IES86644B2 - An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis - Google Patents

An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis Download PDF

Info

Publication number
IES86644B2
IES86644B2 IES20140315A IES20140315A IES86644B2 IE S86644 B2 IES86644 B2 IE S86644B2 IE S20140315 A IES20140315 A IE S20140315A IE S20140315 A IES20140315 A IE S20140315A IE S86644 B2 IES86644 B2 IE S86644B2
Authority
IE
Ireland
Prior art keywords
human
antibody
ligand
il6r
toi
Prior art date
Application number
IES20140315A
Inventor
Clube Jasper
Original Assignee
Kymab Ltd
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 US14/457,536 external-priority patent/US9017678B1/en
Priority claimed from US14/457,566 external-priority patent/US8945560B1/en
Priority claimed from US14/472,685 external-priority patent/US8992927B1/en
Priority claimed from US14/472,698 external-priority patent/US8986694B1/en
Priority claimed from US14/472,818 external-priority patent/US8980273B1/en
Priority claimed from US14/472,828 external-priority patent/US8986691B1/en
Priority claimed from US14/490,160 external-priority patent/US8999341B1/en
Priority claimed from US14/490,175 external-priority patent/US9040052B1/en
Priority claimed from EP14185297.0A external-priority patent/EP2975058A1/en
Priority claimed from US14/500,233 external-priority patent/US9045548B1/en
Priority claimed from US14/507,368 external-priority patent/US9034332B1/en
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
Application filed by Kymab Ltd filed Critical Kymab Ltd
Publication of IES20140315A2 publication Critical patent/IES20140315A2/en
Publication of IES86644B2 publication Critical patent/IES86644B2/en

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

An injectable preparation comprising an antibody or antibody fragment, for use in a method of treating or reducing the risk of rheumatoid arthritis in a human. The antibody or antibody fragment specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO. 78. The antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an amino acid selected from the group consisting of an Asp at position 204 shown in SEQ ID NO. 81 and a Leu at position 206 shown in SEQ ID NO. 81. The 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 amino acid. The human further comprises a nucleotide sequence encoding an IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO. 78.

Description

AN INJECTABLE ANTIBODY PREPARATION FOR USE IN TREATING OR REDUCING THE RISK OF RHEUMATOID ARTHRITIS TECHNICAL FIELD [00011 The technology described herein relates to anti-IL-6 receptor (IL6R) ligands, e.g., antibodies for the treatment of disease.
BACKGROUND id="p-2" id="p-2"
[0002] It is recognized that individual humans differ in their sequence and recently several individuals have had their genomes sequenced, for instance James Watson and Craig Venter. Comparison of the genome sequence of individuals has revealed differences in their sequences in both coding and non-coding parts of the genome. Some of these variations in humans are significant and contribute to phenotypic differences between individuals. In extreme cases these will result in genetic 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. id="p-3" id="p-3"
[0003] lnterleukin-6 (IL-6) is a complex cytokine, which plays a critical role in the regulation of inflammatory responses. Interleukin-6 (IL-6) signals through a ligand-binding IL-6 receptor (IL-6R; also known as CD 126) chain and a common signal-transducing chain glycoprotein 130 (gp 130; also known as CD 130), which is also engaged by receptors specific for IL-11, 1L-27, leukaemia inhibitory factor, oncostatin M (OSM), ciliary neurotrophic factor (CNTF) and cardiotrophin 1 (CT1). A soluble form of the 1L-6R (slL-6R) can be generated by proteolytic cleavage of mlL-6R by the metalloproteinases TNFa-converting enzyme (TACE; also known as ADAMI 7) and ADAM 10 or alternatively spliced mRNA. LL-6 responses can be induced classically in cells expressing mlL-6R through a high-affinity tetrameric complex consisting of IL-6, lL-6Rand two gpl3O molecules (or a hexameric complex consisting of two 1L-6, two mIL-6R and two gp 13 0 molecules). Alternatively, a sIL-6—IL-6R complex directly binds to and signals through gp 130 in cells lacking mIL-6R in a process that has been termed trans-signalling. High levels of IL-6 and sIL-6R have been reported in several chronic inflammatory diseases and in cancer. Several drugs that target different components of the IL-6 and IL-6R system have been described, including JL-6-specific monoclonal antibodies (mAbs) (for example, CNTO 328), IL-6R-specific mAbs (for example, tocilizumab) and soluble gpl30—Fc, ait antagonist of IL-6R trans-signalling. See a review at '’Averting inflammation by targeting the cytokine environment, Manfred Kopf, Martin F. Bachmann & Benjamin J. Marsland, Nature Reviews Drug Discovery 9, 703-718 (September 2010), doi:!0.1038/nrd2805 (incorporated herein by reference). id="p-4" id="p-4"
[0004] Genetic variation in the IL-6 receptor gene is associated with the risk of several human diseases with an inflammatory component, including coronary heart disease, rheumatoid arthritis, and asthma. A non-synonymous variant in the IL-6 receptor gene (IL6R Asp385Ala; 886644 rs2228145 A>C) is associated with the increased susceptibility to asthma. It is thought that the variant exerts its functional mechanism by regulating the balance between sIL6R (generated through cleavage of the cell surface receptor and by alternative splicing of a soluble IL6R isoform) and membrane-bound IL-6R (involved in classic IL6R signalling). Ferreira el al reportedly showed for the first time that the minor allele of this non-synonymous variant (Ala358) directly controls the surface levels of 1L-6R on individual immune cells and that these differences in protein levels translate into a functional impairment in IL6R signaling. id="p-5" id="p-5"
[0005] Ferreira et. al noted that while classic IL6R signalling appears responsible for regulatory T-cell suppression, [L6R trans-signalling (via sIL6R) seems to promote T helper 2 cell polarization in the lung. IL6R has been shown to be expressed in the epithelium, smooth muscle and vascular endothelium of human airways, and in macrophages and granulocytes of bronchoalveolar lavage fluid (BALF). Soluble IL6R levels in BALF are elevated in asthmatic patients compared to controls, and are elevated further upon allergen challenge, indicating an important role of sTL6R in the context of the lung and associated tissues. Consistent with these findings, as noted above 358Ala is also associated with an increased severity of asthma. The authors also provide evidence for the association of this non-synonymous variant with the risk of type 1 diabetes (TID) in two independent populations and confirm that rs2228145 is the major determinant of the concentration of circulating soluble IL6R (sIL6R) levels (reportedly 34.6% increase in sIL6R per copy of the minor allele 358Ala; rs2228145 [C]). The authors provided evidence that the non-synonymous variant rs2228145 regulates the balance of surface and sIL6R, and also affects the responsiveness of immune cells to IL6 stimulation. This mechanism underpins the effect of 358Ala on the IL-6/IL-6R pathway. id="p-6" id="p-6"
[0006] Revez et al comments that the main genetic determinant of soluble interleukin 6 receptor levels is the variant rs2228!45 that maps to the cleavage site of 1L6R. Reportedly, for each Ala allele, sIL6R serum levels increase by about 20 ng ml and asthma risk by 1.09-fold. However, the authors conclude that this variant does not explain the total heritability for slL6R levels.
Additional independent variants in IL6R may therefore contribute to variation in s!L6R levels and influence asthma risk. The authors imputed 471 variants in IL6R and tested these for association with sIL6R serum levels in 360 individuals. An intronic variant (rsl2083537) was associated with sIL6R levels independently of rs4129267 (P=0.0005), a proxy single-nucleotide polymorphism for rs2228145. A significant and consistent association for rs 12083537 was observed in a replication panel of 354 individuals (P=0.033). Each rs!2083537:A allele increased sIL6R serum levels by 2.4 ng ml. Analysis of mRNA levels in two cohorts did not identify significant associations between rsl2083537 and 1L6R transcription levels. On the other hand, results from 16705 asthmatics and 30809 controls showed that the rsl2083537:A allele increased asthma risk by 1.04-fold (P-0.0419). [0007] J C Galicia et al investigated the association of the IL6R polymorphisms with the serum levels of soluble IL6 receptor. In total, 115 healthy volunteers were genotyped, with 70 of them analyzed for sIL6R levels. Using the PCR/RFLP methods, two important polymorphic sites were selected for genotyping: the 48892A/C (D358A) in exon 9 and the -183G/A in the promoter region. In exon 9, C allele carriers had higher sIL6R level (P<0.0001) showing that this sequence variation, which corresponds to the proteolytic cleavage site of IL-6Ralpha, strongly influences the serum slL6R levels. In the promoter region, G allele carriers had lower sIL6R levels (P<0.0082) compared with the A allele carriers. [00081 Esparza-Gordillo et al examined the results of all genome-wide association studies from a public repository and selected 318 genetic markers that were significantly associated with any inflammatory trait. These markers were considered candidates and tested for association with atopic dermatitis (AD) in a 3-step approach including 7 study populations with 7130 patients with AD and 9253 control subjects. Reportedly a functional amino acid change in the IL-6 receptor (IL-6R Asp358Ala; rs2228145) was significantly associated with AD. Investigation of 2 independent population-based birth cohorts showed that 1L-6R 358Ala specifically predisposes to the persistent form of AD. Carriers of 3 58 Ala had increased serum levels of soluble IL6R, with homozygote carriers showing a 2-fold increase. Moreover, the authors reported that sIL6R levels were higher in patients with AD than in control subjects. The authors conclude that the study supports the importance of genetic variants influencing inflammation in the aetiology of AD. Moreover, they identified a functional genetic variant in IL6R influencing disease prognosis and specifically predisposing to persistent AD. id="p-9" id="p-9"
[0009] Antibodies to human IL6R are described in US8043617, US5670373, US5817790, EP409607 and those publications in Table 13 below. Therapeutic methods are described in US5888510, US8043617, US6723319 and those publications in Table 13 below. Actemra™ (tocilizumab) is a humanised example of an anti-IL-6R agent that blocks both classic and transsignaling. When directed to antibodies, the invention instead is directed to antibodies with fully human (not humanised) variable regions (and optionally also human constant regions). id="p-10" id="p-10"
[0010] Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of synovial tissue, leading to destruction of the joint architecture. It is recognized that cytokines such as tumor necrosis factor (TNF), interleukin-1 (IL-1) and interleukin-6 (IL-6) play a role in joint inflammation and cartilage damage observed in RA. IL-6 is a pleiotropic cytokine with biological effects on many cell types. IL-6 is often regarded as being downstream of TNF or IL-1 in inflammatory cytokine cascades and may therefore represent a common pathway factor in a wide range of inflammatory processes. Blockade of IL-6 signaling therefore offers the potential to ameliorate multiple pathogenic features of RA and other inflammatory diseases. id="p-11" id="p-11"
[0011] Therapeutic methods using IL-6R antagonists are mentioned in U.S. Pat. Nos. ,888,510; 6,723,319; and 2001/0001663. Exemplary anti-IL-6R antibodies are described in U.S. Pat. Nos. 7,582,298; 6,410,691; 5,817,790; 5,795,695; 6,670,373; and 7,582,298. id="p-12" id="p-12"
[0012] In certain embodiments, an IL6R 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. "IL6R" has also been referred to as interleukin-6 receptor subunit alpha, CD126; IL-6R-1; IL-6RA; IL6Q; IL6RA; IL6RQ; and gp80.
SUMMARY id="p-13" id="p-13"
[0013] Through the application of human genetic variation analysis and rationallydesigned sequence selection the present invention provides for improved human patient diagnosis and therapy based on human IL6R variation. Importantly, the invention enables tailored medicines that address individual human patient genotypes or phenotypes. id="p-14" id="p-14"
[0014] The inventor's analysis of large numbers of naturally-occurring genomic human TOI sequences reveals that there is significant variation across diverse human populations and provides for the ability for correlation between individual human patients and tailored medical and diagnostic approaches addressing the target. The technical applications of these findings, as per the present invention, thus contribute to better treatment, prophylaxis and diagnosis in humans and provides for patient benefit by 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. id="p-15" id="p-15"
[0015] Furthermore, the inventor surprisingly realised that some rarer natural forms, although present in humans at much lower frequencies than the common form, nevertheless are represented in multiple and ethnically-diverse human populations and usually with many human examples per represented ethnic population. Thus, the inventor realised that targeting such rarer forms would provide for effective treatment, prophylaxis or diagnosis across many human ethnic populations, thereby extending the utility of the present invention and better serving patients in those populations. [0016] With this, 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. In this way, the patient receives drugs and ligands that are tailored to their needs - as determined by the patient’s genetic or phenotypic makeup. Hand-in-hand with this, 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. id="p-17" id="p-17"
[0017] To this end, the invention provides:[0018] In a First Configuration id="p-19" id="p-19"
[0019] 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 said disease or condition, wherein the TOI in said human is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence having a total human genotype frequency of less than 50%; wherein b. Before step (a) said human has been or is genotyped as positive for said nucleotide sequence or phenotyped as positive for said TOI variant. id="p-20" id="p-20"
[0020] In a Second Configuration id="p-21" id="p-21"
[0021] 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 said disease or condition, wherein the TOI in said human is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence having a total human genotype frequency of less than 50%; wherein b. before step (a) the ligand has been or is determined as capable of binding to said TOI variant. id="p-22" id="p-22"
[0022] In a Third Configuration id="p-23" id="p-23"
[0023] 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 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% and/or having a total human genotype frequency of more than 50%; wherein b. Before 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%. id="p-24" id="p-24"
[0024] In a Fourth Configuration id="p-25" id="p-25"
[0025] An anti-human TOI ligand for use in a method of treating and/or preventing a TOImediated 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. id="p-26" id="p-26"
[0026] In a Fifth Configuration id="p-27" id="p-27"
[0027] 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. id="p-28" id="p-28"
[0028] In a Sixth Configuration id="p-29" id="p-29"
[0029] A pharmaceutical composition or kit for treating or preventing a condition or disease mediated by a TOI. id="p-30" id="p-30"
[0030] In a Seventh Configuration id="p-31" id="p-31"
[0031] 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. id="p-32" id="p-32"
[0032] In a Eighth Configuration id="p-33" id="p-33"
[0033] 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. id="p-34" id="p-34"
[0034] In a Ninth Configuration id="p-35" id="p-35"
[0035] A kit for TOI genotyping a human, wherein 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 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. id="p-36" id="p-36"
[0036] In a Tenth Configuration id="p-37" id="p-37"
[0037] 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%. id="p-38" id="p-38"
[0038] In a Eleventh Configuration id="p-39" id="p-39"
[0039] 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. id="p-40" id="p-40"
[0040] In a Twelfth Configuration id="p-41" id="p-41"
[0041] 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. id="p-42" id="p-42"
[0042] In a Thirteenth Configuration id="p-43" id="p-43"
[0043] 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%. id="p-44" id="p-44"
[0044] In a Fourteenth Configuration id="p-45" id="p-45"
[0045] 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%. id="p-46" id="p-46"
[0046] In an example, the TOI is a human TOI selected from the group consisting of PCSK9, VEGF-A and 1L6 receptor. id="p-47" id="p-47"
[0047] A Fifteenth Configuration provides a ligand, method, use, kit or composition of the invention, wherein (i) the ligand (eg, antibody or fragment) comprises (a) a 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 (b) a constant region domain encoded by a C region gene segment; Wherein 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; and (ii) 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. id="p-48" id="p-48"
[0048] 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). id="p-49" id="p-49"
[0049] 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. id="p-50" id="p-50"
[0050] 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. id="p-51" id="p-51"
[0051] 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, [0052] 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 corresponding to position 84 of SEQ ID NO: 127 or a Cys corresponding to position 87 of SEQ ID NO: 127; 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. id="p-53" id="p-53"
[0053] 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*O1 and the genome of the human comprises a human IGHV 1-18*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV 1-18*01; or (ii) IGVH 1-46*01 and the genome of the human comprises a human IGHV 1-46*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV 1-46*01. id="p-54" id="p-54"
[0054] 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-l*01 and the genome of the human comprises a human IGKV4-l*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV4-l*0l; (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 1GLV2-14*O1; or (iii) IGKV 1-13*02 and the genome of the human comprises a human IGKV1-I3*O2 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV 1-13*02. id="p-55" id="p-55"
[0055] Furthermore, the inventor's analysis of large numbers of naturally-occurring genomic human IL6R sequences reveals that there is significant variation across diverse human populations and provides for the ability for correlation between individual human patients and tailored medical and diagnostic approaches addressing the target. The technical applications of these findings, as per the present invention, thus contribute to better treatment, prophylaxis and diagnosis in humans and provides for patient benefit by 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. id="p-56" id="p-56"
[0056] Furthermore, the inventor surprisingly realised that some rarer natural forms, although present in humans at much lower frequencies than the common form, nevertheless are represented in multiple and ethnically-diverse human populations and usually with many human examples per represented ethnic population. Thus, the inventor realised that targeting such rarer forms would provide for effective treatment, prophylaxis or diagnosis across many human ethnic populations, thereby extending the utility of the present invention and better serving patients in those populations. id="p-57" id="p-57"
[0057] With this, the inventor realised that there is significant industrial and medical application for the invention in terms of guiding the choice of an anti-IL6R ligand for administration to human patients for therapy and/or prophylaxis of IL6R-mediated or associated diseases and conditions. In this way, the patient receives drugs and ligands that are tailored to their needs - as determined by the patient’s genetic or phenotypic makeup. Hand-in-hand with this, 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. id="p-58" id="p-58"
[0058] In addition to binding specificity for IL6R variant(s), the invention also relates to tailoring of the ligand (eg, antibody) sequence per se to the recipient human patient. id="p-59" id="p-59"
[0059] To this end, the invention provides:[0060] In a First Configuration id="p-61" id="p-61"
[0061] A method of treating or reducing the risk of an lL6R-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 an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; wherein the genome of said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-62" id="p-62"
[0062] In a first embodiment, the ligand 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 (i) a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109 and wherein said human comprises a VH gene segment encoding a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109, or the human expresses VH domains that comprise a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109; or (ii) a FW3 (framework 3) comprising a Thr at position 33 shown in SEQ ID NO: 111 and wherein said human comprises a VH gene segment encoding a FW3 comprising a Thr at position 110 shown in SEQ ID NO: 111 or the human expresses VH domains that comprise a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111. id="p-63" id="p-63"
[0063] In a second embodiment, 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. id="p-64" id="p-64"
[0064] In a third embodiment, the ligand comprises a Vk domain derived from the recombination of human Vk segment IGKV1-12*01 and a human Jk segment, and wherein said human comprises a JGKV 1-12*01 Vk gene segment or the human expresses Vk domains derived from the recombination of human Vk segment IGKVbl2*01 and a human Jk segment. id="p-65" id="p-65"
[0065] In a fourth embodiment, 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: 113 and wherein said human comprises a Vk gene segment encoding a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113, or the human expresses Vk domains that comprise a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113; or (ii) a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 and wherein said human comprises a Vk gene segment encoding a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 or the human expresses Vk domains that comprise a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115. id="p-66" id="p-66"
[0066] In a fifth embodiment, the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises an IGHG1*O1 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: 81 or a Leu at position 206 shown in SEQ ID NO: 81. id="p-67" id="p-67"
[0067] In a sixth embodiment, 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: 83, an Asn at position 75 shown in SEQ ID NO: 83, a Phe at position 76 shown in SEQ ID NO: 83, a Val at position 161 shown in SEQ ID NO: 83 and an Ala at position 257 shown in SEQ ID NO: 83 and wherein said human comprises an IGHG2*0l 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: 83, Asn at position 75 shown in SEQ ID NO: 83, Phe at position 76 shown in SEQ ID NO: 83, Val at position 161 shown in SEQ ID NO: 83 or Ala at position 257 shown in SEQ ID NO: 83. id="p-68" id="p-68"
[0068] In a seventh embodiment, the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and wherein said human comprises an IGKC1 *01 human heavy 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: 93 or a Cys at position 87 shown in SEQ ID NO: 93.
