting from a treatment selected from the group consisting of centrifugation, filtration, thawing, purification, and any combinations thereof.
  • the test sample can be treated with a chemical and/or biological reagent.
  • Chemical and/or biological reagents can be employed to protect and/or maintain the stability of the sample, including biomolecules (e.g., nucleic acid and protein) therein, during processing.
  • biomolecules e.g., nucleic acid and protein
  • One exemplary reagent is a protease inhibitor, which is generally used to protect or maintain the stability of protein during processing.
  • protease inhibitor which is generally used to protect or maintain the stability of protein during processing.
  • Genotyping refers to a process of determining the specific allelic composition of a cell and/or subject at one or more position within the genome, e.g. by determining the nucleic acid sequence at that position.
  • Genotyping refers to a nucleic acid analysis and/or analysis at the nucleic acid level.
  • phenotyping refers a process of determining the identity and/or composition of an expression product of a cell and/or subject, e.g. by determining the polypeptide sequence of an expression product.
  • Phenotyping refers to a protein analysis and/or analysis at the protein level.
  • nucleic acid amplification refers to the production of additional copies of a nucleic acid sequence and is typically carried out using polymerase chain reaction (PCR) or ligase chain reaction (LCR) technologies well known in the art (Dieffenbach, C. W. and G. S. Dveksler (1995) PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview, N. Y.). Other methods for amplification are also contemplated in aspects of the invention.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • allele-specific amplification refers to a reaction (e.g., PCR reaction) in which at least one of the primers (e.g., allele-specific primer) is chosen from a polymorphic area of gene (e.g., single nucleotide polymorphism), with the polymorphism located at or near the primer's 3'-end.
  • a mismatched primer will not initiate amplification, whereas a matched primer will initiate amplification.
  • the appearance of an amplification product is indicative of the presence of the polymorphism.
  • sequencing refers to the determination of the exact order of nucleotide bases in a strand of DNA (deoxyribonucleic acid) or RNA (ribonucleic acid) or the exact order of amino acids residues or peptides in a protein. Nucleic acid sequencing can be done using Sanger sequencing or next -generation high-throughput sequencing.
  • next-generation sequencing refers to oligonucleotide sequencing technologies that have the capacity to sequence oligonucleotides at speeds above those possible with conventional sequencing methods (e.g. Sanger sequencing), due to performing and reading out thousands to millions of sequencing reactions in parallel.
  • next-generation sequencing methods/platforms include Massively Parallel Signature Sequencing (Lynx
  • 454 pyro-sequencing (454 Life Sciences/ Roche Diagnostics); solid-phase, reversible dye -terminator sequencing (Solexa/Illumina): SOLiD technology (Applied Biosystems); Ion semiconductor sequencing (ION Torrent); DNA nanoball sequencing (Complete Genomics); and technologies available from Pacific Biosciences, Intelligen Bio-systems, Oxford Nanopore
  • nucleic acid hybridization refers to the pairing of complementary RNA and DNA strands as well as the pairing of complementary DNA single strands.
  • nucleic acid hybridization can refer to a method of determining a nucleic acid sequence and/or identity by hybridizing a nucleic acid sample with a probe, e.g. Northern or Southern blot analysis or microarray analysis.
  • the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease is reduced or halted.
  • treatment includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment). For treatment to be effective a complete cure is not contemplated.
  • the term "pharmaceutical composition” refers to the active agent in combination with a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • pharmaceutically acceptable is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • administering refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site.
  • Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
  • compositions can be administered separately or simultaneously. Separate administration refers to the two compositions being administered at different times, e.g. at least 10, 20, 30, or 10-60 minutes apart, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12 hours apart. One can also administer compositions at 24 hours apart, or even longer apart. Alternatively, two or more compositions can be administered simultaneously, e.g. less than 10 or less than 5 minutes apart. Compositions administered simultaneously can, in some aspects, be administered as a mixture, with or without similar or different time release mechanism for each of the components.
  • contacting refers to any suitable means for delivering, or exposing, an agent to at least one complex, enzyme, or cell.
  • exemplary delivery methods include, but are not limited to, direct delivery to cell culture medium, perfusion, injection, or other delivery method well known to one skilled in the art.
  • Obtaining a biological sample from a subject can comprise physical removing a sample from a subject (e.g. drawing blood or taking a hair or saliva sample) without or without active participation from the subject; receiving a sample from a subject (e.g. the subject collects a saliva or hair sample themselves and provides it, e.g. in a container provided for the purpose); or procuring a sample from a storage facility, medical facility, or medical provider.
  • Obtain from the human or subject refers to an active step of, e.g., drawing blood or taking a tissue or cell sample.
  • cholesterol level refers to a level of one or more of total cholesterol, LDL cholesterol, HDL cholesterol, and/or triglycerides. Cholesterol levels can be the level of cholesterol in the blood of a subject.
  • maintaining a particular level refers to a process that results in the cholesterol level not increasing by more than 10% over time. Maintaining may also refer to maintaining a previously achieved level. For example, if a human has received statin treatment, one can maintain the cholesterol level achieved using the statin treatment.
  • the subject treated according to the methods described herein has previously had their cholesterol level reduced.
  • "previously reduced” indicates that at a prior point in time, the subject experienced a decrease in cholesterol levels.
  • the decrease can be due to administration of a pharmaceutical composition (e.g. administration of a composition as described herein or another composition, e.g. a statin) or due to another cause, e.g. a change in diet and/or exercise.
  • An existing treatment for high cholesterol levels is the administration of a statin.
  • a "statin” also known as HMG-CoA reductase inhibitors
  • HMG-CoA reductase inhibitors are inhibitors of the enzyme HMG-coA reductase, which mediates cholesterol production in the liver.
  • Statins by competitively binding HMG-CoA reductase, prevent the binding of HMG-CoA to the enzyme and thereby inhibit the activity of the reductase (e.g. the production of mevalonate).
  • statins can include atorvastatin (LIPITORTM), fluvastatin (LESCOLTM), lovastatin (MEVACORTM, ALTOCORTM), pitavastatin (LIVALOTM), pravastatin (PRAVACHOLTM), rosuvastatin (CRESTORTM), and simvastatin (ZOCORTM).
  • Statins can be administered in combination with other agents, e.g. the combination of ezetimibe and simvastatin.
  • resistant to statin treatment or “reduced responsiveness to statin treatment” refers to a subject exhibiting a statistically significantly lower response to the administration of a statin as compared to a reference level.
  • the reference level can be, e.g., the average response for a population of subjects or the level of the individual subject at an earlier date.
  • a response to statin treatment is readily measured by one of skill in the art, e.g., measurement of cholesterol levels, changes in cholesterol levels, and/or HMG-CoA reductase activity.
  • detectable label refers to a molecule or moiety that can be detected, e.g. measured and/or determined to be present or absent.
  • Detectable labels can comprise, for example, a light-absorbing dye, a fluorescent dye, or a radioactive label.
  • Detectable labels, methods of detecting them, and methods of incorporating them into reagents are well known in the art.
  • detectable labels can include labels that can be detected by spectroscopic, photochemical, biochemical, immunochemical, electromagnetic, radiochemical, or chemical means, such as fluorescence, chemifluoresence, or chemiluminescence, or any other appropriate means.
  • the detectable labels used in the methods described herein can be
  • the detectable label can be linked by covalent or non-covalent means to the reagent.
  • a detectable label can be linked such as by directly labeling a molecule that achieves binding to the reagent via a ligand-receptor binding pair arrangement or other such specific recognition molecules.
  • Detectable labels can include, but are not limited to radioisotopes, bioluminescent compounds, chromophores, antibodies, chemiluminescent compounds, fluorescent compounds, metal chelates, and enzymes.
  • the detectable label can be a fluorescent compound.
  • a detectable label can be a fluorescent dye molecule, or fluorophore including, but not limited to fluorescein, phycoerythrin, phycocyanin, o-phthaldehyde, fluorescamine, Cy3TM, Cy5TM, allophycocyanine, Texas Red, peridenin chlorophyll, cyanine, tandem conjugates such as phycoerythrin-Cy5TM, green fluorescent protein, rhodamine, fluorescein isothiocyanate (FITC) and Oregon GreenTM, rhodamine and derivatives (e.g., Texas red and tetrarhodimine isothiocynate (TRITC)), biotin, phycoerythrin, AMCA, CyDyesTM, 6- carboxyfh
  • a detectable label can be a radiolabel including, but not limited to 3 H, 12 I, 3 "5S, 14 C, 3 J 2 i P, and 3 "3P.
  • a detectable label can be an enzyme including, but not limited to horseradish peroxidase and alkaline phosphatase.
  • An enzymatic label can produce, for example, a chemiluminescent signal, a color signal, or a fluorescent signal.
  • Enzymes contemplated for use as a detectable label can include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-V-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-VI-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • a detectable label is a chemiluminescent label, including, but not limited to lucigenin, luminol, luciferin, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a detectable label can be a spectral colorimetric label including, but not limited to colloidal gold or colored glass or plastic (e.g., polystyrene,
  • reagents can also be labeled with a detectable tag, such as c-Myc, HA, VSV-G, HSV, FLAG, V5, HIS, or biotin.
  • a detectable tag such as c-Myc, HA, VSV-G, HSV, FLAG, V5, HIS, or biotin.
  • Other detection systems can also be used, for example, a biotin-streptavidin system.
  • the antibodies immunoreactive (i. e. specific for) with the biomarker of interest is biotinylated. Quantity of biotinylated antibody bound to the biomarker is determined using a streptavidin-peroxidase conjugate and a chromagenic substrate.
  • streptavidin peroxidase detection kits are commercially available, e. g. from DAKO;
  • a reagent can also be detectably labeled using fluorescence emitting metals such as 152 Eu, or others of the lanthanide series. These metals can be attached to the reagent using such metal chelating groups as diethylenetriaminepentaacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
  • DTPA diethylenetriaminepentaacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • authorization number or "marketing authorization number” refers to a number issued by a regulatory agency upon that agency determining that a particular medical product and/or composition may be marketed and/or offered for sale in the area under the agency's jurisdiction.
  • regulatory agency refers to one of the agencies responsible for evaluating, e.g, the safety and efficacy of a medical product and/or composition and controlling the sales/marketing of such products and/or compositions in a given area.
  • the Food and Drug Administration (FDA) in the US and the European Medicines Agency (EPA) in Europe are but two examples of such regulatory agencies.
  • Other non-limiting examples can include SDA, MPA, MHPRA, IMA, ANMAT, Hong Kong Department of Health-Drug Office, CDSCO, Medsafe, and KFDA.
  • injection device refers to a device that is designed for carrying out injections, an injection including the steps of temporarily fluidically coupling the injection device to a person's tissue, typically the subcutaneous tissue. An injection further includes administering an amount of liquid drug into the tissue and decoupling or removing the injection device from the tissue.
  • an injection device can be an intravenous device or IV device, which is a type of injection device used when the target tissue is the blood within the circulatory system, e.g., the blood in a vein.
  • a common, but non-limiting example injection device is a needle and syringe.
  • a "buffer” refers to a chemical agent that is able to absorb a certain quantity of acid or base without undergoing a strong variation in pH.
  • Packaging refers to how the components are organized and/or restrained into a unit fit for distribution and/or use.
  • Packaging can include, e.g., boxes, bags, syringes, ampoules, vials, tubes, clamshell packaging, barriers and/or containers to maintain sterility, labeling, etc.
  • instructions refers to a display of written, printed or graphic matter on the immediate container of an article, for example the written material displayed on a vial containing a pharmaceutically active agent, or details on the composition and use of a product of interest included in a kit containing a composition of interest. Instructions set forth the method of the treatment as contemplated to be administered or performed.
  • a "solid surface” refers to an object suitable for the attachment of biomolecules.
  • Non-limiting examples of a solid surface can include a particle (including, but not limited to an agarose or latex bead or particle or a magnetic particle), a bead, a nanoparticle, a polymer, a substrate, a slide, a coverslip, a plate, a dish, a well, a membrane, and/or a grating.
  • the solid surface can include many different materials including, but not limited to, polymers, plastics, resins, polysaccharides, silicon or silica based materials, carbon, metals, inorganic glasses, and membranes.
  • classification of a subject refers to determining if the subject has biological ancestors who originated in a particular geographical area, and are therefore likely to have particular genetic variants found in the populations which have historically occupied that area.