J0069] In an eighth embodiment, the ligand comprises a human IGLC1 *01 lambda chain constant region and wherein said human comprises a human IGLC1 *01 lambda chain constant region gene segment, or the human expresses antibodies comprising human IGLC1 *01 lambda chain constant regions. id="p-70" id="p-70"
[0070] These eight embodiments are also individually combinable with any of the other configurations disclosed herein. id="p-71" id="p-71"
[0071] In a Second Configuration id="p-72" id="p-72"
[0072] Corresponding anti-IL6R ligands (eg, an antibody or fragment) for treating or reducing the risk of an IL6R-mediated disease or condition in a human in need thereof, wherein the ligands specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val3851 le in SEQ ID NO: 78. |0073] In a Third Configuration id="p-74" id="p-74"
[0074] A pharmaceutical composition or kit for treating, reducing the risk of, or preventing an IL6R-mediated condition or disease. (0075] In a Fourth Configuration id="p-76" id="p-76"
[0076] A method of producing an anti-human IL6R antibody binding site, the method comprising obtaining a plurality of anti-IL6R antibody binding sites, screening the antibody binding sites for binding to a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and isolating an antibody binding site that binds in the screening step, and optionally producing an IL6R-binding fragment or derivative of the isolated antibody. id="p-77" id="p-77"
[0077] In a Fifth Configuration id="p-78" id="p-78"
[0078] A method of producing an anti-human IL6R antibody, the method comprising immunising a non-human vertebrate (eg, a mouse or a rat that expresses human variable domains) with a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 or a peptide thereof that comprises a mutation Asp358Ala or Val385Ue in SEQ ID NO: 78 and isolating an antibody that binds a human IL6R comprising said mutation or a peptide thereof that said mutation, and optionally producing an IL6R-binding fragment or derivative of the isolated antibody. id="p-79" id="p-79"
[0079] In a Sixth Configuration id="p-80" id="p-80"
[0080] A kit for IL6R genotyping a human, wherein the kit comprises a nucleic acid (i) comprising a sequence of contiguous nucleotides that specifically hybridises to a nucleotide sequence encoding an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, or specifically hybridises to an antisense sequence or an RNA transcript of said sequence, wherein said sequence of contiguous nucleotides hybridises to the nucleotide present in said selected sequence which encodes said mutation or hybridises to an antisense sequence or an RNA transcript thereof; and/or (ii) comprising a sequence of at least 10 contiguous nucleotides of an IL6R nucleotide sequence wherein said contiguous nucleotides encode said mutation or comprising an antisense sequence or RNA version of said contiguous nucleotides. id="p-81" id="p-81"
[0081] In a Seventh Configuration id="p-82" id="p-82"
[0082] Use of an anti-IL6R ligand that specifically binds a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 in the manufacture of a medicament for treating, reducing the risk of, or preventing an IL6R-mediated disease or condition in a human whose genome comprises an IL6R nucleotide sequence encoding said mutation. id="p-83" id="p-83"
[0083] In a Eighth Configuration id="p-84" id="p-84"
[0084] Use of an anti-IL6R ligand that specifically binds a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 in the manufacture of a medicament for targeting said IL6R in a human to treat, reduce the risk of, or prevent a disease or condition mediated by IL6R. id="p-85" id="p-85"
[0085] In a Ninth Configuration id="p-86" id="p-86"
[0086] A method of targeting an IL6R for treating, reducing the risk of, or preventing an IL6R-mediated disease or condition in a human, the method comprising administering an anti-IL6R ligand to a human comprising a nucleotide sequence that encodes a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, whereby an IL6R encoded by said nucleotide sequence is targeted. id="p-87" id="p-87"
[0087] In a Tenth Configuration id="p-88" id="p-88"
[0088] A method of treating, reducing the risk of, or preventing a disease or condition mediated by IL6R in a human, the method comprising targeting a human IL6R that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78 by administering to the human a ligand that specifically binds said 1E6R thereby treating, reducing the risk of, or preventing said disease or condition in the human. id="p-89" id="p-89"
[0089] In a Eleventh Configuration id="p-90" id="p-90"
[0090] A method of IL6R genotyping a nucleic acid sample of a human, the method comprising identifying in the sample the presence of an TL6R nucleotide sequence that encodes a mutation Asp358Ala or Val385He in SEQ ID NO: 78. id="p-91" id="p-91"
[0091] In a Twelfth Configuration id="p-92" id="p-92"
[0092] A method of IL6R typing a protein sample of a human, the method comprising identifying in the sample the presence of a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-93" id="p-93"
[0093] In a Thirteenth Configuration id="p-94" id="p-94"
[0094] A method of treating, reducing the risk o,f or preventing in a human patient a disease or condition selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease, optionally wherein the patient is receiving or has previously received an anti-TNF alpha treatment for said disease or condition, the method comprising typing the patient using a method of the invention and administering a ligand according to the invention whereby the human is treated or said disease or condition is prevented; optionally also reducing or stopping said anti-TNF alpha treatment. id="p-95" id="p-95"
[0095] In an embodiment of any configuration, the disease or condition is severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. id="p-96" id="p-96"
[0096] In an example, the ligand or method • Reduces T helper 2 cell polarization in the lung; • Treats or reduces the risk of a lung disease, eg, asthma; • Treats or reduces the risk of type 1 diabetes; ♦ Treats or reduces the risk of atopic dermatitis; or is a ligand or method for said purpose.
Optionally the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala in SEQ ID NO: 78. In addition, optionally, the human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala in SEQ ID NO: 78.
BRIEF DESCRIPTION OF THE DRAWINGS id="p-97" id="p-97"
[0097] This patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee. id="p-98" id="p-98"
[0098] Figure 1 shows in silica modeling of PCSK9 surface variant residues. id="p-99" id="p-99"
[0099] 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. 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. id="p-100" id="p-100"
[00100] 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 68, 68, 68, 70, 70, 70, 71, 73, 75, 39 and 76, respectively, in order of appearance. Figure 3 discloses the coded amino acid sequences as SEQ ID NOS 69, 69, 69, 69, 69, 69, 72, 74, 74, 38 and 77, respectively, in order of appearance. [00.101] 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 FW1 encoded by VH3-23*04 is depicted (SEQ ]D NO: 40).
DETAILED DESCRIPTION id="p-102" id="p-102"
[00102] 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), that target being human IL6R. id="p-103" id="p-103"
[00103] 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. id="p-104" id="p-104"
[00104] As sources of genomic sequence variation data, the skilled person will be aware of the available databases and resources (including updates thereof) provided by the following:- 1, HapMap (The International HapMap Consortium. 2003; http://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. 2. 1000 Genomes Project (The 1000 Genomes Project Consortium 2010; available on the World Wide Web at www.1000genomes.org/). This resource provides complete genomic sequence for at least 2500 unidentified individuals from one of 25 distinct population groups. For the purposes of the present invention, for example, the 100 Genomes database release 20130502 (see available at ftp.1000genomes.ebi.ac.uk/voll/ftp/release/20130502/) can be used, which is incorporated herein by refrence. 3. Japanese SNP Database( H.Haga et al. 2002; available on the World Wide Web at snp.ims.utokyo.ac.jp/index.html). Based on a study identifying 190,562 human genetic variants. id="p-105" id="p-105"
[00105] The present invention involves the identification and cataloguing of naturallyoccurring human genomic target sequence variants, including those found to be relatively lowfrequency or rare variants that segregate with specific human ethnic populations and in many individual humans. id="p-106" id="p-106"
[00106] 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. In devising this rational design of the present aspect of the invention, 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. As pail of this design, 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. id="p-107" id="p-107"
[00107] Thus, in this aspect of the invention, 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. id="p-108" id="p-108"
[00108] Selection Criteria id="p-109" id="p-109"
[00109] Three or four of the following:- • Naturally-occurring human target variation sequences, e.g., IL6R variation, having a cumulative human allele frequency of 35% or less; • Naturally-occurring human target variation sequences, e.g., IL6R variation, having a total human genotype frequency of about 50% or less; • Naturally-occurring human target variation sequences, e.g., IL6R variation, found in many different human ethnic populations (using the standard categorisation of the 1000 Genomes Project; see Table 12 below); and • Naturally-occurring human target variation sequences, e.g., IL6R variation, found in many individuals distributed across such many different ethnic populations. id="p-110" id="p-110"
[00110] The inventor’s selection included, as a consideration, selection for nucleotide variation that produced amino acid variation in corresponding TOI forms, e.g., IL6R forms, (ie, nonsynonymous variations), as opposed to silent variations that do not alter amino acid residues in the target protein. id="p-111" id="p-111"
[00111] In an embodiment, the cumulative human allele frequency is 30, 25, 20, 15, 10 or % or less, eg, in the range from 1 to 20% or 1 to 15% or 1 to 10%. id="p-112" id="p-112"
[00112] In an embodiment, the total human genotype frequency is 35, 30, 25, 20, 15, 10 or % or less, eg, in the range from I to 25%, 1 to 20%, 1 to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or I to 5% or 1 to about 5%. [00113) In an embodiment, the cumulative human allele frequency is 32, 30, 25, 20, 15, 10 or 5% or less, eg, in the range from 1 to 32% or 5 to 32% or 1 to 10%. id="p-114" id="p-114"
[00114] In an embodiment, the total human genotype frequency is 51, 50, 40, 35, 30, 25, , 15, 10 or 5% or less, eg, in the range from 1 to 51%, 1 to 25%, I to 20%, I to 15%, 1 to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%. id="p-115" id="p-115"
[00115] In an embodiment, 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). id="p-116" id="p-116"
[00116] In an embodiment, 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, 150, 200, 300, 400 or 500 individuals distributed across such many different ethnic populations. id="p-117" id="p-117"
[00117] In an example, the following criteria are applied:- • Naturally-occurring human target, e.g, IL6R, variant sequences having a cumulative human allele frequency of 15% or less; • Naturally-occurring human target, e.g., IL6R, variant sequences having a total human genotype frequency of 20% or less; • Naturally-occurring human target, e.g., TL6R, variant sequences found in at least 5 (4 for 1L6R) different human ethnic populations (using the standard categorisation of the 1000 Genomes Project); and • Naturally-occurring human target, e.g., IL6R, variant sequences found in many individuals distributed across such many different ethnic populations. id="p-118" id="p-118"
[00118] In any aspect, configuration, example, embodiment, clause or concept herein, frequencies may be determined using bioinformatics. id="p-119" id="p-119"
[00119] In any aspect, configuration, example, embodiment, clause or concept herein, frequencies may be determined by reference to a database comprising at least 1000 or 2000 human sequences. id="p-120" id="p-120"
[00120] In any aspect, configuration, example, embodiment, clause or concept herein "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. id="p-121" id="p-121"
[00121] In any aspect, configuration, example, embodiment, clause or concept herein "homozygous human genotype frequency means the cumulative frequency of two occurrences of the variant allele (homozygous state), eg, genotype in a 1000 Genomes Project database. id="p-122" id="p-122"
[00122] In any aspect, configuration, example, embodiment, clause or concept herein "total human genotype frequency" means the total of heterozygous plus homozygous human genotype frequencies. id="p-123" id="p-123"
[00123] In any aspect, configuration, example, embodiment, clause or concept herein "cumulative human allele frequency" refers to the total of all occurrences of the variant allele in the sample or database or in humans, eg, in a 1000 Genomes Project database. id="p-124" id="p-124"
[00124] in an example, the criteria are applied with reference to one or more human genomic sequence databases as described herein. For example, the criteria are those as applied to the 1000 Genomes database. id="p-125" id="p-125"
[00125] For example in any aspect example, embodiment or configuration of the invention, the 1000 Genomes database release 13. For example, the 1000 Genomes database in its most recent version as at 1 October 2013 or at 1 August 2014. In an example, the version is 20110521. In another example it is version 20130502. id="p-126" id="p-126"
[00126] Optionally, further sequence analysis and 3D in silico modelling (eg, see Figure 1) 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. id="p-127" id="p-127"
[00127] The following bioinformatics protocol is envisaged to indentify human sequences for use in the present invention: (a) Identify a genomic region containing a target sequence of interest, e.g., an IL6R target sequence of interest (Target genomic region’) and calculate the genomic coordinates, using coordinates that match the sequence assembly build used by either the 1000 Genomes Project or International HapMap project (or another selected human gene database of choice). (b) Identify genomic variants mapped to the genomic region previously identified in (a). Retrieve allele frequencies for variants for each super population and preferably subpopulation where such data is available. The VWC tools for the 1000 Genomes Project can be used for this step. (c) Filter list of genomic variants from target genomic region to contain only variants classed as either cnon-synonymous’ single nucleotide polymorphisms (SNPs) or genomic ‘insertions or delections’ (indels). Filter furtherto include those that are present in exonic sequences only. Non-synonymous refers to nucleotide variation that produces amino acid variation (ie, excluding silent mutations). (d) Correlate population frequency data for each of the identified variants for each of the super populations (for example ‘European Ancestry’, ‘East Asian ancestry’, ‘West African ancestry’, ‘Americas’, and ‘South Asian ancestry’) to identify those variants that segregate with less than two super-populations. Further correlate all identified variants with each of the sub-populations (for example, ‘European ancestry’ super-population might be subdivided into groups such as ‘CEU · Utah residents with Northern or Western European ancestry’, ‘TSI Toscani in Italia’ and ‘British from England and Scotland’) and produce a second score for rarity of variants within a super-population. (e) Collect one or more sequences that show segregation to specific sub-populations for use in the present invention, eg, according to selection criteria as described herein. id="p-128" id="p-128"
[00128] Human Populations id="p-129" id="p-129"
[00129] Optionally the ethnic populations are selected from those identified in the 1000 Genomes Project database. In this respect, see Table 12 which provides details of the ethnic populations on which the 1000 Genomes Project database is based. id="p-130" id="p-130"
[00130] 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: [00131] African ancestry (Mbuti Pygmies, Biaka Pygmies, San peoples, and speakers of Niger-Kordofanian languages (Bantu, Yoruba or Mandenka populations), id="p-132" id="p-132"
[00132] Eurasian ancestry (European ancestry (Orcadian, Adygei, Basque, French, Russians, Italians, Sardinian, Tuscan), Middle Eastern ancestry (Mozabite, Bedouin, Druze, Palestinians), Central/South Asian ancestry (Balochi, Brahul, Makrani, Sindhi, Pathan, Burusho, Hazara. Uygur, Kalash)), [00133] East Asian ancestry (Han, Dal, Daur, Hezhen, Lahu, Miao, Oroqen, She, Tujia, Tu, Xibo, Yi, Mongola, Naxi, Cambodian, Japanese, Yakut), Oceanic ancestry (Melanesian, Papuan); or id="p-134" id="p-134"
[00134] Americas ancestry (Karitiana, Surui, Colombian, Maya, Pima). id="p-135" id="p-135"
[00135] The International HapMap Project, Nature, 2003 Dec !8;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:19 id="p-136" id="p-136"
[00136] Utah population with Northern or Western European ancestry (samples collected in 1980 by the Centre d’Etude du Polymorphisme Humain (CEPH)); population with ancestry of Yoruba people from Ibadan, Nigeria; population with Japanese ancestry; and population with ancestry of Han Chinese from China. id="p-137" id="p-137"
[00137] The authors, citing earlier publications, suggest that ancestral geography is a reasonable basis for sampling human populations. |00138] A suitable sample of human populations used in the present invention is as follows:- (a) European ancestry (b) Northern European ancestry; Western European ancestry; Toscani ancestry; British ancestry, Finnish ancestry or Iberian ancestry. (c) More specifically, population of Utah residents with Northern and/or Western European ancestry; Toscani population in Italia; British population in England and/or Scotland; Finnish population in Finland; or Iberian population in Spain. (a) East Asian ancestry (b) Japanese ancestry; Chinese ancestry or Vietnamese ancestry. (c) More specifically, Japanese population in Toyko, Japan; Han Chinese population in Beijing, China; Chinese Dai population in Xishuangbanna; Kinh population in Ho Chi Minh City, Vietnam; or Chinese population in Denver, Colorado, USA. (a) West African ancestry (b) Yoruba ancestry; Luhya ancestry; Gambian ancestry; or Malawian ancestry. (c) More specifically, Yoruba population in Ibadan, Nigeria; Luhya population in Webuye, Kenya; Gambian population in Western Division, The Gambia; or Malawian population in Blantyre, Malawi. (a) Population of The Americas (b) Native American ancestry; Afro-Caribbean ancestiy; Mexican ancestry; Pueilo Rican ancestry; Columbian ancestry; or Peruvian ancestry . (c) More specifically, population of African Ancestry in Southwest US; population of African American in Jackson, MS; population of African Caribbean in Barbados; population of Mexican Ancestry in Los Angeles, CA; population of Puerto Rican in Puerto Rico: population of Colombian in Medellin, Colombia; or population of Peruvian in Lima, Peru. (a) South Asian ancestry (b) Ahom ancestry; Kayadtha ancestry; Reddy ancestry; Maratha; or Punjabi ancestry. (c) More specifically, Ahom population in the State of Assam, India; Kayadtha population in Calcutta, India; Reddy population in Hyderabad, India; Maratha population in Bombay, India; or Punjabi population in Lahore, Pakistan. [00139J In any configuration of the invention, in one embodiment, each human population is selected from a population marked "(a)" above. id="p-140" id="p-140"
[00140] In any configuration of the invention, in another embodiment, each human population is selected from a population marked "(b)" above. id="p-141" id="p-141"
[00141] In any configuration of the invention, in another embodiment, each human population is selected from a population marked "(c)" above. id="p-142" id="p-142"
[00142] In one embodiment 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. id="p-143" id="p-143"
[00143] In one embodiment 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 ancestry7; 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 with Peruvian ancestry; or an ethnic population with Ahom ancestry; or an ethnic population with Kayadtha ancestiy; or an ethnic population with Reddy ancestry; or an ethnic population with Maratha; or an ethnic population with Punjabi ancestiy. id="p-144" id="p-144"
[00144] Anti-Target Ligands id="p-145" id="p-145"
[00145] 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, e.g., 1L6R. For example, the ligand specifically binds to a TOI, e.g., IL6R variant as per the invention. The ligand may inhibit or antagonise the activity of the TOI, e.g., IL6R target, eg, the ligand neutralises the target. The skilled person will be familiar with neutralising ligands in genera], 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. id="p-146" id="p-146"
[00146] In an example, the ligand is (or has been determined as) a neutraliser of the TOI, e.g., IL6R. In an example, determination is carried out in a human (eg, in a clinical trial). In an example, 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). id="p-147" id="p-147"
[00147] An antibody "fragment" comprises a portion of an intact antibody, preferably the antigen binding and/or the variable region of the intact antibody. Examples of 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. id="p-148" id="p-148"
[00148] In an embodiment, 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, e.g., anti-IL6R or IL6R-binding or targeting, antibodies and fragments can be prepared according to any known method, eg, using transgenic mice (eg, the Kymouse™ or Velocimouse™, or Omnimouse™ , Xenomouse™, HuMab Mouse™ or MeMo Mouse™), rats (eg, the Omnirat™), camelids, sharks, rabbits, chickens or other non-human animals immunised with the TOI, e.g., 1L6R, followed optionally by humanisation of the constant regions and/or variable regions to produce human or humanised antibodies. In an example, 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. Representative examples of suitable technologies are described in US20120093818 (Amgen, Inc), which is incorporated herein by reference, eg, the methods set out below herein. Although this is with reference to PCSK9, the antibody-generating methods can be applied to other TOIs as per the broadest scopes of the present invention. id="p-149" id="p-149"
[00149] Generally, a VELOCIMMUNE™ 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. 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 ofthe heavy chain and light chain. Such an antibody protein may be produced in a cell, such as a CHO cell. Alternatively, 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. id="p-150" id="p-150"
[00150] Initially, high affinity chimaeric antibodies are isolated having a human variable region and a mouse constant region. As described below, 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 lgG4 (for example, SEQ ID NO: 751,752,753 in US2011/0065902 (which is incorporated by reference herein in its entirety), which sequences are incorporated herein by reference for use in the ligands of the present invention). While the constant region selected may vary according to specific use, high affinity antigen-binding and target specificity characteristics reside in the variable region. id="p-151" id="p-151"
[00151] In an example, the ligand of the invention is or comprises a nucleic acid, eg, RNA, eg, siRNA that hybridises under stringent condition to the TOI, e.g., IL6R, variant sequence, eg, hybridises a nucleotide sequence comprising one or more nucleotides that are variant (versus the most common IL6R sequence, eg, with reference to the 1000 Genomes Project database). id="p-152" id="p-152"
[00152] For example, the nucleic acid hybridises to a region immediately flanking a nucleotide that is variant compared to the corresponding nucleotide of the 1L6R nucleotide sequence having the highest cumulative human allele frequency and/or the highest total human genotype frequency. In an example, the nucleic acid hybridises to at two or more such variant nucleotides. [00153] Specific hybridisation is under stringent conditions, as will be apparent to the skilled person, eg, conditions of 5xSSC, 5*Denhardt's reagent, and 0.5% SDS at 65° C. id="p-154" id="p-154"
[00154] Target binding ability, specificity and affinity (Kd, KOff and/or Kon) can be determined by any routine method in the art, eg, by surface plasmon resonance (SPR). The term "Kd", as used herein, is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction. id="p-155" id="p-155"
[00155] In one embodiment, the surface plasmon resonance (SPR) is carried out at 25°C.
In another embodiment, the SPR is carried out at 37°C. id="p-156" id="p-156"
[00156] In one embodiment, 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)). id="p-157" id="p-157"
[00157] In one embodiment, the SPR is carried out at a physiological salt level, eg, 150mM NaCl. id="p-158" id="p-158"
[00158] In one embodiment, 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-20IM) at 0.05% and ED TA at 3mM. id="p-159" id="p-159"
[00159] In one example, the SPR is carried out at 25°C or 37°C in a buffer at pH7.6, I50mM NaCl, 0.05% detergent (eg, P20) and 3mM EDTA. The buffer can contain lOmM Hepes. In one example, 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). id="p-160" id="p-160"
[00160] In an example, the affinity of the ligand (eg, antibody) 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, Biacore™ 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; 3. Passing the test antigen over the chip’s capture surface at 1024nM, 256nM, 64nM, 16nM, 4nM with a OnM (i.e. buffer alone); and 4. And determining the affinity of binding of test antibody to test antigen using surface plasmon resonance, eg, under an SPR condition discussed above (eg, at 25°C in physiological buffer).
SPR can be carried out using any standard SPR apparatus, such as by Biacore™ or using the ProteOn XPR36™ (Bio-Rad®). id="p-161" id="p-161"
[00161] Regeneration of the capture surface can be carried out with 1 OmM glycine at pHl .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 XPR36™ analysis software. id="p-162" id="p-162"
[00162] In an example, 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). In an example, the ligand is in vitro, eg, in a sterile container. In an example, 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, e.g., IL6R. In an example, the instructions indicate that the human should be genotyped for a TOI, e.g., IL6R, variant sequence of the invention before administering the ligand to the human. In an example, the instructions indicate that the human should be phenotyped for a TOI, e.g., IL6R, variant of the invention before administering the ligand to the human. In an example, the human is of Chinese (eg, Han or CHS) ethnicity and the instructions are in Chinese (eg, Mandarin). In an example, the instructions comprise directions to administer alirocumab or evolocumab to said human. In an example, the TOI is IL6R and the instructions comprise directions to administer sarilumab to said human. id="p-163" id="p-163"
[00163] In an example, the ligand is or comprises soluble human gpl30-Fc. id="p-164" id="p-164"
[00164] The invention relates to the concepts set out in the following clauses. id="p-165" id="p-165"
[00165] 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 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, 70T 80, 90 or 95%) said disease or condition, wherein the TOI in said human is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence having a total human genotype frequency of less than 50%; wherein Before 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. id="p-166" id="p-166"
[00166] In any aspect, configuration, example, embodiment, clause or concept herein, frequencies may be determined using bioinformatics. id="p-167" id="p-167"
[00167] In any aspect, configuration, example, embodiment, clause or concept herein, frequencies may be determined by reference to a database comprising at least 1000 or 2000 human sequences. id="p-168" id="p-168"
[00168] In any aspect, configuration, example, embodiment, clause or concept herein "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. id="p-169" id="p-169"
[00169] In any aspect, configuration, example, embodiment, clause or concept herein "homozygous human genotype frequency means the cumulative frequency of two occurrences of the variant allele (homozygous state), eg, genotype in 1000 Genomes Project database. id="p-170" id="p-170"
[00170] In any aspect, configuration, example, embodiment, clause or concept herein "total human genotype frequency" means the total of heterozygous plus homozygous human genotype frequencies. id="p-171" id="p-171"
[00171] In any aspect, configuration, example, embodiment, clause or concept herein "cumulative human allele frequency" refers to the total of all occurrences of the variant allele in the sample or database or in humans, eg, in the 1000 Genomes Project database. id="p-172" id="p-172"
[00172] Clause!: 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. [00173] In an example, the ligand is (or has been determined as) a neutraliser of the TOL In an example, determination is carried out in a human (eg, in a clinical trial). In an example, 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). id="p-174" id="p-174"
[00174] 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 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 encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence having a total human genotype frequency of less than 50%; wherein b. 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. . id="p-175" id="p-175"
[00175] In an example, the ligand is (or has been determined as) a neutraliser of the TOI. In an example, determination is carried out in a human (eg, in a clinical trial). In an example, 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). id="p-176" id="p-176"
[00176] 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%. id="p-177" id="p-177"
[00177] The TOI variant is not the most frequent. id="p-178" id="p-178"
[00178] 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). id="p-179" id="p-179"
[00179] 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). id="p-180" id="p-180"
[00180] Clause 7: The method of any preceding clause, wherein said frequency is less than 10 or 15% (eg, from 1 to 10%). id="p-181" id="p-181"
[00181] In an embodiment, 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%. id="p-182" id="p-182"
[00182] In an embodiment, 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%. id="p-183" id="p-183"
[00183] 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%). id="p-184" id="p-184"
[00184] In an embodiment, 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%. id="p-185" id="p-185"
[00185] In an embodiment, 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 1 5%, 1 to 10%, 1 to about 10% or 1 to 5% or 1 to about 5%. id="p-186" id="p-186"
[00186] 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); wherein b. Before 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%; or Before step (a) said the method comprises genotyping the human as negative for a variant nucleotide sequence having a cumulative human allele frequency of Jess 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%. id="p-187" id="p-187"
[00187] In an embodiment, in (a) 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%). id="p-188" id="p-188"
[00188] In an embodiment, in (a) 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%). id="p-189" id="p-189"
[00189] In an embodiment, in (b) 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%. id="p-190" id="p-190"
[00190] In an embodiment, in (b) 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 I 5%, I to about 15%, 1 to 10%, 1 to about 10% or 1 to 5% or I to about 5%. id="p-191" id="p-191"
[00191] 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. id="p-192" id="p-192"
[00192] In an embodiment, 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. id="p-193" id="p-193"
[00193] In an embodiment, before 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. id="p-194" id="p-194"
[00194] 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. id="p-195" id="p-195"
[00195] 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). id="p-196" id="p-196"
[00196] By "substantially incapable or neutralising or inhibiting" is meant: Neutralisation or inhibition less than 50, 25, 105 5 or 0.5% inhibition or neutralisation of the most frequent TOI variant. [00197] 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. id="p-198" id="p-198"
[00198] 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%. id="p-199" id="p-199"
[00199] In an embodiment, each TOf 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%). id="p-200" id="p-200"
[00200] In an embodiment, 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%). |00201| 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 14. [00202] 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. In an embodiment, the database is the 1000 Genomes Project database as described herein. id="p-203" id="p-203"
[00203] Clause 17: An anti-human TOI ligand for use in a method of treating and/or preventing a TOLmediated 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. id="p-204" id="p-204"
[00204] In the alternative, 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. [00205] In an embodiment, 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%. id="p-206" id="p-206"
[00206] In an embodiment, 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%. id="p-207" id="p-207"
[00207] 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. id="p-208" id="p-208"
[00208] In the alternative, clause 18 provides a ligand that binds a human TOI comprising an amino acid sequence encoded by a TOT nucleotide sequence having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of Jess 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. [00209] 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. id="p-210" id="p-210"
[00210] 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%. id="p-211" id="p-211"
[00211] 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 total human genotype frequency of less than 50%. [00212] 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%. [00213) 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%; optionally wherein 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). id="p-214" id="p-214"
[00214] 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%. [00215] 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. id="p-216" id="p-216"
[00216] Clause 25: The ligand of clause 24, wherein the ligand is an antibody or antibody fragment. (00217] 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. id="p-218" id="p-218"
[00218] In an embodiment, 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. id="p-219" id="p-219"
[00219] 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 14)). In an example, numbers are with reference to the 1000 Genomes Project database. id="p-220" id="p-220"
[00220] 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 20 individuals distributed across at least 2 said different ethnic populations (eg, found in at least 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). In an example, numbers are with reference to the 1000 Genomes Project database. id="p-221" id="p-221"
[00221] 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 mai’keting authorisation number (eg, an FDA or EMA authorisation number); optionally wherein the kit comprises an injection pen or IV container that comprises the ligand. id="p-222" id="p-222"
[00222] In an example, 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. id="p-223" id="p-223"