  • Classification can comprise, e.g. obtaining information on the subject's family, interviewing the subject or a family member regarding their biological family's ancestry, and/or genetic testing. Classification can be on the basis used for the 1000 Genomes Project, as will be familiar to the skilled person in the art.
  • the subject can be classified as being of a particular ancestry if at least the subject's genome comprises a substantial number of different alleles in common with other humans of that ancestry (eg, determined by reference to the 1000 Genomes Project database), for example, at least 10, 20, 30, 40, 50 or 100 or more alleles in common.
  • Abbreviations for particular ancestral groups are provided in Table 4.
  • compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
  • the term “consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment.
  • compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
  • paragraph 1 provides :
  • a method of treating or preventing a disease or condition in a human, wherein the disease or condition is mediated by a Target of Interest (TOI), wherein the TOI is present in humans as different polymorphic variants comprising comprising administering to said human an anti-TOI ligand (eg, an antibody or antibody fragment) that specifically binds a TOI variant that is encoded by a nucleotide sequence comprising a SNP having a cumulative human allele frequency of less than 50% (eg, less than 40, 35, 30, 35, 20, 15, 10, 5, 4, 3, 2, or 1%) and/or wherein the TOI variant is encoded by a nucleotide sequence comprising a SNP having total human genotype frequency of less than 50% (eg, less than 40, 35, 30, 35, 20, 15, 10, 5, 4, 3, 2, or 1%); wherein the human comprises a (eg, said) TOI-encoding nucleotide sequence comprising said SNP; wherein
  • an anti-TOI ligand eg, an antibody or antibody
  • the ligand (eg, antibody or fragment) comprises a VH domain derived from the recombination of a human VH segment, a human D gene segment and a human JH segment, the human VH segment comprising a SNP, and wherein the human comprises a (eg, said) VH segment comprising said SNP; or
  • the ligand (eg, antibody or fragment) comprises a ⁇ domain derived from the recombination of a human ⁇ segment and a human ⁇ segment, the human ⁇ segment comprising a SNP, and wherein the human comprises a (eg, said) ⁇ segment comprising said SNP; or
  • the ligand (eg, antibody or fragment) comprises a VK domain derived from the recombination of a human VK segment and a human JK segment, the human VK segment comprising a SNP, and wherein the human comprises a (eg, said) VK segment comprising said SNP; or
  • the ligand eg, antibody or fragment
  • the ligand comprises a heavy chain constant region (eg, a gamma- 1, 2, 3 o 4 constant region) encoded by a nucleotide sequence comprising a SNP
  • the human comprises a (eg, said) heavy chain constant region (eg, a gamma- 1, 2, 3 o 4 constant region respectively) comprising said SNP;
  • the ligand (eg, antibody or fragment) comprises a lambda chain constant encoded by a nucleotide sequence comprising a SNP, and wherein the human comprises a (eg, said) lambda chain constant region comprising said SNP; or
  • the ligand (eg, antibody or fragment) comprises a kappa chain constant encoded by a nucleotide sequence comprising a SNP, and wherein the human comprises a (eg, said) kappa chain constant region comprising said SNP.
  • paragraph 1 provides: -
  • a method of treating or preventing a disease or condition in a human, wherein the disease or condition is mediated by a Target of Interest (TOI), wherein the TOI is present in humans as different polymorphic variants comprising comprising administering to said human an anti-TOI ligand (eg, an antibody or antibody fragment) that specifically binds a TOI variant that comprises a single amino acid variation (ie, a single amino acid at a position where there is difference between TOI variants in humans) having a cumulative human allele frequency of less than 50% (eg, less than 40, 35, 30, 35, 20, 15, 10, 5, 4, 3, 2, or 1%) and/or wherein the amino acid variant has a total human genotype frequency of less than 50% (eg, less than 40, 35, 30, 35, 20, 15, 10, 5, 4, 3, 2, or 1%);
  • an anti-TOI ligand eg, an antibody or antibody fragment
  • the human expresses TOI proteins comprising said single amino acid variation;
  • the ligand eg, antibody or fragment
  • the ligand comprises a VH domain comprising the same single amino acid variation, and wherein the human expresses VH domains comprising said single amino acid variation
  • the ligand eg, antibody or fragment
  • the ligand comprises a domain comprising a single amino acid variation, and wherein the human expresses domains comprising the same single amino acid variation
  • the ligand eg, antibody or fragment
  • the ligand comprises a VK domain comprising a single amino acid variation, and wherein the human expresses VK domains comprising the same single amino acid variation
  • the ligand eg, antibody or fragment
  • the ligand comprises a heavy chain constant region (eg, a gamma- 1, 2, 3 o 4 constant region, eg, wherein the constant region is an Fc, CHI, CH2 or CH3) comprising a single amino acid variation
  • the human expresses a heavy chain constant region (eg, a gamma- 1, 2, 3 o 4 constant region, eg, wherein the constant region is an Fc, CHI, CH2 or CH3 respectively) comprising the same single amino acid variation
  • the ligand eg, antibody or fragment
  • the ligand comprises a lambda chain constant region comprising a single amino acid variation, and wherein the human expresses a lambda chain constant region comprising the same single amino acid variation
  • the ligand eg, antibody or fragment
  • the ligand comprises a kappa chain constant region comprising a single amino acid variation
  • the human expresses a kappa chain constant region comprising the same single amino acid variation
  • frequencies herein are according to the 1000 Genomes database, eg, the Phase I database or as shown in Ensembl (available on the world wide web at ensembl.org), eg, at 1 ST October 2013 or 1 ST October 2014.
  • each SNP encodes a non-synonymous amino acid variation, ie, the SNP is not a silent mutation.
  • the method comprises selecting said human comprising the nucleotide sequence prior to administration.
  • paragraph 1 provides:- A method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, the method comprising administering to said human an antibody or antibody fragment that specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation as defined herein (eg, I474V or E670G) in SEQ ID NO: 1, wherein the antibody comprises a VH domain derived from the recombination of a human VH segment, a human D gene segment and a human JH segment, the human VH segment encoding a valine at the amino acid corresponding to position 5 of SEQ ID NO: 40 and wherein said human comprises (i) a VH gene segment encoding the framework 1 of SEQ ID NO: 40 and (ii) a nucleotide sequence encoding a proprotein convertase subtilisin/kexin type 9 (PCSK9) that
  • paragraph 1 provides:- A method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, the method comprising administering to said human an antibody or antibody fragment that specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation as defined herein (eg, I474V or E670G) in SEQ ID NO: 1, wherein the antibody comprises a VL domain derived from the recombination of a human VL segment and a human JL segment, the human VL segment is a or VK disclosed herein
  • PCSK9 proprotein convertase subtilisin/kexin type 9
  • said human comprises (i) said VL gene segment and (ii) a nucleotide sequence encoding a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation in SEQ ID NO: 1.
  • PCSK9 proprotein convertase subtilisin/kexin type 9
  • paragraph 1 provides:- A method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, the method comprising administering to said human an antibody or antibody fragment that specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation as defined herein (eg, I474V or E670G) in SEQ ID NO: 1, wherein the antibody comprises a C domain encoded by a human CH, Ck or CK gene segment disclosed herein and wherein said human comprises (i) said C gene segment and (ii) a nucleotide sequence encoding a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation in SEQ ID NO: 1.
  • PCSK9 proprotein convertase subtilisin/kexin type 9
  • step (a) the ligand has been or is determined as being capable of specifically binding to said TOI variant.
  • step of determining comprises assaying a biological sample obtained from said human for a nucleotide polymorphism encoding said TOI polymorphic variant.
  • step of determining comprises assaying a biological sample obtained from said human for a protein corresponding to the TOI polymorphic variant.
  • the ligand is capable of specifically binding to two or more different TOI variants, each being encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the TOI in said human is a variant encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of more than 50% and/or having a total human genotype frequency of more than 50%.
  • determining comprises that the human has been or is phenotyped as positive for the most frequent TOI variant or genotyped for the nucleotide sequence thereof.
  • determining comprises assaying for the nucleotide sequence to determine the presence of said allele.
  • the ligand is capable of specifically binding to two or more different TOI variants, each being encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of more than 50%.
  • said cumulative human allele frequency is the frequency in a database of naturally-occurring sequences sampled from at least 15 different human ethnic populations and comprising at least 1000 sequences.
  • the ligand is an antibody, antibody fragment or an affibody.
  • the ligand comprises a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the ligand comprises a nucleotide sequence that comprises at least 10 contiguous nucleotides of a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • the genome of said human comprises an allele having a cumulative human allele frequency of less than 50% and the allele is found in at least 2 different ethnic populations.
  • a composition comprising a ligand capable of binding a target of interest encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and the allele is found in at least 2 different ethnic populations and optionally a pharmaceutically acceptable carrier and optionally a label or instructions for use to treat and/or prevent said disease or condition in a human; optionally wherein the label or instructions comprise a marketing authorisation number issued by a regulatory authority.
  • kits for treating or preventing a condition or disease mediated by a target of interest comprising a ligand capable of specifically binding a target of interest encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and the allele is found in at least 2 different ethnic populations ; and optionally in combination with a label or instructions for use to treat and/or prevent said disease or condition in a human; optionally wherein the label or instructions comprise a marketing authorisation number issued by a regulatory agency; optionally wherein the kit comprises an injection pen or IV container that comprises the ligand.
  • a method of producing an anti-human TOI antibody binding site comprising obtaining a plurality of anti-TOI antibody binding sites, screening the antibody binding sites for binding to a TOI comprising an amino acid sequence encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence comprising an allele having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody binding site that binds in the screening step.
  • a method of producing an anti -human TOI antibody comprising immunising a non- human vertebrate with a TOI comprising an amino acid sequence encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI-encoding nucleotide sequence comprising an allele having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody that binds a TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, and optionally producing a TOI -binding fragment or derivative of the isolated antibody.
  • non-human vertebrate is a mouse or a rat.
  • kits for TOI genotyping a human comprising a nucleic acid comprising a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the nucleic acid comprises a nucleotide sequence that comprises at least 10 contiguous nucleotides of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • kits for TOI genotyping or phenotyping a human comprising a ligand capable of binding a target of interest encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% or an antibody, fragment or derivative produced by the method of any one of paragraphs 28 to 30.
  • kits of paragraph 32 wherein the allele is found in at least 2 different ethnic populations.
  • an anti-TOI ligand that specifically binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for targeting said TOI in a human to treat and/or prevent a disease or condition mediated by TOI.
  • a method of targeting a Target of Interest (TOI) for treating and/or preventing a TOI-mediated disease or condition in a human comprising administering an anti-TOI ligand to a human comprising a TOI nucleotide sequence comprising an allele selected as having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, whereby a TOI encoded by said nucleotide sequence is targeted.
  • TOI Target of Interest
  • the method comprises targeting a human TOI comprising an amino acid sequence with said ligand to treat and/or prevent said disease or condition in said human, wherein said amino acid sequence is encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%.
  • a method of Target of Interest (TOI) genotyping a nucleic acid sample of a human comprising assaying in the sample the presence of a TOI nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • a method of Target of Interest (TOI) typing a protein sample of a human the method comprising assaying the sample the presence of a TOI amino acid sequence encoded by a TOI nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the method of paragraph 38 or 39 comprising obtaining a sample of serum, blood, faeces, hair, urine or saliva from a human, whereby the nucleic acid or protein sample is obtained for use in the step of assaying said sequence.
  • a diagnostic kit comprising a ligand that is capable of binding a human Target of Interest (TOI) comprising an amino acid sequence encoded by a TOI nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% and instructions for carrying out the method of any one of paragraphs 38-41.
  • TOI human Target of Interest
  • the diagnostic kit wherein the ligand is selected from an antibody, antibody fragment, antibody portion, affybody, oligonucleotide, modified oligonucleotide, antisense oligonucleotide, siRNA, and microRNA.
  • a diagnostic kit comprising a nucleic acid probe comprising a nucleotide sequence that specifically hybridises a Target of Interest (TOI) nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof and instructions for carrying out the method of paragraph 38 or 39.
  • TOI Target of Interest