[00223] 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 TOl-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. |00224] In an embodiment of any aspect herein, the antibody, fragment or binding site is recombinant. id="p-225" id="p-225"
[00225] In the alternative, 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. id="p-226" id="p-226"
[00226] 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. id="p-227" id="p-227"
[00227] Clause 31: A kit for TOI genotyping a human, wherein 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 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 id="p-228" id="p-228"
[00228] 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. id="p-229" id="p-229"
[00229] Clause 31: A kit for TOI genotyping a human, wherein 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 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. id="p-230" id="p-230"
[00230] For example, 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. In an example, the nucleic acid hybridises to at two or more such variant nucleotides. (00231] Specific hybridisation is under stringent conditions, as will be apparent to the skilled person, eg, conditions of 5*SSC, 5xDenhardt's reagent, and 0.5% SDS at 65° C. id="p-232" id="p-232"
[00232] 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. id="p-233" id="p-233"
[00233] For example, 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. In an example, the ligand specifically binds to an epitope comprising two or more such variant amino acids. In an example, specific binding means binding with an affinity (Kd) of ImM, ΙΟΟηΜ, lOnM or InM or less, eg, as determined by SPR. id="p-234" id="p-234"
[00234] The term "epitope" 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. In certain embodiments, 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. [00235] 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%. id="p-236" id="p-236"
[00236] 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. id="p-237" id="p-237"
[00237] The use of clause 33 or 34, wherein the ligand, human, disease or condition is according to any one of clauses 1 to 27. id="p-238" id="p-238"
[00238] 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. id="p-239" id="p-239"
[00239] 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%. id="p-240" id="p-240"
[00240] 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%. id="p-241" id="p-241"
[00241] In an example, the method comprises obtaining a TOI nucleic acid sample from the human and then carrying out the identifying step. id="p-242" id="p-242"
[00242] 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%. id="p-243" id="p-243"
[00243] In an example, the method comprises obtaining a TOI protein sample from the human and then carrying out the identifying step. id="p-244" id="p-244"
[00244] 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. id="p-245" id="p-245"
[00245] 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. id="p-246" id="p-246"
[00246] 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. id="p-247" id="p-247"
[00247] 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. id="p-248" id="p-248"
[00248] 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 10% and/or a total human genotype frequency from 1 to about 15% or from 1 to 15%. id="p-249" id="p-249"
[00249] Clause 44: The method, ligand, composition, kit or use of any preceding clause wherein 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. id="p-250" id="p-250"
[00250] For example, the TOI is human PCSK9, eg, a mature, cleaved, autocatalysed or active PCSK9. In an example, the disease is a cardiovascular disease such as hyperlipidaemia. id="p-251" id="p-251"
[00251] 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. id="p-252" id="p-252"
[00252] The invention encompasses anti-TOl (eg, PCSK9 or IL6R) antibody ligands having a modified glycosylation pattern. In some applications, 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). In other applications, modification of galactosylation can be made in order to modify complement dependent cytotoxicity (CDC). id="p-253" id="p-253"
[00253] The invention addresses the need to treat humans having naturally-occurring rarer natural IL6R alleles, genotypes and phenotypes (rarer protein forms). In this respect, the invention provides the configurations and embodiments described above, as well as the following examples. [00254] In an embodiment of any configuration, example, embodiment or aspect herein, the ligand, antibody, fragment or binding site of the invention is recombinant. id="p-255" id="p-255"
[00255] In an example, the ligand has been or is determined as capable of specifically binding a human IL6R that comprises a mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. [00256] In an example, the ligand binds (or has been determined to bind) a first human 1L6R that comprises a mutation Asp358Ala and a second human IL6R that comprises a mutation Val385Ile in SEQ ID NO: 78, wherein the IL6Rs are different. In an embodiment, the ligand binds said IL6R when in complex with human gp 130 and/or human IL6. The complex can be a cell-surface bound on human cells or it can be in solution. id="p-257" id="p-257"
[00257] In an example, the ligand comprises a protein domain that specifically binds to a human IL6R that comprises a mutation Asp358Ala or Val385He in SEQ ID NO: 78 id="p-258" id="p-258"
[00258] 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 1 *10"6 M or less (e.g., a smaller KD denotes a tighter binding). Methods for determining whether two ules 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 IL6R may, however, exhibit cross-reactivity to other antigens such as an IL6R molecule from another species. Moreover, multi-specific antibodies (e.g., bispecifics) that bind to human IL6R and one or more additional antigens are nonetheless considered antibodies that "specifically bind" IL6R, as used herein. id="p-259" id="p-259"
[00259] In an example, the ligand comprises or consists of a protein that mimics human IL6 or gp!30. id="p-260" id="p-260"
[00260] In an example, the ligand antagonises said human IL6R that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78. id="p-261" id="p-261"
[00261] In an example, the method comprises (before administering the ligand) the step of determining that the ligand is capable of binding said human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-262" id="p-262"
[00262] In an example, binding is determined by SPR. In an example, binding is determined by ELISA. |00263] In an example, said said IL6R is soluble IL6R. |00264] In another example, said IL6R is human cell-surface bound IL6R. id="p-265" id="p-265"
[00265] The terms "is determined", "is genotyped" or "is phenotyped" and the like herein mean that the method comprises a step of such determining, genotyping or phenotyping. id="p-266" id="p-266"
[00266] In an example, the human has been or is genotyped as positive for an IL6R nucleotide sequence encoding said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-267" id="p-267"
[00267] In an example, the human has been or is phenotyped as positive for a human IL6R that comprises a mutation Asp358Ala or VaJ385Ue in SEQ ID NO: 78. |00268] In an example, the method comprises genotyping the human as positive for a nucleotide sequence encoding said mutation Asp358Ala or Val385He in SEQ ID NO: 78. id="p-269" id="p-269"
[00269] In an example, the method comprises phenotyping the human has positive for a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-270" id="p-270"
[00270] In an example, the human has been or is genotyped as heterozygous for a nucleotide sequence encoding said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-271" id="p-271"
[00271] "Heterozygous" here means that in the human's genotype one allele comprises a nucleotide sequence encoding said mutation Asp358Ala or Val3 851 Ie in SEQ ID NO: 78 and other allele can be any IL6R (eg, the most common form, SEQ ID NO:79, or an allele comprising a nucleotide sequence encoding said mutation Asp358Ala or Val3851le in SEQ ID NO: 78 that is different from the other allele). id="p-272" id="p-272"
[00272] In an example, the method comprises (before administering the ligand) genotyping the human as heterozygous for a nucleotide sequence encoding said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; optionally also genotyping the human as comprising the nucleotide sequence of SEQ ID NO: 9. id="p-273" id="p-273"
[00273] In an example, the genome of the human has been or is genotyped as homozygous for a nucleotide sequence selected encoding said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. ’ id="p-274" id="p-274"
[00274] "Homozygous" here means that in the human’s genotype each allele comprises the same nucleotide sequence selected encoding said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-275" id="p-275"
[00275] In an example, the method comprises genotyping the human as homozygous for a nucleotide sequence encoding said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-276" id="p-276"
[00276] In an example, the ligand comprises an antibody binding site that binds a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and optionally has been or is determined as capable of such binding. id="p-277" id="p-277"
[00277] In an example, the method comprises (before administering the ligand) the step of determining that the ligand is capable of binding to said human IL6R. id="p-278" id="p-278"
[00278] In an example, the binding is specific binding. In an example, the ligand binds (or has been determined as binding) to the IL6R with an affinity (Kd) of ImM, WOnM, lOnM or InM or less. In an embodiment, the affinity is no less than 10, 100 or 1000 fM. id="p-279" id="p-279"
[00279] In an example, binding or affinity is determined by SPR or ELISA. id="p-280" id="p-280"
[00280] In an example, the disease or condition is mediated by a human 1L6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-281" id="p-281"
[00281] In an example, in any context herein the IL6R is complexed with a human gp!30, eg, soluble gp 130 or human cell surface-bound gpl30. id="p-282" id="p-282"
[00282] In an example, the IL6R is bound to human IL6. id="p-283" id="p-283"
[00283] In an example, the ligand is an antibody or antibody fragment. For example, the antibody or antibody fragment is an JL6R antagonist, eg, neutralises 1L6R (eg, when in such a complex with human gp!30 or bound to human IL6 as described above). Examples of such antibodies are disclosed, for instance, in US8043617 or any other reference listed in Table 13, the disclosures and sequences of such antibodies being incorporated herein in their entireties by refere for use in the invention. One specific example is sarilumab, or an IL6R-binding derivative thereof. id="p-284" id="p-284"
[00284] Advantageously, the ligand is or comprises sarilumab. id="p-285" id="p-285"
[00285] In an example, the ligand comprises or consists of a neutralizing antibody that binds to the IL6R, wherein the antibody binds to IL6R and reduces the likelihood that IL6R binds to human IL6 and/or human gp!30. id="p-286" id="p-286"
[00286] In an example, the ligand is an IL6R antagonist, eg, neutralises IL6R. (00287] An example is provided wherein (i) the ligand comprises a sequence of contiguous nucleotides that specifically hybridises to a nucleotide sequence that encodes said mutation(s), or specifically hybridises to an antisense sequence or an RNA transcript of said sequence, wherein said sequence of contiguous nucleotides hybridises to a nucleotide sequence that encodes said mutation(s) or hybridises to an antisense sequence or an RNA transcript thereof respectively; and/or (ii) the ligand comprises a sequence of at least 10 contiguous nucleotides of a nucleotide sequence that encodes said mutation(s) or is an antisense sequence or RNA version of said contiguous nucleotides, wherein said sequence of contiguous nucleotides encodes said mutation(s). id="p-288" id="p-288"
[00288] In an example, said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment. id="p-289" id="p-289"
[00289] An anti-TNF alpha treatment" is optionally selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. For example it is adalimumab treatment. For example, it is etanercept treatment. For example, it is infliximab treatment. id="p-290" id="p-290"
[00290] Reference to "IL6R protein that comprises a mutation Asp358Ala or Val385He in SEQ ID NO: 78" refers to an alanine at a position in an IL6R protein corresponding to position 358 as noted in SEQ ID NO: 78 (see Table 15) herein or an isoleucine at a position in an IL6R protein corresponding to position 385 as noted in SEQ ID NO: 78 (in Table 15). Similarly, phrases such as "a Pro at position 72 shown in SEQ ID NO: 83" mean a proline at a position corresponding to position number 72 as in SEQ ID NO: 83 in Table 15. id="p-291" id="p-291"
[00291] In an example, said human is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. id="p-292" id="p-292"
[00292] In an example, said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate and/or an anti-TNF alpha treatment. id="p-293" id="p-293"
[00293] In an example, said disease or condition is an inflammatory disease or condition. id="p-294" id="p-294"
[00294] In an example, said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. id="p-295" id="p-295"
[00295] In an example, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. id="p-296" id="p-296"
[00296] In an example, said disease or condition is cancer. id="p-297" id="p-297"
[00297] In an example, said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. For example, the human has been diagnosed with moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. id="p-298" id="p-298"
[00298] In an example, said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. id="p-299" id="p-299"
[00299] In an example, the antibody or fragment comprises constant domains that are rabbit, chicken or murine, eg, mouse or rat constant domains. id="p-300" id="p-300"
[00300] In an example, the antibody or fragment constant domains are human. The presence of human variable and constant domains is particularly advantageous for matching to human patients, and thus in an emobidiment the ligand is or comprises an antibody or fragment whose variable and constant domains are human. id="p-301" id="p-301"
[00301] In an example, the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. These endode for the Asp358Ala or Val385Ile mutations. Thus, the human can comprise one or both of these SNPs. id="p-302" id="p-302"
[00302] In an example, said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation, or is for said administration. [00303J In an example, the ligand inhibits human IL6R binding to human IL6 and/or human gpl30 and optionally has been or is determined as capable of such inhibition. id="p-304" id="p-304"
[00304] In an example, the method comprises (before administering the ligand) determining that the ligand is capable of such inhibition. id="p-305" id="p-305"
[00305] Inhibition determination is eg, inhibition in a blood or serum sample, at rtp, at pH7, at 37 degrees centigrade and/or under the physiological conditions of a human body. id="p-306" id="p-306"
[00306] In an example, the ligand is for treating, reducing the risk of or preventing an IL6R-mediated disease or condition in a human (i) whose genome comprises a nucleotide sequence encoding said 1L6R protein comprising mutation Asp358Ala in SEQ ID NO: 78 and wherein the human is of KHV, GBR, CHB, CDX, CLM, MXL, CHS, JPT, GIH, PUR, ESN, FIN, ACB, BEB, YRI, ASW, ITU, PJL, TSI, PEL, MSL, LWK, STU, GWD, IBS or CEU ancestry, eg, the human is of YRI, ASW, GBR, TSI, CLM, CHB, LWK, CHS, MXL, PUR, JPT, IBS, FIN or CEU ancestry; or (ii) whose genome comprises a nucleotide sequence encoding said IL6R protein comprising mutation Val385Ile in SEQ ID NO: 78 and wherein the human is of ASW, YRI, PEL, MSL, LWK, GWD, PUR, IBS, ESN or ACB ancestry eg, the human is of LWK, ASW, YRI or PUR ancestry. id="p-307" id="p-307"
[00307] In an example, the invention provides pharmaceutical composition or kit for treating, reducing the risk of or preventing an IL6R-mediated condition or disease (eg, as recited above), the composition or kit comprising a iigand of the invention and optionally methotrexate and/or an anti-TNF alpha treatment; and optionally in combination with a label or instructions for use to treat, reduce the risk of or prevent said disease or condition in a human (eg, covering treatment of a human as recited herein); optionally wherein the label or instructions comprise a marketing authorisation number (eg, an FDA or EMA authorisation number); optionally wherein the label or instructions comprise directions to administer sarilumab to said human; optionally wherein the kit comprises an TV or injection device that comprises the ligand (and, eg, also methotrexate or said antiTNF alpha treatment). For example, the invention provides pharmaceutical composition or kit for treating, reducing the risk of or preventing an IL6R-mediated condition or disease (eg, as recited above), the composition or kit comprising a ligand of the invention in combination with a label or instructions for use to treat arthritis in a human, wherein the label or instructions comprise a marketing authorisation number (eg, an FDA or EMA authorisation number), wherein the label or instructions comprise directions covering administration of sarilumab to said human; optionally wherein the kit comprises an IV or injection device that comprises the ligand (and, eg, also methotrexate or said anti-TNF alpha treatment). id="p-308" id="p-308"
[00308] In an example, the invention provides a method of producing an anti-human IL6R antibody binding site, the method comprising obtaining a plurality of anti-IL6R antibody binding sites, screening the antibody binding sites for binding to a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 or a peptide thereof that comprises mutation Asp358Ala or Val3 85He in SEQ ID NO: 78 and isolating an antibody binding site that binds in the screening step, and optionally producing an IL6R-binding fragment or derivative of the isolated antibody. id="p-309" id="p-309"