  • the TOI is encoded by a nucleotide sequence having a cumulative human allele frequency from 1 to 10% and/or a total human genotype frequency from 1 to about 15% or from 1 to 15%.
  • the TOI is a human TOI selected from Table 5 ; optionally for treating and/or preventing a corresponding disease or condition as set out in Table 5.
  • a method of treating or preventing a disease or condition in a human, wherein the disease or condition is mediated by a Target of Interest (TOI), wherein the TOI is present in humans as different polymorphic variants comprising administering to the human determined to be positive for the TOI polymorphic variant, wherein the TOI in said human is encoded by a nucleotide sequence comprising an allele having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence comprising an allele having total human genotype frequency of less than 50% an anti-TOI ligand to target the TOI in the human to treat or prevent said disease or condition.
  • TOI Target of Interest
  • an antibody portion an antibody fragment, an affibody, an antisense oligonucleotide, an siRNA, and a microRNA.
  • administration to a human or is comprised by an injectable preparation.
  • the present invention contemplates ligands (eg, antibodies and fragments) whose binding site specificities have been matched to one or more variant human TOIs (eg, PCSK9 or IL6R). Additionally or alternatively (and as further illustrated in the non- limiting Examples below), an optional aspect of the invention provides for matching of other features of the ligand to the patient's genotype or phenotype. In this respect, for example, the invention includes the ability to match amino acid sequence variation in a human patient to one or more ligand sequences or domains outside of the binding sites.
  • the ligand comprises or consists of a human TOI-binding antibody or an anti-human TOI receptor Fc fusion
  • this aspect of the invention provides for more tailored matching of one or more constant region domains (eg, the Fc) to the patient genotype or phenotype.
  • sequence variation in the binding site can be similarly matched to the patient's genotype or phenotype. The present inventor has done this by considering the SNP occurrences in sequences encoding one or more parts of the ligand, eg, SNP occurrences in one, more or all of the gene segments from which the variable domain(s) and/or constant region domain(s) are derived.
  • Matching could involve designing the ligand specifically for a patient of known phenotype and/or genotype, or matching could involve choosing a ligand by determining that there is correspondence between variation in the patient's phenotype or genotype with the variation in the ligand amino acid and/or corresponding nucleotides.
  • a key consideration for the inventor was the desire to promote compatibility of the ligand with the patient's body, and in particular, the possible patient immune system responses to administered ligands.
  • human patients receiving human or humanised antibody drugs may mount an immune response against the incoming antibody (a so-called HAHA response) which results in the patient producing anti-drug antibodies as a result of the patient's immune system recognising the drug as foreign.
  • HUMIRATM adalimumab
  • HUMIRATM adalimumab
  • the present aspect of the invention by more closely tailoring the ligand itself (as well as its specificity) to the patient, helps to address these considerations when designing and administering anti-human TOI medicines for treating and/or preventing human TOI -related diseases and conditions.
  • the inventor also considered the desirability to tailor the variation in the ligand constant region (eg, for an antibody or Fc-containing ligand), mindful that then the constant region being administered to the patient would be tuned to the various components, such as patient's Fc receptors, that would interact with the constant region in the patient.
  • Good Fc/Fc receptor interactions can be important for drug recycling (via the FcRn) to provide for useful half -lives in vivo or for use in cell killing, eg, for cancer indications.
  • FcRn drug recycling
  • the effector function of the constant region (eg, Fc) to the patient more closely, to promote efficacy.
  • more efficacious drugs are desirable for better patient treatment and may provide the possibility of lowered dosing and/or dosing frequencies.
  • the ligand eg, antibody or fragment
  • variable domain that is encoded by a human V region nucleotide sequence, wherein the V nucleotide sequence is derived from recombination of human VH, D and JH gene segments or human VL and JL gene segments; or
  • a first gene segment of said gene segments of (a), or said C region gene segment of (b) comprises a first single nucleotide polymorphism (SNP) encoding a first amino acid polymorphism
  • the genome of said human comprises said first SNP or wherein said human expresses (a') an antibody variable domain comprising said first amino acid polymorphism or (b') an antibody constant domain comprising said first amino acid polymorphism.
  • ELISA is used.
  • segment or a second segment of said segments of (a), or said C region gene segment of (b), comprises a second SNP encoding a second amino acid polymorphism; and wherein the genome of said human comprises said second SNP or wherein said human expresses (a") an antibody variable domain comprising said second amino acid polymorphism or (b") an antibody constant region domain comprising said first and second amino acid polymorphisms.
  • antibody variable domain comprising said first and second amino acid polymorphisms.
  • the first and second SNPs of the genome are comprised by an IGHG1*01 gene segment and said first segment of (a) is an IGHG1*01 gene segment.
  • the first and second SNPs of the genome are comprised by an IGHG2*01 gene segment and said first segment of (a) is an IGHG2*01 gene segment.
  • each SNP is a constant region gene segment SNP, eg each SNP is a gamma- 1 constant region gene segment SNP, or a gamma-2 constant region gene segment SNP, or a gamma-3 constant region gene segment SNP or a gamma-4 constant region gene segment SNP.
  • each SNP is a variable domain SNP, eg, a VH domain SNP, or a VK domain SNP, or a ⁇ SNP.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 13), wherein the ligand comprises or consists of an antibody or fragment that comprises a human antibody variable domain derived from the recombination of a human V gene segment and a human J gene segment (and optionally a human D gene segment when the variable domains are VH domains); and wherein the genome of the human comprises said human V gene segment and/or the human expresses antibodies comprising antibody variable domains derived from the recombination of said human V gene segment and a human J gene segment (and optionally a human D gene segment).
  • the V gene segment is any of the V gene segments disclosed in WO2013041844, a 1000 Genomes database and/or www.imgt.org, the disclosures of which (including disclosure relating to sequence) is explicitly incorporated herein by reference for use in the present invention.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 14), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor) comprises a human heavy chain constant domain encoded by a first constant region nucleotide sequence; and wherein the genome of the human comprises a heavy chain constant region nucleotide sequence that is identical to said first constant region nucleotide sequence and/or the human expresses antibodies comprising said human constant domain.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor
  • the genome of the human comprises a heavy chain constant region nucleotide sequence that is identical to said first constant region nucleotide sequence and/or the human expresses antibodies comprising said human constant domain.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 15), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor) comprises a human gamma heavy chain CHI domain encoded by a CHI nucleotide sequence; and wherein the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CHI nucleotide sequence and/or the human expresses antibodies comprising said human gamma CHI domain.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CHI nucleotide sequence and/or the human expresses antibodies comprising said human gamma CHI domain.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 16), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor) comprises a human gamma heavy chain CH2 domain encoded by a CH2 nucleotide sequence; and wherein the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH2 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH2 domain.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH2 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH2 domain.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 17), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor) comprises a human gamma heavy chain CH3 domain encoded by a CH3 nucleotide sequence; and wherein the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH3 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH3 domain.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH3 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH3 domain.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 18), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor) comprises a human gamma heavy chain CH4 domain encoded by a CH4 nucleotide sequence; and wherein the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH4 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH4 domain.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH4 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH4 domain.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 19), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor) comprises a human gamma heavy chain Fc region encoded by a Fc nucleotide sequence; and wherein the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said Fc nucleotide sequence and/or the human expresses antibodies comprising said human gamma Fc region.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said Fc nucleotide sequence and/or the human expresses antibodies comprising said human gamma Fc region.
  • ligand comprises a human IGHG1 *01 gamma-1 heavy chain constant region.
  • ligand comprises a human IGHG2*01 gamma-1 heavy chain constant region.
  • human as positive for said gamma heavy chain constant region, eg, positive for said gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc; positive for said human IGHG1*01 gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc; or positive for said human IGHG2*01 gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc.
  • IGH-gamma-1 SNPs 204D observed cumulative frequency of 0.296
  • 206L observed cumulative frequency of 0.283 individually or in combination. These residues are part of the CH3 domain, and as such they form part of antibody Fc regions. Thus, matching of these CH3 variations with the patient is especially beneficial for reasons as discussed above.
  • this example provides aspects set out in the following clauses.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 31), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused TOI receptor) comprises a human gamma- 1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 42 or a Leu corresponding to position 206 of SEQ ID NO: 42 and wherein the genome of the human comprises a gamma-1 heavy chain constant region nucleotide sequence that encodes such an Asp or Leu or the human expresses antibodies comprising human gamma-1 constant regions comprising such an Asp or Leu.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused TOI receptor
  • the genome of the human comprises a gamma-1 heavy chain constant region nucleotide sequence that encodes such an Asp or Leu or the human expresses antibodies comprising human gamma-1
  • the skilled person will be familiar with techniques for determining genome sequences of a human, eg, by using a sample containing genomic DNA and/or RNA, sequencing and comparing using bioinformatics or other computer tools to compare the sampled sequence with sequences of human alleles (eg, as shown in the IMGT, 100 genomes or other database as disclosed herein).
  • the sample is a blood or saliva or cheek swab sample.
  • ligand comprises a human IGHG1*01 gamma-1 heavy chain constant region, eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG1*01.
  • a human IGHG1*01 gamma-1 heavy chain constant region eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG1*01.
  • ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 61.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 31), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused TOI receptor) comprises a human gamma-2 heavy chain constant region that comprises an amino acid selected from the group consisting of a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44, a Phe corresponding to position 76 of SEQ ID NO: 44, a Val corresponding to position 161 of SEQ ID NO: 44 and an Ala corresponding to position 257 of SEQ ID NO: 44; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such a selected amino acid or the human expresses antibodies comprising human gamma-2 constant regions comprising such a selected amino acid.
  • the genome of the human comprises a gamma-2 heavy chain constant region nucleot
  • the ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44, a Phe corresponding to position 76 of SEQ ID NO: 44 and optionally (ii) a Val corresponding to position 161 of SEQ ID NO: 44 and/or an Ala corresponding to position 257 of SEQ ID NO: 44; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such amino acids of (i) or the human expresses antibodies comprising human gamma-2 constant regions comprising such amino acids of (i).
  • This example focuses on CHI variation.
  • ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Val corresponding to position 161 of SEQ ID NO: 44 and an Ala corresponding to position 257 of SEQ ID NO: 44 and optionally (ii) an amino acid selected from the group consisting of a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44 and a Phe corresponding to position 76 of SEQ ID NO: 44; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such amino acids of (i) or the human expresses antibodies comprising human gamma-2 constant regions comprising such amino acids of (i).
  • This example focuses on Fc variation.
  • ligand comprises a human IGHG2*01 gamma-2 heavy chain constant region, eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG2*01.
  • a human IGHG2*01 gamma-2 heavy chain constant region eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG2*01.
  • ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 63 or 65.
  • any one of clauses 45 to 58 comprising selecting a said human whose phenotype comprises one, more or all of said Pro, Asn, Phe, Val and Ala; a human IGHG2*01 region; or a human IGHG2*01 CHI, CH2 and/or CH3.