[00309] In an enbodiment of this and the next example, the plurality of binding sites comprises or consists of a plurality of 4-chain antibodies or fragments thereof, eg, dAbs, Fabs or scFvs. Suitable methods for producing pluralities of binding sites for screening include phage display (producing a phage display library of antibody binding sites), ribosome display (producing a ribosome display library of antibody binding sites), yeast display (producing a yeast display library of antibody binding sites), or immunisation of a non-human vertebrate (eg, a rodent, eg, a mouse or rat, eg, a Velocimouse™, Kymouse™, Xenomouse™, Aliva Mouse™, HuMab Mouse™, Omnimouse™, Omnirat™ or MeMo Mouse™) with an IL6R epitope and isolation of a repertoire of antibodyproducing cells (eg, a B-cell, plasma cell or plasmablast repertoire) and/or a repertoire of isolated antibodies. id="p-310" id="p-310"
[00310] In an example, the method comprises selecting one or more antibody binding sites or a derivative thereof that each specifically binds to a human IL6R epitope comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-311" id="p-311"
[00311] In an example, specific binding means binding with an affinity (Kd) of ImM, ΙΟΟηΜ, lOnM or InM or less, eg, as determined by SPR. id="p-312" id="p-312"
[00312] The term "epitope" 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. In certain embodiments, 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. [00313] In an example the invention provides a method of producing an anti-human IL6R antibody, the method comprising immunising a non-human vertebrate (eg, a mouse or a rat that expresses human antibody variable domains) with a human IL6R comprising a mutation Asp358Ala or Val385He in SEQ ID NO: 78 or a peptide thereof that comprises a mutation Asp358AIa or Val385He in SEQ ID NO: 1 and isolating an antibody that binds a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 or a peptide thereof that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, and optionally producing an IL6R-binding fragment or derivative of the isolated antibody. id="p-314" id="p-314"
[00314] In an example the method further comprises 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. id="p-315" id="p-315"
[00315] In an example the method further comprises isolating a cell (eg, B-cell, plasmablast, plasma cell or memory cell) comprising the nucleic acid, wherein the cell is obtained from a non-human vertebrate that has been immunised with the IL6R epitope. id="p-316" id="p-316"
[00316] In an example the method provides a kit for IL6R genotyping a human, wherein the kit comprises a nucleic acid (i) comprising a sequence of 10 or more (eg, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100 or more) contiguous nucleotides that specifically hybridises to a nucleotide sequence encoding a human IL6R comprising a mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, or specifically hybridises to an antisense sequence or an RNA transcript of said sequence, wherein said sequence of contiguous nucleotides hybridises to at least one nucleotide encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or hybridises to an antisense sequence or an RNA transcript thereof; and/or (ii) comprising a sequence of at least 10 or more (eg, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100 or more) nucleotides of a nucleotide sequence encoding an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. id="p-317" id="p-317"
[00317] In an example the invention provides a kit for IL6R genotyping or phenotyping a human, wherein the kit comprises a ligand according to the invention or an antibody, fragment or derivative produced by the method of the invention. . id="p-318" id="p-318"
[00318] In an example the invention provides the use of an anti-IL6R ligand that binds a human IL6R comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 in the manufacture of a medicament for treating, reducing the risk of or preventing an IL6R-mediated disease or condition in a human whose genome comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally for treating, reducing the risk of or preventing an IL6R-mediated disease or condition in a human as recited herein. id="p-319" id="p-319"
[00319] In an example the invention provides the use of an anti-IL6R ligand that binds a human IL6R comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 in the manufacture of a medicament for targeting said IL6R in a human to treat, reduce the risk of or prevent a disease or condition mediated by IL6R, optionally for targeting IL6R in a human as recited above. [00320] In an example the invention provides a method of targeting an 1L6R for treating, reducing the risk of or preventing an IL6R-mediated disease or condition in a human, the method comprising administering an anti-IL6R ligand to a human comprising a nucleotide sequence encoding an IL6R protein comprising a mutation Asp358Ala or VaI385Ile in SEQ ID NO: 78, whereby an 1L6R encoded by said nucleotide sequence is targeted. id="p-321" id="p-321"
[00321] The ligand can be any anti-IL6R ligand disclosed herein. id="p-322" id="p-322"
[00322] In an example the invention provides a method of treating, reducing the risk of or preventing a disease or condition mediated by IL6R in a human, the method comprising targeting a human IL6R comprising a mutation Asp358Ala orVa!385IIe in SEQ ID NO: 78 by administering to the human a ligand that binds said IL6R thereby treating, reducing the risk of or pi-eventing said disease or condition in the human. id="p-323" id="p-323"
[00323] The ligand can be any anti-IL6R ligand disclosed herein. id="p-324" id="p-324"
[00324] In an example the human has been or is phenotyped as positive for a human IL6R comprising a mutation Asp358Ala or Val385He in SEQ ID NO: 78. In an example the human has been or is genotyped as positive for a nucleotide sequence encoding a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-325" id="p-325"
[00325] In an example the method comprises genotyping the human as positive for a nucleotide sequence encoding an IL6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-326" id="p-326"
[00326] In an example the method comprises phenotyping the human as positive for an 1L6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-327" id="p-327"
[00327] In an example the human has been or is genotyped as heterozygous for a nucleotide sequence encoding an IL6R protein comprising a mutation Asp358Ala or Va!38511e in SEQ ID NO: 78; optionally wherein the human has been or is also genotyped as comprising the nucleotide sequence of SEQ ID NO: 79. id="p-328" id="p-328"
[00328] In an example the genome of the human has been or is genotyped as homozygous for a nucleotide sequence selected encoding an IL6R protein comprising a mutation Asp358Ala or Val38511e in SEQ ID NO: 78. id="p-329" id="p-329"
[00329] In an example the method comprises genotyping the human for a nucleotide sequence encoding an IL6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 before administering the ligand to the human, wherein the ligand is determined to be capable of binding to an IL6R encoded by said selected sequence. id="p-330" id="p-330"
[00330] In an example the method comprises administering said ligand and methotrexate and/or an anti-TNF alpha treatment (eg, infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), or etanercept (eg, Enbrel®) treatment) to the human. In an example the method comprises administering said ligand and adalimumab. In an example the method comprises administering said ligand and etanercept. In an example the method comprises administering said ligand and infliximab. id="p-331" id="p-331"
[00331] In an example the ligand and anti-TNF alpha treatment are administered separately. id="p-332" id="p-332"
[00332] In an example the ligand and anti-TNF alpha treatment are administered simultaneously. id="p-333" id="p-333"
[00333] In an example the ligand is administered by subcutaneous injection. [00334J The invention also provides a method of IL6R genotyping a nucleic acid sample of a human, the method comprising identifying in the sample the presence of a nucleotide sequence encoding an IL6R protein comprising a mutation Asp358Ala or Val3851 le in SEQ ID NO: 78, and optionally administering to the human a ligand of the invention. id="p-335" id="p-335"
[00335] The invention also provides a method of IL6R typing a protein sample of a human, the method comprising identifying in the sample the presence of a human IL6R comprising a mutation Asp358Ala or Val38511e in SEQ ID NO: 78, and optionally administering to the human a ligand of the invention. id="p-336" id="p-336"
[00336] In an example, the method comprises obtaining an IL6R protein sample from the human and then carrying out the identifying step. For example the sample is obtained by obtaining a sample of serum, blood, faeces, hair, tissue, cells, urine or saliva from a human, whereby the nucleic acid or protein sample is obtained and used in the step of identifying said sequence. In an example a ligand according of the invention is used to carry out said identifying step. id="p-337" id="p-337"
[00337] The invention als provides a method of treating, reducing the risk of, or preventing in a human patient an IL6R-mcdiated disease or condition (eg, rheumatoid arthritis), wherein the patient is receiving or has previously received an anti-TNF alpha treatment for said disease or condition, the method comprising typing the patient using a method of the invention as described above and administering a ligand of the invention to the human, whereby the human is treated or said disease or condition is prevented; optionally also reducing or stopping anti-TNF alpha treatment. In an embodiment, the ligand is co-administered with methotrexate. (00338] In an example, said reducing or stopping comprises reducing the dose and/or dosing frequency of anti-TNF alpha treatment. id="p-339" id="p-339"
[00339] The invention also provides a diagnostic, therapeutic or prophylactic kit comprising a ligand that is capable of binding to or has been or is determined as capable of binding to an IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and instructions for carrying out a method of the invention and/or a label or instructions indicating or covering administration of the ligand to a human as per the invention (eg, a human comprising a nucleotide sequence encoding an 1L6R comprising a mutation Asp358Ala or Val385He in SEQ ID NO: 78). id="p-340" id="p-340"
[00340] The invention also provides a diagnostic, therapeutic or prophylactic kit comprising a nucleic acid probe comprising a nucleotide sequence that specifically hybridises to a nucleotide sequence encoding an LL6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 or an antisense sequence or RNA transcript thereof and instructions for carrying out a method of the invention. (00341] In embodiments of any of the configuration, examples or aspects described herein, optionally, the 1L6R is human LL6R, eg, a soluble or cell-surface bound human ]L6R. In an example, the disease is an inflammatory disease such as rheumatoid arthritis. |00342] In examples of the present invention, the ligand specifically binds to human IL6R that comprises a mutation Asp358Ala in SEQ ID NO: 78. In examples of the present invention, the ligand specifically binds to human IL6R that comprises a mutation Val38511e in SEQ ID NO: 78. id="p-343" id="p-343"
[00343] Determination of such binding can be performed by any antibody binding test as known in the art, eg, by surface plasmon resonance. Binding to each such form is, for example, respectively with a Kd of at least ImM, ΙΟΟηΜ, InM, lOOpM, lOpM or lpM, id="p-344" id="p-344"
[00344] In an example, the ligand binds human IL6R that comprises a mutation Asp358Ala in SEQ ID NO: 78, wherein the ligand binding to said IL6R is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to human IL6R comprising SEQ ID NO: 78. id="p-345" id="p-345"
[00345] In an example, the ligand binds human IL6R that comprises a mutation Val385lie in SEQ ID NO: 78, wherein the ligand binding to said IL6R is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to human IL6R comprising SEQ ID NO: 78. id="p-346" id="p-346"
[00346] In examples of the present invention, the ligand neutralises human IL6R that comprises a mutation Asp358Ala in SEQ ID NO: 78 and optionally also human IL6R comprising SEQ ID NO: 78. In examples of the present invention, the ligand neutralises human IL6R that comprises a mutation Val385Ile in SEQ ID NO: 78 and optionally also human IL6R comprising SEQ ID NO: 78. Determination of neutralisation can be performed, for example, by any neutralisation assay method disclosed in US8043617 (Regeneron Pharmaceuticals, Inc), eg, as disclosed in Example therein, or disclosed in any of the patents or applications in Table 13. Ligands of the invention that bind or target IL6R are useful, for example, for therapeutic and prophylactic applications disclosed in US8043617 or disclosed in any of the patents or applications in Table 13, these specific disclosures being incorporated herein by reference in their entirety for use in the present invention and for possible inclusion in claims herein. id="p-347" id="p-347"
[00347] In embodiments where the ligand is used for therapeutic applications, ligand of the invention can inhibit, interfere with or modulate one or more biological activities of an IL6R (eg, human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and optionally also human IL6R comprising SEQ ID NO: 78). In one embodiment, ligand binds specifically to human IL6R (eg, human IL6R that comprises a mutation Asp358Ala or Val385He in SEQ ID NO: 78 and optionally also human IL6R comprising SEQ ID NO: 78) and/or substantially inhibits binding of human IL6R (eg, human IL6R that comprises a mutation Asp358Ala or Val385He in SEQ ID NO: 78 and optionally also human IL6R comprising SEQ ID NO:78) to human IL6 and/or human gpl30 by at least 20%, eg, 20%-40%, 40-60%, 60-80%, 80-85%, or more (for example, by measuring binding in an in vitro competitive binding assay). In an example, the ligand is or comprises an antibody or IL6R-binding fragment thereof (eg, a Fab or scFv). id="p-348" id="p-348"
[00348] In an embodiment, the ligand has a Kd of less (binding more tightly) than 10 7, 8, 10 9, 1010, 1011, 10"12, 10 13 M for binding to one or more of a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and optionally also human IL6R comprising SEQ ID NO: 78. In an example, Kd is determined using SPR. id="p-349" id="p-349"
[00349] In an embodiment, the ligand has an IC50 for blocking the binding of human IL6 to one or more of a human IL6R that comprises a mutation Asp358Ala or Val385Ue in SEQ ID NO: 78 (and optionally also human 1L6R comprising SEQ ID NO: 78) of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM. id="p-350" id="p-350"
[00350] In an embodiment, the ligand has an IC50 for blocking the binding of human gp 130 to one or more of a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 (and optionally also human IL6R comprising SEQ ID NO: 78) of less than 1 microM, 1000 nM to 100 nM, lOOnMto 10 nM, lOnMto 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM. id="p-351" id="p-351"
[00351] In an embodiment, the ligand has an IC50 for blocking the binding of human 1L6 to human IL6R comprising SEQ ID NO: 78 that is no more than 1000, 100, 90, 80, 70, 60, 50, 40, 30, 20 or 10-fold (ie, more inhibitory) than the IC50 for blocking the binding of human IL6 to one or more of a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. Additionally or alternatively, for example, the ligand has an IC50 for blocking the binding of human IL6 to (i) human IL6R comprising SEQ ID NO: 78 of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM, eg, in the range of ImM to IpM (eg, ImM to ΙΟΟρΜ; lOnM to lOOpM; InM to lOpM; or lOOpM to IpM) and (ii) one or more more of a human 1L6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 of less than 1 microM, 1000 nM to 100 nM, 100 nMto 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to I pM, eg, in the range of ImM to IpM (eg, ImM to ΙΟΟρΜ; lOnM to lOOpM; InM to lOpM; or lOOpM to IpM). id="p-352" id="p-352"
[00352] In an embodiment, the ligand has an 1C50 for blocking the binding of human gp 130 to human IL6R comprising SEQ ID NO: 78 that is no more than 1000, 100, 90, 80, 70, 60, 50, 40, 30, 20 or 10-fold (ie, more inhibitory) than the IC50 for blocking the binding of human gpl30 to one or more of a human JL6R that comprises a mutation Asp358Ala or Val385 lie in SEQ ID NO: 78. Additionally or alternatively, for example, the ligand has an 1C50 for blocking the binding of human gp!30 to (i) human IL6R comprising SEQ ID NO: 78 of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM, eg, in the range of 1 mM to IpM (eg, ImM to lOOpM; lOnM to lOOpM; InM to lOpM; or lOOpM to IpM) and (ii) one or more more of a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM, eg, in the range of ImM to IpM (eg, ImM to lOOpM; lOnM to ΙΟΟρΜ; InM to lOpM; or ΙΟΟρΜ to IpM). id="p-353" id="p-353"
[00353] In an embodiment, the ligand binds to human IL6R comprising SEQ ID NO: 78 with a binding affinity that is greater than up to 10%, greater than up to 20%, greater than up to 40%, greater than up to 50%, greater than up to 55%, greater than up to 60%, greater than up to 65%, greater than up to 70%, greater than up to 75%, greater than up to 80%, greater than up to 85%, greater than up to 90%, greater than up to 95% or greater than up to 100% (ie, affinity is double, so Kd is half) relative to binding to a human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. Such binding measurements can be made using a variety of binding assays known in the art, eg, using surface plasmon resonance (SPR), such as by Biacore™ or using the ProteOn XPR36™ (Bio-Rad®), or using KinExA® (Sapidyne Instruments, Inc). id="p-354" id="p-354"
[00354] In one embodiment, the surface plasmon resonance (SPR) is carried out at 25°C.
In another embodiment, the SPR is carried out at 37°C. id="p-355" id="p-355"
[00355] In one embodiment, 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)). id="p-356" id="p-356"
[00356] In one embodiment, the SPR is carried out at a physiological salt level, eg, 150mMNaCI. . id="p-357" id="p-357"
[00357] In one embodiment, 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. id="p-358" id="p-358"
[00358] In one example, the SPR is carried out at 25°C or 37°C in a buffer at pH7.6, 150mM NaCI, 0.05% detergent (eg, P20) and 3mM EDTA. The buffer can contain lOmM Hepes. In one example, 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). id="p-359" id="p-359"
[00359] In an example, the affinity of the ligand, which is an antibody, is determined using SPR by 1. Coupling anti-mouse (or other relevant vertebrate) IgG (eg, Biacore BR-1008-38) to a biosensor chip (eg, GLM chip) such as by primary amine coupling; 2. Exposing the anti-mouse IgG (vertebrate antibody) to a test IgG antibody to capture test antibody on the chip; 3. Passing the test antigen over the chip’s capture surface at 1024nM, 256nM, 64nM, 16nM, 4nM with a OnM (i.e. buffer alone); and 4. And determining the affinity of binding of test antibody to test antigen using surface plasmon resonance, eg, under an SPR condition discussed above (eg, at 25°C in physiological buffer). SPR can be carried out using any standard SPR apparatus, such as by Biacore™ or using the ProteOn XPR36™ (Bio-Rad®). id="p-360" id="p-360"
[00360] Regeneration of the capture surface can be carried out with lOmM glycine at pH] .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 XPR361M analysis software. |00361] In an embodiment, assaying or testing of a ligand of the invention is carried out at or substantially at pH7 (eg, for in vitro tests and assays) and at or substantially at rtp. |00362] In one example an anti-IL6R antibody ligand of the present invention has of an IgG2 heavy chain constant region comprising or consisting of the amino acid sequence as shown in SEQ ID NO: 154, FIG. 3KK of US20120093818A1, which sequence is incorporated herein by reference. id="p-363" id="p-363"
[00363] In one example an anti-lL6R antibody ligand of the present invention has of an IgG4 heavy chain constant region comprising or consisting of the amino acid sequence as shown in SEQ ID NO: 155, FIG. 3KK of US20120093818A1, which sequence is incorporated herein by reference. id="p-364" id="p-364"
[00364] In an example, the antibody of the invention comprises a human IGHG1*O1 or IGHG2*01 heavy chain constant region. Additionally or alternatively, the antibody of the invention comprises a human IGKC*0I or IGLC1 *01 light chain constant region. In an example, the antibody of the invention comprises a human IGHGl*01 heavy chain constant region and a human 1GKC*O1 light chain constant region. id="p-365" id="p-365"
[00365] In an example, the antibody of the invention comprises the heavy chain constant region of SEQ ID NO: 104 or 106. id="p-366" id="p-366"
[00366] In an example, the antibody of the invention comprises the light chain constant region of SEQ ID NO: 105 or 107. id="p-367" id="p-367"
[00367] In an example, the antibody of the invention comprises SEQ ID NO: 104 or 106.
In an example, the antibody of the invention comprises SEQ ID NO; 105 or 107. id="p-368" id="p-368"
[00368] In examples of the present invention, the ligand is or comprises a fully human antibody (ie, the variable domains and constant domains are human). id="p-369" id="p-369"
[00369] In an example, the ligand of the invention specifically binds to an epitope of a human IL6R comprising at least one amino acid that is not found in SEQ ID NO: 78. For example, the amino acid is 358Ala or 385Ile (numbering as used in SEQ ID NO:78). For example, the ligand of the invention specifically binds to an epitope comprising 35 8 Ala and to an epitope comprising 385Ile. Optionally, the ligand also specifically binds to a human IL6R comprising SEQ ID NO: 78. [00370] Reference is made to US8043617 (Regeneron, Inc) the entire disclosure of which is incorporated herein by reference. This patent application discloses relevant ligands for use in the present invention, as well as examples and methods of producing and testing ligands that can be used with reference to the present invention. For example, in one embodiment, the antibody or antigenbinding portion of the antibody of the invention comprises a heavy chain variable region (HCVR) selected from the group consisting of SEQ ID NO:3, 227, 19, 231, 35, 51,67, 83, 99, 1 15, 131, 147, 239, 241, 163, 179, 235, 195 and 211, or substantially similar sequence thereof, In a more specific embodiment, the antibody or antigen-binding fragment thereof further comprises a light chain variable region (LCVR) selected from the group consisting of SEQ ID NO: 11, 229, 27, 233, 43, 59, 75, 91, 107, 123, 139, 155, 171, 187, 203 and 219, or a substantially similar sequence thereof In specific embodiments, the antibody or antigen-binding fragment thereof comprise HCVR/LCVR pairs selected from the group consisting of SEQ IDNO:3/11; 227/229; 19/27; 231/233; 35/43; 51/59; 67/75; 83/91; 99/107; 115/123; 131/139; 147/155; 239/155; 241; 155; 163/171; 179/187; 235/237; 195/203; and 211/219, or substantially similar sequences thereof (these sequences, SEQ ID NOs and pairs being those disclosed in US8568721, which is incorporated herein by reference as though written herein and for potential inclusion the invention and claims herein). In an example, the human antibody or antigen-binding fragment comprises the complementarity determining region (CDR) sequences of a heavy chain variable region (HCVR) and light chain variable region (LCVR) pair (HCVR/LCVR) set forth in SEQ ID NO:19/27 of US8568721, which are incorporated herein by reference as though written herein and for potential inclusion the invention and claims herein. In an example, the ligand of the invention comprises or consists of an antibody disclosed in any of Tables 1 to 4 of US8568721, the associated disclosures (including sequences) of which are incorporated herein by reference. id="p-371" id="p-371"
[00371] In an example, the ligand is or comprises sarilumab or is an IL6R-binding derivative thereof. id="p-372" id="p-372"
[00372] In an example, the antibody or other ligand of the invention is glycosylated, eg, has human glycosylation (eg, produced by a CHO, Cos or Hek293 cell). In an example, the ligand of the invention is produced in CHO. id="p-373" id="p-373"
[00373] The ligand can be used for the treatment, therapy, prophylaxis and/or diagnosis of one or more diseases or conditions or susceptibility thereto, wherein such diseases or conditions comprise those disclosed in US8043617 (Regeneron Pharmaceuticals, Inc), which disclosure is incorporated herein by reference in its entirety for inclusion in one more claims herein or one or more disease or conditions disclosed herein. id="p-374" id="p-374"
[00374] The ligand can be administered to a human characterised as described in US8043617 (Regeneron Pharmaceuticals, Inc) which disclosure is incorporated by reference herein in its entirety. id="p-375" id="p-375"
[00375] The ligand can be administered in a form or combination disclosed in US8043617 (Regeneron Pharmaceuticals, Inc), which disclosure is incorporated herein by reference. id="p-376" id="p-376"
[00376] The ligand can be used in a method of diagnosis. id="p-377" id="p-377"
[00377] Diagnostic Applications id="p-378" id="p-378"
[00378] In some embodiments, the ligand of the invention is a diagnostic tool. The ligand can be used to assay the amount of IL6R present in a sample and/or subject. As will be appreciated by one of skill in the art, such ligands need not be neutralizing ligands. In some embodiments, the diagnostic ligand is not a neutralizing ligand. In some embodiments, the diagnostic ligand binds to a different epitope than a neutralizing ligand binds to. In some embodiments, the two ligands do not compete with one another. id="p-379" id="p-379"
[00379] In some embodiments, the ligands of the invention are used or provided in an assay kit and/or method for the detection of IL6R in mammalian tissues or cells in order to screen/diagnose for a disease or disorder associated with changes in levels of IL6R. The kit comprises a ligand that binds IL6R and means for indicating the binding of the ligand with IL6R. if present, and optionally IL6R protein levels. Various means for indicating the presence of a ligand can be used. For example, fluorophores, other molecular probes, or enzymes can be linked to the ligand and the presence of the ligand can be observed in a variety of ways. The method for screening for such disorders can involve the use of the kit, or simply the use of one of the disclosed ligands and the determination of whether the ligand binds to IL6R in a sample. As will be appreciated by one of skill in the art, high or elevated levels of IL6R will result in larger amounts of the ligand binding to IL6R in the sample. Thus, degree of ligand binding can be used to determine how much IL6R is in a sample. Subjects or samples with an amount of IL6R that is greater than a predetermined amount (e.g., an amount or range that a person without an IL6R related disorder would have) can be characterized as having an IL6R mediated disorder. id="p-380" id="p-380"
[00380] In some embodiments, the ligand is a non-neutralizing ligand and is used to determine the amount of IL6R in a subject receiving an anti-TNF alpha treatment. id="p-381" id="p-381"
[00381] In some embodiments, an isolated nucleic acid molecule is provided and it encodes the ligand. In some embodiments an expression vector is provided and comprises the nucleic acid molecule. In some embodiments, a pharmaceutical composition is provided and comprises the ligand and a pharmaceutically acceptable carrier. In some embodiments, a method is provided for treating a disease or condition which is ameliorated, improved, inhibited or prevented with an 1L6R antagonist ligand of the invention. The method can comprise administering a therapeutic amount of the pharmaceutical composition or ligand to a subject in need thereof. In some embodiments, the subject is a human subject suffering from an inflammatory disease, eg, rheumatoid arthritis. In some embodiments, a method of receiving treatment or therapy is provided, the method can comprise receiving a ligand thereof at a frequency of once every 60 to 90 days. id="p-382" id="p-382"
[00382] In one aspect, the invention provides a ligand of the invention which is or comprises an human antibody or antigen-binding fragment of a human antibody that specifically binds and inhibits a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 (and optionally a human IL6R comprising SEQ ID NO: 781), characterized by the ability to reduce human IL6 activity in a human by 40-80% over a 24, 60 or 90 day period relative to predose levels, eg, as determined using the assay set out in Example 4 of US8043617 (incorportated herein by reference). [00383] In one embodiment, the antibody or antigen-binding fragment is characterized as exhibiting an enhanced binding affinity (Kd) for hIL6R at pH 5.5 relative to the Kd at pH 7.4, as measured by plasmon surface resonance. In a specific embodiment, the antibody or fragment thereof exhibits at least a 20-fold, at least a 40-fold or at least a 50-fold enhanced affinity for IL6R at an acidic pH relative to a neutral pH, as measured by surface plasmon resonance. id="p-384" id="p-384"
[00384] In one embodiment, the antibody or antigen-binding fragment is characterized as not exhibiting an enhanced binding affinity for IL6R at an acidic pFI relative to a neutral pH, as measured by surface plasmon resonance. In a specific embodiment, the antibody or fragment thereof exhibits a decreased binding affinity at an acidic pH. id="p-385" id="p-385"
[00385] In another embodiment, the antibody or antigen-binding fragment specifically binds a human IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and also specifically binds one or more of cynomolgus monkey, rhesus monkey, mouse, rat and hamster 1L6R (eg, binds human and mouse IL6R, or binds human and cynomolgus monkey 1L6R or binds human and rhesus monkey IL6R). id="p-386" id="p-386"
[00386] In one embodiment, the antibody or antigen-binding fragment binds human and cyno and/or rhesus monkey IL6R, but does not bind mouse, rat or hamster IL6R. id="p-387" id="p-387"
[00387] In one embodiment, the invention comprises an antibody or antigen-binding fragment of an antibody comprising one or more of a heavy chain variable region (HCVR), light chain variable region (LCVR), HCDR1, HCDR2, and HCDR3 disclosed in US8043617, the disclosure of which is incorporated herein by reference in its entirety. id="p-388" id="p-388"
[00388] In an example, the invention features a pharmaceutical composition comprising a ligand of the invention, wherein the ligand comprises or consists of a recombinant human antibody or fragment thereof which specifically binds hIL6R and a pharmaceutically acceptable carrier. In one embodiment, the invention features a composition which is a combination of a ligand of the invention (eg, an antibody or antigen-binding fragment of an antibody), and a second therapeutic agent. The second therapeutic agent may be any agent that is advantageously combined with the ligand of the invention, for example, an anti-inflammatory agent, eg, an anti-TNF alpha agent (eg, antibody) or methotrexate. id="p-389" id="p-389"
[00389] In an example, the invention provides a method for inhibiting hIL6R activity using the anti-lL6R ligand of the invention (eg, an antibody or antigen-binding portion of the antibody of the invention), wherein the therapeutic methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an antibody or antigen-binding fragment of an antibody of the invention. The disorder treated is any disease or condition which is improved, ameliorated, inhibited or prevented by removal, inhibition or reduction of IL6R activity. id="p-390" id="p-390"
[00390] The term "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 (I) 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. Typically, an "isolated7' ligand, antibody, protein etc constitutes al 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. Preferably, 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. [00391] For example, an "isolated" antibody is one that has been identified, separated and/or recovered from a component of its production environment (eg, naturally or recombinantly). Preferably, 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. In preferred embodiments, 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. id="p-392" id="p-392"
[00392] Immunoconjugates id="p-393" id="p-393"
[00393] 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. 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, which is incorporated by reference herein in its entirety. id="p-394" id="p-394"
[00394] Bispecifics id="p-395" id="p-395"
[00395] 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 anti-TOI (e.g., anti-IL6R) mAbs can be linked to or coexpressed with another functional molecule, e.g., another peptide or protein. For example, 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 multi specific antibody with a second binding specificity. id="p-396" id="p-396"
[00396] 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 1g CH3 domain, wherein the first and second 1g 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. In one embodiment, 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 V821 (by IMGT; Q355R, N3845, K392N, V397M, R409K, E419Q, and V422I by EU) in the case of IgG4 antibodies. Variations on the bi-specific antibody format described above are contemplated within the scope of the present invention. id="p-397" id="p-397"
[00397] Therapeutic Administration and Formulations id="p-398" id="p-398"
[00398] The invention provides therapeutic compositions comprising the anti-TOI ligand, eg, antibodies or antigen-binding fragments thereof, of the present invention. The invention provides therapeutic compositions comprising the anti-IL6R ligands, 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. 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. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as L1POFECTFNT™), 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. id="p-399" id="p-399"
[00399] 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. When the ligand, eg, antibody, of the present invention is used for treating various conditions and diseases associated with IL6R in an adult patient, it is advantageous to intravenously administer the ligand or 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. Depending on the severity of the condition, the frequency and the duration of the treatment can be adjusted. id="p-400" id="p-400"
[00400] Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, thus the composition invention provides the ligand 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 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. id="p-401" id="p-401"
[00401] The 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.). id="p-402" id="p-402"
[00402] In certain situations, the pharmaceutical composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201). In another embodiment, polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974). In yet another embodiment, 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). id="p-403" id="p-403"
[00403] 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. As the 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. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared is preferably filled in an appropriate ampoule. A pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe. In addition, with respect to subcutaneous delivery, 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. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty , 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. In 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. id="p-404" id="p-404"
[00404] Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPEN™ (Owen Mumford, Inc., Woodstock, UK), DISETRONIC™ pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25™ pen, HUMALOG™ pen, HUMALIN 70/30™ pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPEN™!, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIOR™ (Novo Nordisk, Copenhagen, Denmark), BD™ pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENT™, OPT1PEN PRO™, OPTIPEN STARLET™, and OPTICLIKT™ (sanofi-aventis, Frankfurt, Germany), to name only a few. Examples of 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 SOLOSTAR™ pen (sanofi-aventis), the FLEXPEN™ (Novo Nordisk), and the KWIKPEN™ (Eli Lilly). id="p-405" id="p-405"
[00405] Advantageously, the pharmaceutical compositions for ora! or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients. Such 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. id="p-406" id="p-406"
[00406] The invention provides therapeutic methods in which the ligand, eg, antibody or antibody fragment, of the invention is useful to treat an inflammatory disease or condition associated with elevated hIL6R and/or IL6R activity. The anti-lL6R ligands, eg, antibodies or antibody fragments, of the invention are particularly useful for the treatment of arthritis. id="p-407" id="p-407"
[00407] Ligands of the invention are useful, for instance, in specific binding assays, for genotyping or phenotyping humans, affinity purification of the TOI, e.g., IL6R, and in screening assays to identify other antagonists of TOI, e.g., IL6R, activity. Some of the ligands of the invention are useful for inhibiting binding of TOI, e.g., IL6R, to a congnate human receptor or protein, or inhibiting TOI, e.g., IL6R, -mediated activities. id="p-408" id="p-408"
[00408] The invention encompasses anti-TOl (eg, anti-IL6R) antibody ligands having a modified glycosylation pattern. In some applications, 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). In other applications, modification of galactosylation can be made in order to modify complement dependent cytotoxicity (CDC). id="p-409" id="p-409"
[00409] In an example, the invention features 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 a TOI (e.g., a rare variant as described herein) (e.g., IL6R) as described herein and a pharmaceutically acceptable carrier. In one embodiment, 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. id="p-410" id="p-410"
[00410] "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. id="p-411" id="p-411"
[00411] In an example, the invention features a method for inhibiting TOI, e.g., IL6R, activity using the anti TOI, e.g., anti-IL6R, 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, e.g., IL6R, activity. id="p-412" id="p-412"
[00412] By the phrase "therapeutically 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). id="p-413" id="p-413"
[00413] 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. id="p-414" id="p-414"
[00414] Genotyping & Phenotyping id="p-415" id="p-415"
[00415] The skilled person will be familiar with techniques that can be used for accurate genotyping and application to the invention. These include the following.