  • the present aspect of the invention also provides the following.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 60), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused TOI receptor) comprises a human kappa light chain constant region that comprises a Val corresponding to position 84 of SEQ ID NO: 50 or a Cys corresponding to position 87 of SEQ ID NO: 50; and wherein the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val or Cys or the human expresses antibodies comprising human kappa light chain constant regions comprising such a Val or Cys.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc- fused TOI receptor
  • the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val or Cys or the human expresses antibodies comprising human kappa light chain constant
  • ligand comprises a human kappa light chain constant region that comprises a Val corresponding to position 84 of SEQ ID NO: 50 and a Cys corresponding to position 87 of SEQ ID NO: 50.
  • ligand comprises or consists of an antibody, wherein the antibody comprises a light chain variable domain derived from recombination of a human VK gene segment and a human JK gene segment, wherein the JK gene segment is IGKJ2*01 (SEQ ID NO: 57).
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 60), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc- fused TOI receptor) comprises a human IGLC2*01 light chain constant region; and wherein the genome of the human comprises a human IGLC2*01 nucleotide sequence or the human expresses antibodies comprising human light chain IGLC2*01 constant regions.
  • the ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 79), wherein the ligand comprises or consists of an antibody or fragment, wherein the antibody or fragment comprises a VH domain that is derived from the recombination of a human VH gene segment, a human D gene segment and a human JH gene segment, wherein the VH gene segment is selected from the group consisting of (i) IGHV 1-18*01 and the genome of the human comprises a human IGHV1-18*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV1 -18*01 ; or (ii) IGVH1 -46*01 and the genome of the human comprises a human IGHV1 -46*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV1-46*01. 81.
  • the ligand comprises or consists of an antibody or fragment, wherein the antibody or fragment comprises a VL domain that is derived from the recombination of a human VL gene segment and a human JL gene segment, wherein the VL gene segment is selected from the group consisting of (i) IGKV4-1*01 and the genome of the human comprises a human IGKV4-1*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV4-1*01 ; (ii) IGLV2- 14*01 and the genome of the human comprises a human IGLV2- 14*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGLV2- 14*01 ; or (iii) IGKV1-13*02 and the genome
  • clause 88 comprising genotyping the human as positive for said selected VL gene segment, eg, genotyping the human as positive for human IGKV4-1*01, IGLV2- 14*01 or IGKV1 -13*02. 90.
  • Kd dissociation constant
  • the ligand, antibody or fragment of present invention comprises an Fc region, wherein the Fc region comprises at least one non-native amino acid residue selected from the group consisting of 234D, 234E, 234N, 234Q, 234T, 234H, 234Y, 2341, 234V, 234F, 235A, 235D, 235R, 235W, 235P, 235S, 235N, 235Q, 235T, 235H, 235Y, 2351, 235V, 235F, 236E, 239D, 239E, 239N, 239Q, 239F, 239T, 239H, 239Y, 2401, 240A, 240T, 240M, 241W, 241 L, 241 Y, 241E, 241 R.
  • the Fc region comprises at least one non-native amino acid residue selected from the group consisting of 234D, 234E, 234N, 234Q, 234T, 234H
  • the Fc region may comprise additional and/or alternative non-native amino acid residues known to one skilled in the art (see, e.g., U.S. Patents 5,624,821 ; 6,277,375 ; 6,737,056 ; PCT Patent
  • the ligand, antibody or fragment comprises one or more human binding sites that specifically bind the TOI (eg, PCSK9, IL4Ra, IL6R, VEGFA or Navl.7).
  • the binding sites comprise or consist of human antibody variable domains or human receptors for the TOI (eg, Human receptor binding sites for VEGFA).
  • the ligand, antibody or fragment can be optionally fully human (eg, comprising human constant regions, eg, human Fc regions with or without human CL regions).
  • the ligand is aflibercept, alirocumab, sarilumab or dupilumab. Fully human ligands maximise compatibility with the human patient when used in the context of the invention wherein the V and/or C regions are tailored to the human genotype and/or phenotype as per the description herein.
  • step (a) said human has been or is genotyped as positive for said nucleotide
  • step (a) the ligand has been or is determined as being capable of binding to said TOI variant.
  • step (a) the ligand has been or is determined as capable of binding to said TOI variant.
  • step (a) said nucleotide sequence has been or is determined as having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the ligand is capable of binding to two or more different TOI variants, each being encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • step (a) said human has been or is genotyped as negative for a variant nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50% ; or phenotyped as negative for a TOI variant encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • step (a) the human has been or is phenotyped as positive for the most frequent TOI variant or genotyped for the nucleotide sequence thereof.
  • step (a) the ligand has been or is determined as being capable of binding to the most frequent TOI variant.
  • step (b) The method of paragraph 9, 10 or 11, wherein before step (a) the ligand has been or is determined as being substantially incapable of neutralising or inhibiting said TOI variant recited in step (b).
  • step (b) The method of any one of paragraphs 9 to 12, wherein the ligand is capable of binding to the most frequent TOI variant.
  • step (a) has been or is determined as being present in at least 2 different human ethnic populations.
  • said human frequency is the frequency in a database of naturally-occurring sequences sampled from at least 15 different human ethnic populations and comprising at least 1000 sequences.
  • An anti-human TOI ligand for use in a method of treating and/or preventing a TOI-mediated disease or condition in a human, wherein the TOI is present in humans as different polymorphic variants and wherein the genome of said human comprises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, the method comprising administering the ligand to the human.
  • a ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, for use in a method comprising the step of using the ligand to target said TOI in a human to treat and/or prevent a disease or condition mediated by TOI, the method comprising administering the ligand to the human.
  • a pharmaceutical composition or kit for treating or preventing a condition or disease mediated by a TOI as recited in any preceding paragraph the composition or kit comprising a ligand of any one of paragraphs 17 to 27; and optionally in combination with a label or instructions for use to treat and/or prevent said disease or condition in a human; optionally wherein the label or instructions comprise a marketing authorisation number (eg, an FDA or EMA authorisation number); optionally wherein the kit comprises an injection pen or IV container that comprises the ligand.
  • a marketing authorisation number eg, an FDA or EMA authorisation number
  • a method of producing an anti-human TOI antibody binding site comprising obtaining a plurality of anti-TOI antibody binding sites, screening the antibody binding sites for binding to a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI -encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody binding site that binds in the screening step.
  • a method of producing an anti -human TOI antibody comprising immunising a non- human vertebrate (eg, a mouse or a rat) with a TOI comprising an amino acid sequence encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, or to a peptide thereof that comprises an amino acid variation from the corresponding sequence encoded by the TOI -encoding nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency, and isolating an antibody that binds a TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, and optionally producing a TOI -binding fragment or derivative of the isolated antibody.
  • a non- human vertebrate eg, a mouse or a rat
  • a TOI comprising an
  • the method of paragraph 29 or 30, comprising the step of obtaining a nucleic acid encoding the antibody, fragment, derivative or binding site and optionally inserting the nucleic acid in an expression vector.
  • kits for TOI genotyping a human comprising a nucleic acid comprising a nucleotide sequence that specifically hybridises to a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof; and/or the nucleic acid comprises a nucleotide sequence that comprises at least 10 contiguous nucleotides of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or is an antisense sequence thereof.
  • kits for TOI genotyping or phenotyping a human wherein the kit comprises a ligand according to any one of paragraphs 17 to 27 or an antibody, fragment or derivative produced by the method of any one of paragraphs 29 to 31.
  • an anti-TOI ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for treating and/or preventing a TOI-mediated disease or condition in a human whose genome comprises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • an anti-TOI ligand that binds a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, in the manufacture of a medicament for targeting said TOI in a human to treat and/or prevent a disease or condition mediated by TOI.
  • a method of targeting a TOI for treating and/or preventing a TOI-mediated disease or condition in a human comprising administering an anti-TOI ligand to a human comprising a TOI nucleotide sequence selected having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%, whereby a TOI encoded by said nucleotide sequence is targeted.
  • the method comprises targeting a human TOI comprising an amino acid sequence with said ligand to treat and/or prevent said disease or condition in said human, wherein said amino acid sequence is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%.
  • a method of TOI genotyping a nucleic acid sample of a human comprising identifying in the sample the presence of a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • a method of TOI typing a protein sample of a human comprising identifying in the sample the presence of a TOI amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%.
  • the method of paragraph 38 or 39 comprising obtaining a sample of serum, blood, faeces, hair, urine or saliva from a human, whereby the nucleic acid or protein sample is obtained for use in the step of identifying said sequence.
  • the method of any one of paragraphs 38 to 40 comprising using a ligand according to any one of paragraphs 17 to 27 to carry out said identifying step.
  • a diagnostic kit comprising a ligand that is capable of binding a human TOI comprising an amino acid sequence encoded by a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% and instructions for carrying out the method of paragraph 38 or 39.
  • a diagnostic kit comprising a nucleic acid probe comprising a nucleotide sequence that specifically hybridises a TOI nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50% or an RNA transcript thereof and instructions for carrying out the method of paragraph 38 or 39.
  • the TOI is encoded by a nucleotide sequence having a cumulative human allele frequency from 1 to 10% and/or a total human genotype frequency from 1 to about 15% or from 1 to 15%.
  • the TOI is a human TOI selected from Table 5; optionally for treating and/or preventing a corresponding disease or condition as set out in table 5.
  • the ligand eg, antibody or fragment
  • variable domain that is encoded by a human V region nucleotide sequence, wherein the V nucleotide sequence is derived from recombination of human VH, D and JH gene segments or human VL and JL gene segments; or
  • a first gene segment of said gene segments of (a), or said C region gene segment of (b) comprises a first single nucleotide polymorphism (SNP) encoding a first amino acid polymorphism
  • the genome of said human comprises said first SNP or wherein said human expresses (a') an antibody variable domain comprising said first amino acid polymorphism or (b') an antibody constant domain comprising said first amino acid polymorphism.
  • blood of said human comprises substantially no antibodies that specifically bind to the domain comprising said first amino acid polymorphism as determined in an in vitro binding assay.
  • the ligand, method, use, kit or composition of any one of paragraphs 46 to 48 wherein the genome of said human comprises said first gene segment (when (a) applies) or said C region gene segment (when (b) applies).
  • the ligand, method, use, kit or composition of any one of paragraphs 46 to 49 wherein said first segment or a second segment of said segments of (a), or said C region gene segment of (b), comprises a second SNP encoding a second amino acid polymorphism; and wherein the genome of said human comprises said second SNP or wherein said human expresses (a") an antibody variable domain comprising said second amino acid polymorphism or (b") an antibody constant region domain comprising said first and second amino acid polymorphisms.
  • each SNP is a variable region gene segment SNP.
  • each SNP is a constant region gene segment SNP, eg each SNP is a gamma- 1 constant region gene segment SNP, or a gamma-2 constant region gene segment SNP, or a gamma-3 constant region gene segment SNP or a gamma-4 constant region gene segment SNP.
  • the ligand, method, use, kit or composition of paragraph 54 wherein the first SNP is a CHI, CH2, CH3 or CH4 gene segment SNP and/or the second SNP is a CHI, CH2, CH3 or CH4 gene segment SNP.
  • each SNP is a variable domain SNP, eg, a VH domain SNP, or a VK domain SNP, or a ⁇ SNP.
  • ligand eg, antibody or fragment
  • ligand has been determined to specifically bind one or more human TOI variants as disclosed herein, for example, with a KD of InM or less (eg, 100 or ⁇ or less) as determined by SPR.
  • any preceding paragraph eg, according to any one of paragraphs 46 to 58
  • the ligand comprises or consists of an antibody or fragment that comprises a human antibody variable domain derived from the recombination of a human V gene segment and a human J gene segment (and optionally a human D gene segment when the variable domains are VH domains); and wherein the genome of the human comprises said human V gene segment and/or the human expresses antibodies comprising antibody variable domains derived from the recombination of said human V gene segment and a human J gene segment (and optionally a human D gene segment).
  • any preceding paragraph eg, according to any one of paragraphs 46 to 59
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc -fused human TOI receptor
  • the genome of the human comprises a heavy chain constant region nucleotide sequence that is identical to said first constant region nucleotide sequence and/or the human expresses antibodies comprising said human constant domain.
  • any preceding paragraph eg, according to any one of paragraphs 46 to 60