Hybridization-based methods 1.1 Dynamic allele-specific hybridization 1.2 Molecular beacons 1.3 SNP microarrays Enzyme-based methods 2.1 Restriction fragment length polymorphism 2.2 PCR-based methods 2.3 Flap endonuclease 2.4 Primer extension 2.5 5’- nuclease 2.6 Oligonucleotide Ligation Assay Other post-amplification methods based on physical properties of DNA 3.1 Single strand conformation polymorphism 3.2 Temperature gradient gel electrophoresis 3.3 Denaturing high performance liquid chromatography 3.4 High-resolution melting of the entire amplicon 3.5 Use of DNA mismatch-binding proteins 3.6 SNPlex (SNPlex™ is a proprietary genotyping platform sold by Applied Biosystems). id="p-416" id="p-416"
[00416] Next-generation sequencing technologies such as pyrosequencing is also useful. id="p-417" id="p-417"
[00417] Reference is also made to GB2444410A and the genotyping method disclosed therein, which is incorporated herein by reference in its entirety. id="p-418" id="p-418"
[00418] Miniaturized assays, such as microarrays with oligonucleotide reagents immobilized on small surfaces, are frequently proposed for large-scale mutation analysis and highthroughput genotyping (Large-scale identification, mapping, and genotyping of single-nucleotide polymorphisms in the human genome (Wang DG, Fan JB, Siao CJ, Bemo 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. 1998 May 15; 280(5366):1077-82). Other high-throughput methods discriminate alleles by differential hybridization, primer extension, ligation and cleavage of an allele-specific probe (Review Accessing genetic variation: genotyping single nucleotide polymorphisms, Syvanen AC, Nat Rev Genet. 2001 Dec; 2(12):930-42; Review Techniques patents for SNP genotyping, Twyman RM, Primrose SB, Pharmacogenomics. 2003 Jan; 4(1):67-79). id="p-419" id="p-419"
[00419] An approach for a fully automated, large-scale SNP analysis is the ’homogeneous’ assay, i.e. a single-phase assay without separation steps, permitting continual monitoring during amplification. The TaqMan™ assay (Applied Biosystems), originally designed for quantitative realtime PCR, is a homogeneous, single-step assay also used in determination of mutation status of DNA (see, eg, A.A. Komar (ed.); Single Nucleotide Polymorphisms, Methods in Molecular Biology 578, DOI 10.1007/978-1-60327-411-119, Humana Press, a part of Springer Science+Business Media, LLC; and Single Nucleotide Polymorphisms, Methods in Molecular Biology™ Volume 578, 2009, pp 293-306, The TaqMan Method for SNP Genotyping, Gong-Qing Shen et al). The TaqMan SNP Genotyping Assay exploits the 5'-exonuclease activity of AmpliTaq Gold™ DNA polymerase to cleave a doubly labeled probe hybridized to the SNP-containing sequence of ssDNA. 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. id="p-420" id="p-420"
[00420] 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. 2001 Jul; 1 1(7): 1262-8; Assessment of two flexible and compatible SNP genotyping platforms: TaqMan SNP Genotyping Assays and the SNPlex Genotyping System, De la Vega FM, Lazaruk KD, Rhodes MD, Wenz MFI, Mutat Res. 2005 Jun 3; 573(1-2):11135). The results of the assay can be automatically determined by genotyping software provided with real-time thermal cyclers (e.g. IQ software of Bio-Rad, Sequence Detection Software of Applied Biosystems). id="p-421" id="p-421"
[00421] Single nucleotide polymorphisms (SNPs) can be determined using TaqManIM 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: PMC 1440877). 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. id="p-422" id="p-422"
[00422] The skilled person will be familiar with techniques that can be used for accurate phenotyping and application to the invention. These include the use of amino acid sequencing of isolated target protein and comparison of sequences from different variants (eg, with the most common variant). An antibody that specifically and selectively binds in the area of a SNP under stringent conditions can also be used to identify a particular variant. In another method, the genotype is determined and a corresponding amino acid sequence (phenotype) determined, eg, by in silica translation. id="p-423" id="p-423"
[00423] Exemplary TOls id="p-424" id="p-424"
[00424] 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; A1CDA; AW1; A1G1; AKAP1; AKAP2; AIYIH; amyloid-beta; AMHR2; ANGPT1; ANGPT2; ANGPTL3; ANGPTL4; ANPEP; APC; APOCI; AR; Axl; AZGP1 (zinc-a-glycoprotein); B7.1; B7.2; BAD; BAFF; BAG); BA11; BCL2; BCL6; BDNF; BLNK; BLR1 (MDR15); BlyS; BMPI; BMP2; BMP3B (GDF1O); BMP4; BMP6; BMP8; BMPR1A; BMPR1B; BMPR2; BPAG1 (plectin); BRCA1; C19orflO (lL27w); C3; C4A; C5; C5R1; CANT1; CASP1; CASP4; CAV1; CBI; CCBP2 (D6 /JAB61); CCL1 (1-309); CCL11 (eotaxin); CCL13 (MCP-4); CCL15 (MIP-id); CCL16 (HCC-4); CCL17 (TARC); CCL18 (PARC); CCL19 (MIP-3b); CCL2 (MCP-1); MCAF; CCL2O (MIP-3a); CCL21 (MIP-2); SLC; exodus-2; CCL22 (MDC / STC-1); CCL23 (MPIF-1); CCL24 (MPIF-2 1 eotaxin-2); CCL25 (TECK); CCL26 (eotaxin-3); CCL27 (CTACK /1LC); 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 I RA);CCR3 (CKR3 / CMKBR3); CCR4; CCR5 (CMKBR5 / ChemR13); CCR6 (CMKBR6 / CKR-L3 / STRL22 / DRY6); CCR7 (CKR7 / EBI 1); CCR8 (CMKBR8 /TERI /CKR-L1); CCR9 (GPR-9-6); CCRLI (VSHK1): CCRL2 (L-CCR); CD7, CD164; CDI9; CD1C; CD2O; CD200; CD-22; CD24; CD28; CD3; CD37; CD38; CD3E; CD3G; CD3Z; CD4; CD4O; CD4OL; CD44; CD45RB; CD52; CD69; CD72; CD74; CD79A; CD79B; CD8; CD8O; CD8I; CD83; CD86; CD96; CD207; CDH1 (E-cadherin); CDH10; CDH12; CDHI3; CDH I8; CDHI9; CDH20; CDH5; CDH7; CDH8; CDH9; CDK2; CDK3; CDK4; CDK5; CDK6; CDK7; CDK9; CDKN1A (p2IWapl/Cipl); CDKN1B (p27Kipl); CDKNIC; CDKN2A (p!61NK4a); CDKN2B; CDKN2C; CDKN3; CEBPB; CELSR3; CER1; CHGA; CHGB; Chitinase; CHRNG; CHST10; CKLFSF2; CKLFSF3; CKLFSF4; CKLFSF5; CKLFSF6; CKLFSF7; CKLFSF8; CLDN3; CLDN7 (claudin-7); CLN3; CLU (clusterin); CMKLR1; CMKOR1 (RDC1); CNR1; COL18A1; COL1AI; COL4A3; COL6A1; CR2; CRP; CSF1 (M-CSF); CRLF2; CSF2 (GM-CSF); CSF3 (GCSF); CTLA4; CTNNB1 (b-catenin); CTSB (cathepsin B); CX3CL1 (SCYDi); CX3CR1 (V28); CXCR6; CXCL1 (GRO1); CXCL1O (IP-10); CXCL11 (I-TAC / IP-9); CXCL12 (SDFI); CXCL13; CXCL14; CXCL16; CXCL2 (GRO2); CXCL3 (GRO3); CXCL5 (ENA-78 I LIX); CXCL6 (GCP-2); CXCL9 (MIG); CXCR3 (GPR9/CKR-L2); CXCR4; CXCR6 (TYMSTR ISTRL33 1 Bonzo); CYB5; CYC1; CYSLTR1; DAB2TP; DAND5; DES; DKFZp451 JOI 18; DNCL1; DPP4; E2F1; ECGF1; EDG1; EFNAI; EFNA3; EFNB2; EGF; EGFR; ELAC2; ENG; ENO1; ENO2; ENO3; EphA4; EPHB4; EPO; ERBB2 (Her-2); EREG; ERK8; ESRI; ESR2; F3 (TF); FADD; FasL; FASN; FCER1A; FCER2; FCGR3A; FCRL4; FGF; FGF1 (aFGF); FGF10; FGFI1; FGF12; FGF12B; FGF13; FGF14; FGFI6; FGFI 7; FGF18; FGF19; FGF2 (bFGF); FGF2O; FGF21; FGF22; FGF23; FGF3 (int-2); FGF4 (HST); FGF5; FGF6 (HST-2); FGF7 (KGF); FGF8; FGF9; FGFR3; FIGF (VEGFD); FILI (EPSILON); FILI (ZETA); FLJ12584; FLJ25530; FLRT1 (fibronectin); FLT1; FOS; FOSLI (FRA-I); FY (DARC); GABRP (GABAa); GAGEB1; GAGECI; Galectin-3; GALNAC4S65T; GATA3; GDF5; GFII; GGT1; GHR; GM-CSF; GNASI; GNRH1; GPR2 (CCR10); GPR31; GPR44; GPR81 (FKSG8O); GPR87; GPR137C; GRCC10 (CIO); GRP; GSN (Gelsolin); GSTP1; HAVCR1; HAVCR2; HDAC4; EDAC5; HDAC7A; HDAC9; hepcidin; hemojuvelin; HGF; HIF1A; HIP I; histamine and histamine receptors; HLA-A; HLA-DRA; HM74; HMOX1; HUMCYT2A; ICEBERG; ICOS; 1D2; IFN-a; IFNA1; IFNA2; IFNA4; IFNA5; IFNA6; IFNA7; IFNB1; IFNgamma; TFNW1; IGBP1; IGF1; IGF1R; IGF2; IGFBP2; IGFBP3; IGFBP6; IL-1; IL 10; IL 1 ORA; IL1ORB;1LU; 1L11RA; IL-12; IL12A; IL12B; IL12RB1; IL12RB2; 1L13; IL13RA1; IL13RA2; 1L14; 1L15;IL15RA; IL16; 1L17; ILI7B; IL17C; IL17R; 1L18; IL18BP; IL18R1; IL18RAP; 1L19; ILIA; IL1B; 1L1F1O; IL1F5; IL1F6; IL1F7; IL1F8; IL1F9; 1L1HY1; IL1R1; IL1R2; IL1RAP; IL1RAPL1; IL1RAPL2; IL1RL1;ILIRL2 IL1RN; 1L2; 1L20; IL2ORA; IL21R; 1L22; 1L22R; 1L22RA2; 1L23; 1L24; 1L25; 1L26; 1L27; 1L28A; 1L28B; 1L29; 1L2RA; IL2RB; IL2RG; 1L3; 1L30; IL3RA; 1L4; IL4R; 1L5; IL5RA; 1L6; IL6 receptor; 1L6ST (glycoprotein 130); 1L7; TL7R; 1L8; IL8RA; IL8RB; EL8RB; 1L9; IL9R; ILK; INHA; INHBA; INSL3; INSL4; IRAKI; IRAK2; ITGAI; ITGA2; 1TGA3; ITGA6 (a6 integrin); ITGAV; ITGB3; ITGB4 (b4 integrin); JAG1; JAKI; JAK3; JUN; K6HF; KAI1; KDR; MTLG; KLF5 (GC Box BP); KLF6; KLK10; KLKI2; KLK13; KLK14; KLK15; KLK3; KLK4; KLK5; KLK6; KLK9; KRT1; KRT19 (Keratin 19); KRT2A; KRTHB6 (hair-specific type II keratin); LAG3; LAMA5; LEP (leptin); LIGHT; Lingo-p75; LingoTroy; LPS; LRP5; LTA (TNF-b); LTB; LTB4R (GPR16); LTB4R2; LTBR; MACMARCKS; MAG orOmgp: MAP2K7 (c-Jun); MDK; MIB1; midkine; MIF; MIP-2; MK167 (Ki-67); MMP2;MMP9; MS4A1; MSMB; MT3 (metallothionectin-ifi); MTSS 1; MUC 1 (mucin); MYC; MYD88; NCK2; neurocan; Nav1.7; NavL8; NFKB 1; NFKB2; NGFB (NGF); NGFR; NgR-Lingo; NgR-Nogo66 (Nogo); NgR-p75; NgR-Troy; NME1 (NM23A); NOX5; NPPB; NROB1; NROB2; NR1D1;NR1D2; NR1H2; NR1H3; NR1H4; NR1I2; NR1I3; NR2C1; NR2C2; NR2E1; NR2E3; NR2FI; NR2F2; NR2F6; NR3C1; NR3C2; NR4A1; NR4A2; NR4A3; NR5AI; NR5A2; NR6A1; NRP1; NRP2; NT5E; NTN4; ODZ1; OPG; OPRD1; OX40L; 0X40; P2RX7; PAP; PARTI; PATE; PAWR; PC A3; 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 (COX2); PTN; RAC2 (p21Rac2); RARB; RGS1; RGSI3; RGS3; RNF110 (ZNFI44); ROBO2; ROR1; S100A2; SCGB1D2 (lipophilin B); SCGB2A1 (mammaglobin 2); SCGB2A2 (mammaglobin 1); SCYE1 (endothelial Monocyte-activating cytokine); SDF2; SERPINA1; SERPINIA3; SERPINB5 (maspin); SERP1NE1 (PAT-i); SERPINF1; SHBG; SLA2; SLC2A2; SLC33AI; SLC43A1; SLIT2; SPPI; SPRR1B (Spri); ST6GAL1; STAB1; STAT6; STEAP; STEAP2; TB4R2; TBX21; TCP1O; TDGFI; TEK; TGFA; TGFB1; TGFB111; TGFB2; TGFB3; TGFBI; TGFBR1; TGFBR2; TGFBR3; TH IL, THBS1 (thrombospondin-1); THBS2; THBS4; THPO; TIE (Tie-i); TIM3; TMP3; tissue factor; TLR1O; TLR2; TLR3; TLR4; TLR5; TLR6; TLR7; TLR8; TLR9; TMPRSS6; TNF; TNF-a; TNFAIP2 (B94); TNFAIP3; TNFRSF1 1 A; TNFRSF1A; TNFRSF1B; TNFRSF21; TNFRSF5; TNFRSF6 (Fas); TNFRSF7; TNFRSF8; TNFRSF9; TNFSF10 (TRAIL); TNFSFI 1 (TRANCE); TNFSF12 (APO3L); TNFSF13 (April); TNFSF13B; TNFSFI4 (HVEM-L); TNFSFI 5 (VEGI); TNFSFI 8; TNFSF4 (0X40 ligand); TNFSF5 (CD4O ligand); TNFSF6 (FasL); TNFSF7 (CD27 ligand); TNFSF8 (CD3O ligand); TNFSF9 (4-1BB ligand); TOLLIP; Toll-like receptors; TOP2A (topoisomerase lia); TP53; TPM1; TPM2; TRADD; TRAF1; TRAF2; TRAF3; TRAF4; TRAF5; TRAF6; TRAIL; TREM1; TREM2; TRPC6; TSLP; TWEAK; VEGFA; VEGFB; VEGFC; versican; VHL C5; VLA-4; Wnt7A; XCL1 (lymphotactin); XCL2 (SCM-lb); XCR1 (GPR5 /CCXCR1); YY1; and ZFPM2. (00425] In an example, the TOI is a human TOI selected from Table 5. (00426] In an example, the TOI is 0X40 ligand. id="p-427" id="p-427"
[00427] In an example, the TOI is 0X40. id="p-428" id="p-428"
[00428] In an example, the TOI is PCSK9. id="p-429" id="p-429"
[00429] In an example, the TOI is IL6 receptor (IL-6R). id="p-430" id="p-430"
[00430] In an example, the TOI is LIGHT. id="p-431" id="p-431"
[00431] In an example, the TOI is VEGF-A. id="p-432" id="p-432"
[00432] In an example, the TOI is TNF alpha. id="p-433" id="p-433"
[00433] In an example, the TOI is PIGF. id="p-434" id="p-434"
[00434] In an example, the TOI is IGF1R. id="p-435" id="p-435"
[00435] In an example, the TOI is OPG. id="p-436" id="p-436"
[00436] In an example, the TOI is ICOS id="p-437" id="p-437"
[00437] In an example, the TOI is NGF. id="p-438" id="p-438"
[00438] In an example, the TOI is BMP6. id="p-439" id="p-439"
[00439] In an example, the TOI is ferroportin. id="p-440" id="p-440"
[00440] In an example, the TOI is TMPRSS6. id="p-441" id="p-441"
[00441] In an example, the TOI is hemojuvelin. id="p-442" id="p-442"
[00442] In an example, the TOI is VEGF receptor. id="p-443" id="p-443"
[00443] In an example, the TOI is PDGF receptor. id="p-444" id="p-444"
[00444] In an example, the TOI is stem cell factor receptor. id="p-445" id="p-445"
[00445] In an example, the TOI is hepcidin. id="p-446" id="p-446"
[00446] In an example, the TOI is IL-4 receptor alpha. id="p-447" id="p-447"
[00447] Tn an example, the TOI is sclerostin. id="p-448" id="p-448"
[00448] In an example, the TOI is IL-13 receptor. id="p-449" id="p-449"
[00449] In an example, the TOI is CD7. id="p-450" id="p-450"
[00450] In an example, the TOI is delta-like ligand-4 (D114). id="p-451" id="p-451"
[00451] In an example, the TOI is HGF. id="p-452" id="p-452"
[00452] In an example, the TOI is angiopoietin-2 (Ang2). id="p-453" id="p-453"
[00453] In an example, the TOI is GDF8. id="p-454" id="p-454"
[00454] In an example, the TOI is ERBB3. id="p-455" id="p-455"
[00455] In an example, the TOI is IL-17 receptor. id="p-456" id="p-456"
[00456] In an example, the TOI is CD40. id="p-457" id="p-457"
[00457] In an example, the TOI is CD40 ligand. id="p-458" id="p-458"
[00458] In an example, the TOI is EGFR. id="p-459" id="p-459"
[00459] For convenience, the meaning of some terms and phrases used in the specification, examples, and appended claims, are provided below. Unless stated otherwise, or implicit from context, the following terms and phrases include the meanings provided below. The definitions are provided to aid in describing particular embodiments, and are not intended to limit the claimed invention, because the scope of the invention is limited only by the claims. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. If there is an apparent discrepancy between the usage of a term in the art and its definition provided herein, the definition provided within the specification shall prevail. id="p-460" id="p-460"
[00460] For convenience, certain terms employed herein, in the specification, examples and appended claims are collected here. id="p-461" id="p-461"
[00461] The terms "decrease", "reduced", or "reduction" are all used herein to mean a decrease by a statistically significant amount. In some embodiments, "reduce," "reduction" or "decrease" typically means a 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. As used herein, "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." id="p-462" id="p-462"
[00462] In certain aspects of all embodiments of the invention, the term "inhibition" is used. 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. id="p-463" id="p-463"