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc -fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CHI nucleotide sequence and/or the human expresses antibodies comprising said human gamma CHI domain.
  • any preceding paragraph eg, according to any one of paragraphs 46 to 61
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc -fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH2 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH2 domain.
  • any preceding paragraph eg, according to any one of paragraphs 46 to 62
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc-fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH3 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH3 domain.
  • any preceding paragraph eg, according to any one of paragraphs 46 to 63
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc-fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said CH4 nucleotide sequence and/or the human expresses antibodies comprising said human gamma CH4 domain.
  • any preceding paragraph eg, according to any one of paragraphs 46 to 64
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc-fused human TOI receptor
  • the genome of the human comprises a gamma heavy chain constant region nucleotide sequence that is identical to said Fc nucleotide sequence and/or the human expresses antibodies comprising said human gamma Fc region.
  • said human gamma heavy chain is a human gamma-2 heavy chain.
  • ligand comprises a human IGHG1 *01 gamma- 1 heavy chain constant region.
  • any one of paragraphs 61 to 69 and 71 to 74 comprising genotyping the human as positive for said gamma heavy chain constant region nucleotide sequence, eg, positive for said gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc nucleotide sequence; positive for said human IGHG1*01 gamma heavy chain constant region, CHI, CH2, CH3, CH4 or Fc nucleotide sequence; or positive for said human IGHG2*01 gamma heavy chain constant region, CHI, CH2, CH3, CH4 or Fc nucleotide sequence.
  • ligand eg, comprising or consisting of an antibody or fragment or an Fc -fused TOI receptor
  • the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 42 or a Leu corresponding to position 206 of SEQ ID NO: 42 and wherein the genome of the human comprises a gamma-1 heavy chain constant region nucleotide sequence that encodes such an Asp or Leu or the human expresses antibodies comprising human gamma- 1 constant regions comprising such an Asp or Leu.
  • human gamma- 1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 42 and a Leu corresponding to position 206 of SEQ ID NO: 42.
  • IGHG1 *01 gamma-1 heavy chain constant region eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG1*01.
  • genome of the human comprises a human IGHG1*01 nucleotide sequence or the human expresses antibodies comprising human constant domains encoded by a human IGHG1*01 nucleotide sequence.
  • ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 61.
  • the ligand comprises a human gamma-2 heavy chain constant region that comprises an amino acid selected from the group consisting of a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44, a Phe corresponding to position 76 of SEQ ID NO: 44, a Val corresponding to position 161 of SEQ ID NO: 44 and an Ala corresponding to position 257 of SEQ ID NO: 44; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such a selected amino acid or the human expresses antibodies comprising human gamma-2 constant regions comprising such a selected amino acid.
  • ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44, a Phe
  • the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such amino acids of (i) or the human expresses antibodies comprising human gamma-2 constant regions comprising such amino acids of (i).
  • ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Val corresponding to position 161 of SEQ ID NO: 44 and an Ala corresponding to position 257 of SEQ ID NO: 44 and optionally (ii) an amino acid selected from the group consisting of a Pro corresponding to position 72 of SEQ ID NO: 44, an Asn corresponding to position 75 of SEQ ID NO: 44 and a Phe corresponding to position 76 of SEQ ID NO: 44; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes such amino acids of (i) or the human expresses antibodies comprising human gamma-2 constant regions comprising such amino acids of (i).
  • ligand comprises a human IGHG2*01 gamma-2 heavy chain constant region, eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG2*01.
  • a human IGHG2*01 gamma-2 heavy chain constant region eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG2*01.
  • genome of the human comprises a human IGHG2*01 nucleotide sequence or the human expresses antibodies comprising human constant domains encoded by a human IGHG2*01 nucleotide sequence.
  • the ligand (eg, comprising or consisting of an antibody or fragment or an Fc -fused TOI receptor) comprises a human kappa light chain constant region that comprises a Val corresponding to position 84 of SEQ ID NO: 50 or a Cys corresponding to position 87 of SEQ ID NO: 50; and wherein the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val or Cys or the human expresses antibodies comprising human kappa light chain constant regions comprising such a Val or Cys.
  • the ligand eg, comprising or consisting of an antibody or fragment or an Fc -fused TOI receptor
  • the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val or Cys or the human expresses antibodies comprising human kappa light chain constant regions comprising such a Val or Cys.
  • ligand comprises a human kappa light chain constant region that comprises a Val corresponding to position 84 of SEQ ID NO: 50 and a Cys corresponding to position 87 of SEQ ID NO: 50.
  • the ligand comprises or consists of an antibody, wherein the antibody comprises light chains that comprise SEQ ID NO: 62 or 66.
  • ligand comprises or consists of an antibody, wherein the antibody comprises a light chain variable domain derived from recombination of a human VK gene segment and a human JK gene segment, wherein the JK gene segment is IGKJ2*01 (SEQ ID NO: 57).
  • ligand eg, comprising or consisting of an antibody or fragment or an Fc -fused TOI receptor
  • the ligand comprises a human IGLC2*01 light chain constant region
  • the genome of the human comprises a human IGLC2*01 nucleotide sequence or the human expresses antibodies comprising human light chain IGLC2*01 constant regions.
  • the ligand comprises or consists of an antibody or fragment, wherein the antibody or fragment comprises a VH domain that is derived from the recombination of a human VH gene segment, a human D gene segment and a human JH gene segment, wherein the VH gene segment is selected from the group consisting of (i) IGHV1-18*01 and the genome of the human comprises a human IGHVl - 18*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV1-18*01 ; or (ii) IGVH1 -46*01 and the genome of the human comprises a human IGHVl -46*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHVl -46*01.
  • fragment comprises a one more or all of a CHI domain, CH2 domain, CH3 domain, hinge or Fc encoded by human IGHG2*01.
  • the ligand comprises or consists of an antibody or fragment, wherein the antibody or fragment comprises a VL domain that is derived from the recombination of a human VL gene segment and a human JL gene segment, wherein the VL gene segment is selected from the group consisting of (i) IGKV4- 1*01 and the genome of the human comprises a human IGKV4-1*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV4-1*01 ; (ii) IGLV2- 14*01 and the genome of the human comprises a human IGLV2- 14*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGLV2- 14*01 ; or (iii) IGKV1 -13*02 and the genome of the human comprises a human IGKV1
  • the antibody or fragment comprises a light chain variable domain derived from recombination of a human VK gene segment and a human JK gene segment, wherein the JK gene segment is IGKJ2*01 (SEQ ID NO: 57; wherein (i) or (iii) applies.
  • ligand eg, antibody or fragment
  • Kd dissociation constant of InM or less as determined by SPR, (eg, 100, 10 or lpM or less).
  • PCSK9 Proprotein convertase subtilisin kexin type 9
  • LDLR low density lipoprotein receptor
  • mice have shown that increasing PCSK9 protein levels decreases levels of LDLR protein in the liver (Benjannet et al., 2004; Lagace et al., 2006; Maxwell et al., 2005; Park et al., 2004), while PCSK9 knockout mice have increased levels of LDLR in the liver (Rashid et al., 2005). Additionally, various human PCSK9 mutations that result in either increased or decreased levels of plasma LDL have been identified (Kotowski et al., 2006; Zhao et al., 2006). PCSK9 has been shown to directly interact with the LDLR protein, be endocytosed along with the LDLR, and co- immunofluoresce with the LDLR throughout the endosomal pathway (Lagace et al., 2006).
  • PCSK9 is a prohormone -proprotein convertase in the subtilisin (S8) family of serine proteases (Seidah et al., 2003). Humans have nine prohormone -proprotein convertases that can be divided between the S8A and S8B subfamilies (Rawlings et al., 2006). Furin, PC1/PC3, PC2, PACE4, PC4, PC5/PC6 and PC7/PC8/LPC/SPC7 are classified in subfamily S8B.
  • Prohormone-proprotein convertases are expressed as zymogens and they mature through a multi step process.
  • the function of the pro-domain in this process is two-fold.
  • the pro-domain first acts as a chaperone and is required for proper folding of the catalytic domain (Ikemura et al., 1987).
  • Once the catalytic domain is folded autocatalysis occurs between the pro-domain and catalytic domain.
  • the pro-domain remains bound to the catalytic domain where it then acts as an inhibitor of catalytic activity (Fu et al., 2000).
  • maturation proceeds with a second autocatalytic event at a site within the pro-domain (Anderson et al., 1997). After this second cleavage event occurs the pro-domain and catalytic domain dissociate, giving rise to an active protease.
  • 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.
  • the protein can be referred to as the "mature,” “cleaved”, “processed” or “active” PCSK9.
  • the protein can be referred to as the "inactive”, “pro-form”, or "unprocessed” form of PCSK9.
  • PCSK9 also encompasses PCSK9 molecules incorporating post-translational modifications of the PCSK9 amino acid sequence, such as PCSK9 sequences that have been glycosylated, 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 (see, e.g., FIGS. 1A and IB of US20120093818A1 ; which is incorporated by reference herein in its entirety).
  • the present invention provides anti-PCSK9 ligands; and PCSK9-binding or targeting ligands as described herein.
  • the ligands have a variety of utilities. Some of the ligands, for instance, are useful in specific binding assays, for genotyping or phenotyping humans, 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 ligands of the invention are useful for inhibiting binding of PCSK9 to LDLR, or inhibiting PCSK9-mediated activities.
  • Anti-PCSK9 ligands eg, antibodies and anti-sense RNA
  • Anti-PCSK9 ligands have been developed based on targeting and neutralising so-called "wild-type" human PCSK9, which is a commonly-occurring form (see, eg, US20120093818A1 and US20110065902A1 ; each of which is incorporated by reference herein in its entirety). While such therapies are useful for human patients harbouring this form of human PCSK9, the inventor considered it useful to investigate the possibility of targeting much rarer - but still naturally-occurring - forms of PCSK9 amongst human populations.
  • Such forms or “alleles” comprise multiple changes at the nucleotide and amino acid levels from the corresponding common form nucleotide and amino acids sequences, ie, there are multiple non-synonymous changes at the nucleotide level that translate into multiple corresponding changes in the protein target in humans.
  • the inventor realised that there is significant industrial and medical application for the invention in terms of guiding the choice of anti-PCSK9 ligand for administration to human patients for therapy and/or prophylaxis of PCSK9-mediated or associated diseases or conditions.
  • the patient receives drugs and ligands that are tailored to their needs - as determined by the patient's genetic or phenotypic makeup.
  • the invention provides for the genotyping and/or phenotyping of patients in connection with such treatment, thereby allowing a proper match of drug to patient. This increases the chances of medical efficacy, reduces the likelihood of inferior treatment using drugs or ligands that are not matched to the patient (eg, poor efficacy and/or side-effects) and avoids pharmaceutical mis-prescription and waste.
  • the present inventor decided to determine a set of human PCSK9 variants on the basis of the following criteria, these being criteria that the inventor realised would provide for useful medical drugs and diagnostics to tailored need in the human population.
  • the inventor selected variants having at least 3 of the 4 following criteria: -
  • PCSK9 variants having a cumulative human allele frequency in the range from 1 to 10%
  • PCSK9 variants having a total human genotype frequency in the range from 1 to about 15%;
  • the inventor's selection included, as a consideration, selection for nucleotide variation that produced amino acid variation in corresponding PCSK9 forms (ie, non-synonymous variations), as opposed to silent variations that do not alter amino acid residues in the target protein.
  • Table 1 Human PCSK9 variants distributed over several human ethnic populations & having a total human genotype frequency in the range of 1 to about 15%
  • x in a box indicates that the amino acid for the variant form is different from the amino acid at that position in form a, the variant amino acid being shown in " Amino Acid Position & Variation" of the table and the form a amino acid being shown in the first row of the table; amino acids at all other positions of each variant form are identical to those found in form a.
  • Amino acid numbering is per the numbering shown for the pro-form in Table 2 below.
  • Heterozygous human genotype frequency ie, cumulative frequency of all genotypes having one occurrence of the variant allele and one occurrence of another allele (heterozygous state), eg, ac genotype in 1000 Genomes database;
  • Homozygous human genotype frequency ie, cumulative frequency of two occurrences of the variant allele (homozygous state), eg, cc genotype in 1000 Genomes database;
  • Total human genotype frequency ie, total of heterozygous plus homozygous human genotype frequencies.
  • Form a' is identical to form a with the exception that form a' has a glycine (G) at position 620 (see US20120093818 (Amgen, Inc)); form a has E at this position .
  • G glycine
  • x in a box indicates that a variant allele comprises the non-synonymous nucleotide variation indicated in the 5th row.
  • chromosome number all positions are on human chromosome Incoordinate number (Ensembl release 73 - September 2013, Genome assembly: GRCh37 (GCA_000001405.13);
  • NCBI dbSNP reference number NCBI dbSNP Build 138 released on Apr 25, 2013.

Abstract

L'invention concerne des cibles humaines d'intérêt (TOI), des ligands anti-TOI, des kits, des compositions et une méthode.
PCT/GB2015/053361 2014-11-07 2015-11-06 Traitement de maladie par liaison de ligand à des cibles d'intérêt WO2016071701A1 (fr)

Applications Claiming Priority (22)

Application Number Priority Date Filing Date Title
US14/536,129 US9062105B1 (en) 2014-07-15 2014-11-07 Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
US14/536,129 2014-11-07
US14/536,049 2014-11-07
US14/536,049 US9045545B1 (en) 2014-07-15 2014-11-07 Precision medicine by targeting PD-L1 variants for treatment of cancer
US14/537,403 2014-11-10
US14/537,403 US9067998B1 (en) 2014-07-15 2014-11-10 Targeting PD-1 variants for treatment of cancer
GBPCT/GB2014/053729 2014-12-17
PCT/GB2014/053729 WO2015092393A2 (fr) 2013-12-17 2014-12-17 Cibles humaines
DE202014010421.2U DE202014010421U1 (de) 2013-12-17 2014-12-17 Menschliche Ziele
DE202014010421.2 2014-12-17
US14/659,910 2015-03-17
US14/659,910 US9109034B1 (en) 2014-07-15 2015-03-17 Precision medicine by targeting PD-L1 variants for treatment of cancer
US14/665,579 US9139648B1 (en) 2014-07-15 2015-03-23 Precision medicine by targeting human NAV1.9 variants for treatment of pain
US14/665,532 US9150660B1 (en) 2014-07-15 2015-03-23 Precision Medicine by targeting human NAV1.8 variants for treatment of pain
US14/665,579 2015-03-23
US14/665,532 2015-03-23
US14/731,727 2015-06-05
US14/731,727 US9187562B1 (en) 2014-07-15 2015-06-05 Methods for treating anaemia
EPPCT/EP2015/068491 2015-08-11
PCT/EP2015/068491 WO2016023916A1 (fr) 2014-08-12 2015-08-11 Traitement de maladie par la liaison d'un ligand à des cibles présentant un intérêt
US14/868,952 2015-09-29
US14/868,952 US9303089B2 (en) 2014-07-15 2015-09-29 Methods of treating anaemia

Publications (1)

Publication Number Publication Date
WO2016071701A1 true WO2016071701A1 (fr) 2016-05-12

Family

ID=55908648

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2015/053361 WO2016071701A1 (fr) 2014-11-07 2015-11-06 Traitement de maladie par liaison de ligand à des cibles d'intérêt

Country Status (1)

Country Link
WO (1) WO2016071701A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US10336824B2 (en) 2015-03-13 2019-07-02 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of thereof
CN110460238A (zh) * 2019-08-09 2019-11-15 华中科技大学 一种输入串联输出并联的dab变换器的解耦控制方法及装置
US10513558B2 (en) 2015-07-13 2019-12-24 Cytomx Therapeutics, Inc. Anti-PD1 antibodies, activatable anti-PD1 antibodies, and methods of use thereof
CN110662763A (zh) * 2017-02-21 2020-01-07 武汉大学 Nav1.9的靶点多肽、与其结合的抗体及抗体片段和相关药物组合物
WO2021105389A1 (fr) * 2019-11-29 2021-06-03 Kymab Limited Traitement de surcharge de fer physiologique
WO2021207072A1 (fr) * 2020-04-07 2021-10-14 Mabwell Therapeutics Inc. Anticorps anti-tmprss6 et leurs utilisations
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, and methods of use thereof

Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5599905A (en) 1988-10-31 1997-02-04 Immunex Corporation Interleukin-4 receptors
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5714146A (en) 1992-08-26 1998-02-03 Board Of Regents Of The University Of Washington IL-4 bone therapy
US5985280A (en) 1988-04-06 1999-11-16 Royal Postgraduate Medical School Of Hammersmith Hospital Diagnosis and/or therapy of tumours using monoclonal antibodies specific for the human IL-4 receptor
WO2001058957A2 (fr) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Amelioration de la demi-vie circulante de proteines de fusion a base d'anticorps
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
WO2001092340A2 (fr) 2000-05-26 2001-12-06 Immunex Corporation Utilisation d'antagonistes vis-a-vis de l'interleukine-4 (il-4) et compositions correspondantes
WO2002006919A2 (fr) 2000-07-18 2002-01-24 Aegis Analytical Corporation Systeme, procede et produit programme d'ordinateur pour la mise en correspondance de donnees provenant de plusieurs bases de donnees
WO2004016750A2 (fr) 2002-08-14 2004-02-26 Macrogenics, Inc. Anticorps specifiques du recepteur fc$g(g)riib et procedes d'utilisation de ces anticorps
WO2004029207A2 (fr) 2002-09-27 2004-04-08 Xencor Inc. Variants fc optimises et methodes destinees a leur generation
WO2004035752A2 (fr) 2002-10-15 2004-04-29 Protein Design Labs, Inc. Modification d'affinites de liaison pour fcrn ou de demi-vies seriques d'anticorps par mutagenese
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
WO2005040217A2 (fr) 2003-10-17 2005-05-06 Cambridge University Technical Services Limited Polypeptides comprenant des regions constantes modifiees
WO2005047331A2 (fr) 2003-11-07 2005-05-26 Immunex Corporation Anticorps liant un recepteur de l'interleucine 4
WO2005103081A2 (fr) 2004-04-20 2005-11-03 Genmab A/S Anticorps monoclonaux humains diriges contre cd20
US20060252077A1 (en) 2004-12-30 2006-11-09 Helicos Biosciences Corporation Stabilizing a nucleic acid for nucleic acid sequencing
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
JP2007020563A (ja) * 2005-06-13 2007-02-01 Sutaagen:Kk 糖尿病網膜症発症及び/又は進展リスクの診断方法
US20070070349A1 (en) 2005-09-23 2007-03-29 Helicos Biosciences Corporation Optical train and method for TIRF single molecule detection and analysis
US7226720B2 (en) 2003-09-08 2007-06-05 General Electric Company Limited play data storage media and method for limiting access to data thereon
US7279563B2 (en) 1999-10-05 2007-10-09 Helicos Biosciences Corporation Compounds for protecting hydroxyls and methods for their use
US7282337B1 (en) 2006-04-14 2007-10-16 Helicos Biosciences Corporation Methods for increasing accuracy of nucleic acid sequencing
US20080008697A1 (en) 2006-06-30 2008-01-10 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
WO2008054606A2 (fr) 2006-10-02 2008-05-08 Regeneron Pharmaceuticals, Inc. Anticorps humains à haute affinité vis-à-vis du récepteur il-4 humain
WO2008057459A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057458A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057457A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
GB2444410A (en) 2006-11-30 2008-06-04 Navigenics Inc Genetic profiling method
WO2008125623A2 (fr) 2007-04-13 2008-10-23 Novartis Ag Molécules et procédés de modulation de proprotéine convertase subtilisine/kexine de type 9 (pcsk9)
WO2008133647A2 (fr) 2006-11-07 2008-11-06 Merck & Co., Inc. Antagonistes de pcsk9
WO2009055783A2 (fr) 2007-10-26 2009-04-30 Schering Corporation Anti-pcsk9 et méthodes de traitement de troubles du métabolisme lipidique et du cholestérol
WO2009100318A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de 1b20 pcsk9
WO2009100297A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de pcsk9 1d05
WO2010029513A2 (fr) 2008-09-12 2010-03-18 Rinat Neuroscience Corporation Antagonistes de pcsk9
WO2010056981A2 (fr) * 2008-11-13 2010-05-20 Massachusetts General Hospital Procédés et compositions pour la régulation de l'homéostasie du fer par modulation de la protéine bmp-6
WO2010077854A1 (fr) 2008-12-15 2010-07-08 Regeneron Pharamaceuticals, Inc. Anticorps humains à grande affinité contre pcsk9
WO2011037791A1 (fr) 2009-09-25 2011-03-31 Merck Sharp & Dohme Corp. Antagonistes de pcsk9
WO2011053759A1 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes de la pcsk9 avec anticorps fab ax189 et ax1, et variantes afférentes
WO2011053783A2 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes et variants ax213 et ax132 pcsk9
WO2011051351A1 (fr) 2009-10-27 2011-05-05 Ucb Pharma S.A. Procédé pour générer des anticorps dirigés contre des canaux ioniques
WO2011072263A1 (fr) 2009-12-11 2011-06-16 Irm Llc Antagonistes de pcsk9
WO2011111007A2 (fr) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation Anticorps présentant une liaison à l'antigène dépendante du ph
WO2011128096A1 (fr) * 2010-04-16 2011-10-20 Roche Diagnostics Gmbh Marqueurs de polymorphisme destinés à prédire la réponse à un traitement par un médicament anticorps monoclonal inhibant les récepteurs de l'interleukine-6
WO2012041332A2 (fr) * 2010-10-01 2012-04-05 Rigshospitalet Variations génétiques du gène du récepteur d'interleukine-6 permettant de prédire la réponse de patients au traitement basé sur des inhibiteurs dudit récepteur d'interleukine-6
US20120093818A1 (en) 2007-08-23 2012-04-19 Simon Mark Jackson Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
US20130071405A1 (en) 2011-09-16 2013-03-21 Eli Lilly And Company Antibodies to pcsk9 and uses thereof
WO2013041844A2 (fr) 2011-09-19 2013-03-28 Kymab Limited Anticorps, domaines variables & chaînes adaptées pour une utilisation humaine
EP2604628A2 (fr) 2007-12-21 2013-06-19 Medimmune Limited Éléments de liaison pour le récepteur alpha interleukin-4 (IL-4R) - 173
WO2013090633A2 (fr) * 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer

Patent Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5985280A (en) 1988-04-06 1999-11-16 Royal Postgraduate Medical School Of Hammersmith Hospital Diagnosis and/or therapy of tumours using monoclonal antibodies specific for the human IL-4 receptor
US6716587B2 (en) 1988-10-31 2004-04-06 Immunex Corporation Antibodies to interleukin-4 receptors and uses thereof
US5717072A (en) 1988-10-31 1998-02-10 Immunex Corporation Antibodies that are immunoreactive with interleukin-4 receptors
US5767065A (en) 1988-10-31 1998-06-16 Immunex Corporation Use of interleukin-4 receptors to inhibit biological responses mediated by interleukin-4
US5840869A (en) 1988-10-31 1998-11-24 Immunex Corporation DNA encoding interleukin-4 receptors
US5599905A (en) 1988-10-31 1997-02-04 Immunex Corporation Interleukin-4 receptors
US5714146A (en) 1992-08-26 1998-02-03 Board Of Regents Of The University Of Washington IL-4 bone therapy
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6911345B2 (en) 1999-06-28 2005-06-28 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US7279563B2 (en) 1999-10-05 2007-10-09 Helicos Biosciences Corporation Compounds for protecting hydroxyls and methods for their use
WO2001058957A2 (fr) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Amelioration de la demi-vie circulante de proteines de fusion a base d'anticorps
WO2001092340A2 (fr) 2000-05-26 2001-12-06 Immunex Corporation Utilisation d'antagonistes vis-a-vis de l'interleukine-4 (il-4) et compositions correspondantes
WO2002006919A2 (fr) 2000-07-18 2002-01-24 Aegis Analytical Corporation Systeme, procede et produit programme d'ordinateur pour la mise en correspondance de donnees provenant de plusieurs bases de donnees
WO2004016750A2 (fr) 2002-08-14 2004-02-26 Macrogenics, Inc. Anticorps specifiques du recepteur fc$g(g)riib et procedes d'utilisation de ces anticorps
WO2004029207A2 (fr) 2002-09-27 2004-04-08 Xencor Inc. Variants fc optimises et methodes destinees a leur generation
WO2004035752A2 (fr) 2002-10-15 2004-04-29 Protein Design Labs, Inc. Modification d'affinites de liaison pour fcrn ou de demi-vies seriques d'anticorps par mutagenese
US7226720B2 (en) 2003-09-08 2007-06-05 General Electric Company Limited play data storage media and method for limiting access to data thereon
WO2005040217A2 (fr) 2003-10-17 2005-05-06 Cambridge University Technical Services Limited Polypeptides comprenant des regions constantes modifiees
WO2005047331A2 (fr) 2003-11-07 2005-05-26 Immunex Corporation Anticorps liant un recepteur de l'interleucine 4
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
WO2005103081A2 (fr) 2004-04-20 2005-11-03 Genmab A/S Anticorps monoclonaux humains diriges contre cd20
US20060252077A1 (en) 2004-12-30 2006-11-09 Helicos Biosciences Corporation Stabilizing a nucleic acid for nucleic acid sequencing
US7220549B2 (en) 2004-12-30 2007-05-22 Helicos Biosciences Corporation Stabilizing a nucleic acid for nucleic acid sequencing
JP2007020563A (ja) * 2005-06-13 2007-02-01 Sutaagen:Kk 糖尿病網膜症発症及び/又は進展リスクの診断方法
US20070070349A1 (en) 2005-09-23 2007-03-29 Helicos Biosciences Corporation Optical train and method for TIRF single molecule detection and analysis
US7282337B1 (en) 2006-04-14 2007-10-16 Helicos Biosciences Corporation Methods for increasing accuracy of nucleic acid sequencing
US20080008697A1 (en) 2006-06-30 2008-01-10 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
WO2008054606A2 (fr) 2006-10-02 2008-05-08 Regeneron Pharmaceuticals, Inc. Anticorps humains à haute affinité vis-à-vis du récepteur il-4 humain
WO2008133647A2 (fr) 2006-11-07 2008-11-06 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057459A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057457A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008063382A2 (fr) 2006-11-07 2008-05-29 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057458A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
GB2444410A (en) 2006-11-30 2008-06-04 Navigenics Inc Genetic profiling method
WO2008125623A2 (fr) 2007-04-13 2008-10-23 Novartis Ag Molécules et procédés de modulation de proprotéine convertase subtilisine/kexine de type 9 (pcsk9)
US20120093818A1 (en) 2007-08-23 2012-04-19 Simon Mark Jackson Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
WO2009055783A2 (fr) 2007-10-26 2009-04-30 Schering Corporation Anti-pcsk9 et méthodes de traitement de troubles du métabolisme lipidique et du cholestérol
EP2604628A2 (fr) 2007-12-21 2013-06-19 Medimmune Limited Éléments de liaison pour le récepteur alpha interleukin-4 (IL-4R) - 173
WO2009100297A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de pcsk9 1d05
WO2009100318A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de 1b20 pcsk9
WO2010029513A2 (fr) 2008-09-12 2010-03-18 Rinat Neuroscience Corporation Antagonistes de pcsk9
WO2010056981A2 (fr) * 2008-11-13 2010-05-20 Massachusetts General Hospital Procédés et compositions pour la régulation de l'homéostasie du fer par modulation de la protéine bmp-6
WO2010077854A1 (fr) 2008-12-15 2010-07-08 Regeneron Pharamaceuticals, Inc. Anticorps humains à grande affinité contre pcsk9
US20110065902A1 (en) 2008-12-15 2011-03-17 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to pcsk9
WO2011037791A1 (fr) 2009-09-25 2011-03-31 Merck Sharp & Dohme Corp. Antagonistes de pcsk9
WO2011051351A1 (fr) 2009-10-27 2011-05-05 Ucb Pharma S.A. Procédé pour générer des anticorps dirigés contre des canaux ioniques
WO2011053759A1 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes de la pcsk9 avec anticorps fab ax189 et ax1, et variantes afférentes
WO2011053783A2 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes et variants ax213 et ax132 pcsk9
WO2011072263A1 (fr) 2009-12-11 2011-06-16 Irm Llc Antagonistes de pcsk9
WO2011111007A2 (fr) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation Anticorps présentant une liaison à l'antigène dépendante du ph
WO2011128096A1 (fr) * 2010-04-16 2011-10-20 Roche Diagnostics Gmbh Marqueurs de polymorphisme destinés à prédire la réponse à un traitement par un médicament anticorps monoclonal inhibant les récepteurs de l'interleukine-6
WO2012041332A2 (fr) * 2010-10-01 2012-04-05 Rigshospitalet Variations génétiques du gène du récepteur d'interleukine-6 permettant de prédire la réponse de patients au traitement basé sur des inhibiteurs dudit récepteur d'interleukine-6
US20130071405A1 (en) 2011-09-16 2013-03-21 Eli Lilly And Company Antibodies to pcsk9 and uses thereof
WO2013041844A2 (fr) 2011-09-19 2013-03-28 Kymab Limited Anticorps, domaines variables & chaînes adaptées pour une utilisation humaine
WO2013090633A2 (fr) * 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer

Non-Patent Citations (84)

* Cited by examiner, † Cited by third party
Title
"Association and gene-gene interactions of eight common single - nucleotide polymorphisms with pediatric asthma in middle china", J ASTHMA, vol. 47, no. 3, April 2010 (2010-04-01), pages 238 - 44
"The Merck Manual of Diagnosis and Therapy, 19th Edition,", 2006, MERCK RESEARCH LABORATORIES, ISBN: 0-911910-19-0
A. CALLEGARO ET AL., NUCLEIC ACIDS RES., vol. 34, no. 7, 14 April 2006 (2006-04-14), pages E56
A.A. KOMAR: "Single Nucleotide Polymorphisms, Methods in Molecular Biology", vol. 578, HUMANA PRESS
ANDREWS ET AL., J. BIOL. CHEM, vol. 277, 2002, pages 46073 - 46078
BENJAMIN LEWIN: "Genes X", 2009, JONES & BARTLETT PUBLISHING, ISBN: 0763766321
BENJANNET ET AL.: "NARC-1/PCSK9 and its natural mutants: zymogen cleavage and effects on the low density lipoprotein (LDL) receptor and LDL cholesterol", J BIOL CHEM., vol. 279, no. 47, 9 September 2004 (2004-09-09), pages 48865 - 75
BENTLEY, D. R, CURR OPIN GENET DEV, vol. 16, 2006, pages 545 - 52
CELL, vol. 157, no. 6, 22 May 2014 (2014-05-22), pages 1393 - 404
CHEN ET AL.: "A common PCSK9 haplotype, encompassing the E670G coding single nucleotide polymorphism, is a novel genetic marker for plasma low-density lipoprotein cholesterol levels and severity of coronary atherosclerosis", J AM COLL CARDIOL., vol. 45, no. 10, 21 April 2005 (2005-04-21), pages 1611 - 9
CHIARAMONTE ET AL., J CLIN INVEST, vol. 104, no. 6, 1999, pages 777 - 85
CHIARAMONTE, HEPATOLOGY, vol. 34, no. 2, 2001, pages 273 - 82
CHOTHIA, C ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLIN EXP ALLERGY, vol. 33, August 2003 (2003-08-01), pages 1111 - 7
COLIGAN ET AL.,: "Current Protocols in Protein Sciences", 2009, WILEY INTERSCIENCES
CUNNINGHAM ET AL.: "Structural and biophysical studies of PCSK9 and its mutants linked to familial hypercholesterolemia", NAT STRUCT MOL BIOL, vol. 14, no. 5, 15 April 2007 (2007-04-15), pages 413 - 9
DAVIS ET AL.: "Basic Methods in Molecular Biology", 1995, ELSEVIER SCIENCE PUBLISHING, INC.
DE LA VEGA FM; LAZARUK KD; RHODES MD; WENZ MH: "Assessment of two flexible and compatible SNP genotyping platforms: TaqMan SNP Genotyping Assays and the SNPlex Genotyping System", MUTAT RES., vol. 573, no. 1-2, 3 June 2005 (2005-06-03), pages 111 - 35
DE WILDT ET AL., EUR J. IMMUNOL., vol. 26, no. 3, 1996, pages 629 - 39
DIEFFENBACH, C. W.; G. S. DVEKSLER: "PCR Primer, a Laboratory Manual", 1995, COLD SPRING HARBOR PRESS
DORDO ET AL., J. MOL BIOL, vol. 217, 1999, pages 721 - 739
DRENTH JP; WAXMAN SG: "Mutations in sodium-channel gene SCN9A cause a spectrum of human genetic pain disorders", J CLIN INVEST, vol. 117, no. 12, December 2007 (2007-12-01), pages 3603 - 9
FALLON ET AL., J IMMUNOL, vol. 164, no. 5, 2000, pages 2585 - 91
GESSNER ET AL., IMMUNOBIOLOGY, vol. 201, 2000, pages 285
GONG-QING SHEN, SINGLE NUCLEOTIDE POLYMORPHISMS, METHODS IN MOLECULAR BIOLOGY, vol. 578, 2009, pages 293 - 306
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
GOODSON: "Medical Applications of Controlled Release", vol. 2, 1984, pages: 115 - 138
HANCOCK ET AL., AM J RESPIR CELL MOL BIOL, vol. 18, no. 1, 1998, pages 60 - 5
HASEGAWA ET AL., J RHEUMATOL, vol. 24, no. 2, 1997, pages 328 - 32
HAUBER ET AL., J. CYST FIBR, vol. 2, 2003, pages 189
HORTON ET AL.: "Molecular biology of PCSK9: its role in LDL metabolism", TRENDS BIOCHEM SCI, vol. 32, no. 2, 9 January 2007 (2007-01-09), pages 71 - 7
JAMA, vol. 305, no. 14, 2011, pages 1460 - 1468
JENNIE P. MATHER AND DAVID BARNES: "Animal Cell Culture Methods", vol. 57, 1998, ACADEMIC PRESS
JOHN E. COLIGAN, ET. AL.,: "Current Protocols in Protein Science (CPPS)", JOHN WILEY AND SONS, INC
JUAN S. BONIFACINO ET. AL.: "Current Protocols in Cell Biology (CPCB)", JOHN WILEY AND SONS, INC.
KABAT, E. A. ET AL.: "Sequences of Proteins of Immunological Interest, Fifth Edition,", 1991, NIH PUBLICATION NO. 91-3242
KENDREW ET AL.: "Molecular Biology and Biotechnology: a Comprehensive Desk Reference", 1995, VCH PUBLISHERS, INC.
KOTOWSKI ET AL.: "A spectrum of PCSK9 alleles contributes to plasma levels of low-density lipoprotein cholesterol", AM J HUM GENET, vol. 78, no. 3, 20 January 2006 (2006-01-20), pages 410 - 22
LAGACE ET AL.: "Secreted PCSK9 decreases the number of LDL receptors in hepatocytes and in livers of parabiotic mice", J CLIN INVEST, vol. 116, no. 11, November 2006 (2006-11-01), pages 2995 - 3005
LANGER AND WISE: "Medical Applications of Controlled Release", 1974, CRC PRES.
LANGER AND WISE: "Medical Applications of Controlled Release", CRC PRES.
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
MARDIS: "The impact of next-generation sequencing technology on genetics", TRENDS IN GENETICS, vol. 24, no. 3, 2007, pages 133 - 141
MAXWELL ET AL.: "Overexpression of PCSK9 accelerates the degradation of the LDLR in a post-endoplasmic reticulum compartment", PROC NATL ACAD SCI USA., vol. 102, no. 6, 27 January 2005 (2005-01-27), pages 2069 - 74
N A ROSENBERG ET AL., SCIENCE, vol. 298, no. 5602, 20 December 2002 (2002-12-20), pages 2342 - 2343
NATURE, vol. 426, no. 6968, 18 December 2003 (2003-12-18), pages 789 - 96
NYREN, P ET AL., ANAL BIOCHEM, vol. 208, 1993, pages 17175
PARK ET AL.: "Post-transcriptional regulation of low density lipoprotein receptor protein by proprotein convertase subtilisin/kexin type 9a in mouse liver", J BIOL CHEM., vol. 279, no. 48, 22 September 2004 (2004-09-22), pages 50630 - 8
PISCIOTTA ET AL.: "Additive effect of mutations in LDLR and PCSK9 genes on the phenotype of familial hypercholesterolemia", ATHEROSCLEROSIS, vol. 186, no. 2, 23 September 2005 (2005-09-23), pages 433 - 40
POWELL ET AL.: "Compendium of excipients for parenteral formulations", J PHARM SCI TECHNOL, vol. 52, 1998, pages 238 - 311
R. IAN FRESHNEY: "Culture of Animal Cells: A Manual of Basic Technique; 5th edition", WILEY-LISS
RANADE K; CHANG MS; TING CT; PEI D; HSIAO CF; OLIVIER M; PESICH R; HEBERT J; CHEN YD; DZAU VJ, GENOME RES, vol. 11, no. 7, July 2001 (2001-07-01), pages 1262 - 8
RASHID ET AL.: "Decreased plasma cholesterol and hypersensitivity to statins in mice lacking Pcsk9", PROC NATL ACAD SCI USA, vol. 102, no. 15, 1 April 2005 (2005-04-01), pages 5374 - 9
REIMANN ET AL.: "Pain perception is altered by a nucleotide polymorphism in SCN9A", PROC NATL ACAD SCI USA., vol. 107, no. 11, 8 March 2010 (2010-03-08), pages 5148 - 53
RICHARD M. DURBIN ET AL: "A map of human genome variation from population-scale sequencing", NATURE, vol. 467, no. 7319, 28 October 2010 (2010-10-28), pages 1061 - 1073, XP055048531, ISSN: 0028-0836, DOI: 10.1038/nature09534 *
RISMA ET AL., J.IMMUNOL., vol. 169, no. 3, 2002, pages 1604 - 1610
ROBERT S. PORTER ET AL.: "The Encyclopedia of Molecular Biology", 1994, BLACKWELL SCIENCE LTD., ISBN: 0-632-02182-9
S. FRENCH; B. ROBSON, J. MOL. EVOL., vol. 19, 1983, pages 171
S. L. BERGER AND A. R. KIMMEL: "Methods in Enzymology: Guide to Molecular Cloning Techniques", vol. 152, 1987, ACADEMIC PRESS INC.
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual (4 ed.),", 2012, COLD SPRING HARBOR LABORATORY PRESS
SCIENCE, vol. 280, no. 5366, 15 May 1998 (1998-05-15), pages 1077 - 82
SEFTON, CRC CRIT. REF. BIOMED. ENG, vol. 14, 1987, pages 201
SEFTON, CRC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
SEIDAH ET AL.: "The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation", PROC NATL ACAD SCI USA., vol. 100, 27 January 2003 (2003-01-27), pages 928 - 33
SEIDAH; PRAT: "The proprotein convertases are potential targets in the treatment of dyslipidemia", J MOL MED (BERL, vol. 85, no. 7, 10 March 2007 (2007-03-10), pages 685 - 96
SHENDURE ET AL.: "Next-generation DNA sequencing", NATURE, vol. 26, no. 10, 2008, pages 1135 - 1145
SHIELD ET AL., JBC, vol. 277, 2002, pages 26733
SLUITER ET AL., EUR RESPIR J
STRAUSBERG, R. L. ET AL., DRUG DISC TODAY, vol. 13, 2008, pages 569 - 77
SU ET AL.: "Next-generation sequencing and its applications in molecular diagnostics", EXPERT REV MOL DIAGN, vol. 11, no. 3, 2011, pages 333 - 43
SYVANEN AC: "Review Accessing genetic variation: genotyping single nucleotide polymorphisms", NAT REV GENET, vol. 2, no. 12, December 2001 (2001-12-01), pages 930 - 42
TASHKIN ET AL., AM J RESPIR CRIT CARE MED, vol. 153, 1996, pages 1802 - 11
TAYLOR ET AL., J. THEOR. BIOL., vol. 119, 1986, pages 205 - 218
TREAT ET AL.: "Liposomes in the Therapy of Infectious Disease and Cancer", 1989, LISS, pages: 353 - 365
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60 - 69
TWYMAN RM; PRIMROSE SB: "Review Techniques patents for SNP genotyping", PHARMACOGENOMICS, vol. 4, no. 1, January 2003 (2003-01-01), pages 67 - 79
VAN DER POUW KRAAN ET AL., GENES IMMUN, vol. 3, no. 7, 2002, pages 436 - 9
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WU ET AL., J. BIOL. CHEM., vol. 262, 1987, pages 4429 - 4432
WU X ET AL.: "Haplotypes of the interleukin-4 receptor alpha chain gene associate with susceptibility to and severity of atopic asthma", CLIN EXP ALLERGY, vol. 34, no. 10, October 2004 (2004-10-01), pages 1570 - 5
ZHANG ET AL.: "The impact of next-generation sequencing on genomics", J GENET GENOMICS, vol. 38, no. 3, 2011, pages 95 - 109
ZHAO ET AL.: "Molecular characterization of loss-of-function mutations in PCSK9 and identification of a compound heterozygote", AM J HUM GENET, vol. 79, no. 3, 18 July 2006 (2006-07-18), pages 514 - 23
ZHENG ET AL., J CLIN INVEST, vol. 106, no. 9, 2000, pages 1081 - 93

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US10336824B2 (en) 2015-03-13 2019-07-02 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of thereof
US10669339B2 (en) 2015-03-13 2020-06-02 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of use thereof
US11174316B2 (en) 2015-03-13 2021-11-16 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of use thereof
US10513558B2 (en) 2015-07-13 2019-12-24 Cytomx Therapeutics, Inc. Anti-PD1 antibodies, activatable anti-PD1 antibodies, and methods of use thereof
CN110662763A (zh) * 2017-02-21 2020-01-07 武汉大学 Nav1.9的靶点多肽、与其结合的抗体及抗体片段和相关药物组合物
CN110662763B (zh) * 2017-02-21 2021-06-18 易森荟(武汉)生物医药有限公司 Nav1.9的靶点多肽、与其结合的抗体及抗体片段和相关药物组合物
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, and methods of use thereof
CN110460238A (zh) * 2019-08-09 2019-11-15 华中科技大学 一种输入串联输出并联的dab变换器的解耦控制方法及装置
WO2021105389A1 (fr) * 2019-11-29 2021-06-03 Kymab Limited Traitement de surcharge de fer physiologique
WO2021207072A1 (fr) * 2020-04-07 2021-10-14 Mabwell Therapeutics Inc. Anticorps anti-tmprss6 et leurs utilisations
US11866513B2 (en) 2020-04-07 2024-01-09 Mabwell Therapeutics, Inc. Anti-TMPRSS6 antibodies and uses thereof

Similar Documents

Publication Publication Date Title
US20230024543A1 (en) Methods of treating anaemia
US9109034B1 (en) Precision medicine by targeting PD-L1 variants for treatment of cancer
JP7202431B2 (ja) 対象とするヒト標的に特異的に結合するリガンド
US9062105B1 (en) Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
US9067998B1 (en) Targeting PD-1 variants for treatment of cancer
US8980273B1 (en) Method of treating atopic dermatitis or asthma using antibody to IL4RA
WO2015092393A2 (fr) Cibles humaines
US8945560B1 (en) Method of treating rheumatoid arthritis using antibody to IL6R
WO2016071701A1 (fr) Traitement de maladie par liaison de ligand à des cibles d'intérêt
US9017678B1 (en) Method of treating rheumatoid arthritis using antibody to IL6R
US8992927B1 (en) Targeting human NAV1.7 variants for treatment of pain
US8986691B1 (en) Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8986694B1 (en) Targeting human nav1.7 variants for treatment of pain
WO2016023916A1 (fr) Traitement de maladie par la liaison d'un ligand à des cibles présentant un intérêt
US9150660B1 (en) Precision Medicine by targeting human NAV1.8 variants for treatment of pain
TWI713444B (zh) 人類標靶
DE202014010421U1 (de) Menschliche Ziele
IES86644B2 (en) An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis
IES20140312A2 (en) An injectable antibody preparation for use in treating or reducing the risk of atopic dermatitis or asthma
GB2521355A (en) Human targets I

Legal Events

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

Ref document number: 15802184

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15802184

Country of ref document: EP

Kind code of ref document: A1