[00463] The terms "increased", "increase", "enhance", or "activate" are all used herein to mean an increase by a statically significant amount. In some embodiments, 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. In the context of a marker or symptom, an "increase" is a statistically significant increase in such level. id="p-464" id="p-464"
[00464] As used herein, the term "substantially" refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term "substantially0 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, id="p-465" id="p-465"
[00465] As used herein, 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. In some embodiments, 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. In an alternative embodiment, where administration to a human is mentioned in the context of the invention herein, the alternative is that administration is to a different, non-human, subject. Similarly, alternative to a human comprising said nucleotide sequence, such a sequence may be comprised by a non-human subject. This may be useful for the assaying, treatment, prevention or diagnosis in a non-human animal (eg, non-human primate) that comprises one or both of the, e.g., IL6R mutations. id="p-466" id="p-466"
[00466] Preferably, 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. id="p-467" id="p-467"
[00467] 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. Alternatively, 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. For example, 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. id="p-468" id="p-468"
[00468] 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. [00469) As used herein, the terms "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. The terms '’protein", and polypeptide" refer to a polymer of amino acids with natural amino acids. When referring to "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. Thus, 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. id="p-470" id="p-470"
[00470] As used herein, the term "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 singlestranded nucleic acid not derived from any double-stranded DNA. In one aspect, the nucleic acid can be DNA. In another aspect, the 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. id="p-471" id="p-471"
[00471] As used herein, the term "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. In some embodiments, a nucleic acid probe can further comprise a detectable label. In some embodiments, a nucleic acid probe can be attached to a solid surface. In some embodiments, a nucleic acid from is from about 5 nt to about 100 nt in length. id="p-472" id="p-472"
[00472] As used herein, the term "siRNA" refers to a nucleic acid that forms an RNA molecule comprising two individual strands of RNA which are substantially complementary to each other. Typically, 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). In some embodiments, a siRNA can be blunt-ended. In some embodiments, 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. In some embodiments, 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. id="p-473" id="p-473"
[00473] As used herein, "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). PCSK9 is a prohormone-proprotein convertase in the subtilisin (S8) family of serine proteases (Seidah et al., 2003). 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. id="p-474" id="p-474"
[00474] PCSK9 exists as both a pro-form and a mature form. Autocatalysis of the PCSK9 proform occurs between Gin 152 and Seri53 (VFAQ|SIP (SEQ ID NO: 67)) (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 PCSK.9 isoforms are provided herein, see, e.g. SEQ ID NOs: 1-27. id="p-475" id="p-475"
[00475] As used herein, the "catalytic domain" of PCSK9 refers to the portion of a PCSK9 polypeptide conesponding to positions 153 to 449 of PCSK.9, e.g. of SEQ ID NO: 1. As used herein, 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. |00476] As used herein, 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. Non-limiting examples of such diseases or conditions can include, for example, a lipid disorder, hyperlipoproteinemia, hyperlipidemia; dyslipidemia; hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication, type II diabetes, high blood pressure, and a cardiovascular disease or condition. In an example, the disease or condition is an inflammatory or autoimmune disease or condition. Methods of identifying and/or diagnosing such diseases and conditions are well known to medical practitioners of ordinary skill. id="p-477" id="p-477"
[00477] 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 low64 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, id="p-478" id="p-478"
[00478] As used herein, a disease or condition "mediated by IL6R" refers to a disease or condition which is caused by or characterized by a change (eg, increase) in IL6R, e.g. a change in expression level, a change in activity, and/or the presence of a variant or mutation of IL6R. Nonlimiting examples of such diseases or conditions include, for example, an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. For example, moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. Methods of identifying and/or diagnosing such diseases and conditions are well known to medical practitioners of ordinary skill. id="p-479" id="p-479"
[00479] A subject at risk of having or developing a disease or condition mediated by 1L6R 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. comprising one or more genetic polymorphisms known to predispose to the disease or condition, e.g., comprising a nucleotide sequence encoding an IL6R protein comprising a mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 1, or comprising such an IL6R. id="p-480" id="p-480"
[00480] As used herein, "ligand" refers to a molecule which can bind, e.g., specifically bind, to a second molecule or receptor. In some embodiments, a ligand can be, e.g., an antibody, antibody fragment, antibody portion, and/or affibody. id="p-481" id="p-481"
[00481] The term variant" as used herein 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 a i 000 Genomes Project database) by one or more arnino 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. Such conservative substitutions are well known in the art.
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. In some embodiments amino acid substitutions are conservative. Also encompassed within the term variant when used with reference to a 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). id="p-482" id="p-482"
[00482] 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: 1 -27 and in Table L 2 or 6. id="p-483" id="p-483"
[00483] In some aspects, one can use "synthetic variants", "recombinant variants", or "chemically modified" polynucleotide variants or polypeptide variants isolated or generated using methods well known in the art. "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. id="p-484" id="p-484"
[00484] The invention contemplates chemically modified synthetic or recombinant ligand (or ligand-encoding nucleotide sequences) derivatives isolated or generated using methods well known in the art. "Modified derivatives" 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. id="p-485" id="p-485"
[00485] The invention contemplates chemically modified, synthetic or recombinant ligand (or ligand-encoding nucleotide sequences) derivatives isolated or generated using methods well known in the art. "Modified derivatives" 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 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. id="p-486" id="p-486"
[00486] ’’Conservative 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. Thus, a 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)). Conservative substitution tables providing functionally similar amino acids are well known in the art. For example, 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. Freeman and Company (1984), incorporated by reference in its entirety.) In some embodiments, individual substitutions, deletions or additions that alter, add or delete a single amino acid or a small percentage of amino acids can also be considered conservative substitutions if the change does not reduce the activity of the peptide. Insertions or deletions are typically in the range of about 1 to 5 amino acids. The choice of conservative amino acids may be selected based on the location of the amino acid to be substituted in the peptide, for example if the amino acid is on the exterior of the peptide and expose to solvents, or on the interior and not exposed to solvents. id="p-487" id="p-487"
[00487] In alternative embodiments, one can select the amino acid which will substitute an existing amino acid based on the location of the existing amino acid, i.e. its exposure to solvents (i.e. if the amino acid is exposed to solvents or is present on the outer surface of the peptide or polypeptide as compared to internally localized amino acids not exposed to solvents). Selection of such conservative amino acid substitutions are well known in the art, for example as disclosed in Dordo et al, J. Mol Biol, 1999, 217, 721-739 and Taylor et al, J. Theor. Biol. 119(1986);205-218 and S. French and B. Robson, J. Mol. Evol. 19(1983)171. Accordingly, one can select conservative amino acid substitutions suitable for amino acids on the exterior of a protein or peptide (i.e. amino acids exposed to a solvent), 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. id="p-488" id="p-488"
[00488] In alternative embodiments, one can also select conservative amino acid substitutions encompassed suitable for amino acids on the interior of a protein or peptide, for example one can use suitable conservative substitutions for amino acids is on the interior of a protein or peptide (i.e. the amino acids are not exposed to a solvent), for example but not limited to, one can use the following conservative substitutions: where Y is substituted with F, T with A or S, I with L or V, W with Y, M with L, N with D, G with A, T with A or S, D with N, I with L or V, F with Y or L, S with A or T and A with S, G, T or V. In some embodiments, non-conservative amino acid substitutions are also encompassed within the term of variants. id="p-489" id="p-489"
[00489] As used herein an "antibody" includes an IgG (eg, IgGl, 2, 3 or 4), IgM, IgA, IgD or IgE 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-1 inked 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. (00490] As described herein, an antigen" is a molecule that is bound by a binding site on an antibody agent. Typically, 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. The term 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. id="p-491" id="p-491"
[00491] As used herein, the term ‘‘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. In some embodiments, an antibody fragment can comprise a monoclonal antibody or a polypeptide comprising an antigenbinding domain of a monoclonal antibody. For example, 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). In another example, an antibody includes two heavy (H) chain variable regions and two light (L) chain variable regions. The term 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 or 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, human antibodies, humanized antibodies, chimeric antibodies, and the like. id="p-492" id="p-492"
[00492] As used herein, antibody variable domain refers to the portions of the light and heavy chains of antibody molecules that include amino acid sequences of Complementarity Determining Regions (CDRs; ie., CDR1, CDR2, and CDR3), and Framework Regions (FRs). VH refers to the variable domain of the heavy chain. VL refers to the variable domain of the light chain. According to the methods used in this invention, the amino acid positions assigned to CDRs and FRs may be defined according to Rabat (Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991)) or according to IMGT nomenclature. id="p-493" id="p-493"
[00493] 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. id="p-494" id="p-494"
[00494] 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. id="p-495" id="p-495"
[00495] The 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 or "FW"). The extent of the framework region and CDRs has been precisely defined (see, 1MGT or Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, and Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917; which are incorporated by reference herein in their entireties). Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminns to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. id="p-496" id="p-496"
[00496] The terms 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. Examples of 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 CH 1 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 consists of a VH or VL domain; and (vi) an isolated complementarity determining region (CDR) that retains specific antigen-binding functionality. id="p-497" id="p-497"
[00497] As used herein, 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. For example, the polypeptide comprises a CDR3 (eg, HCDR3). For example the polypeptide comprises CDRs 1 and 2 (eg, HCDRl and 2) or CDRs 1-3 of a variable domain of an antibody (eg. HCDRsl-3). In an example, the antibody binding site is provided by a single variable domain (eg, a VH or VL domain). In another example, the binding site comprises a VH/VL pair or two or more of such pairs. id="p-498" id="p-498"
[00498] As used herein, 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. For example, in a diagnostic test the specific binding of a ligand can distinguish between two variant IL6R proteins as described herein. In some embodiments, 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. In the context of oligonucleotide strands which interact via hybridization, specific binding can be "specific hybridization." id="p-499" id="p-499"
[00499] Additionally, and as described herein, a recombinant human(ized) antibody can be further optimized to decrease potential immunogenicity, while maintaining functional activity, for therapy in humans. In this regard, 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. id="p-500" id="p-500"
[00500] The term '’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. id="p-501" id="p-501"
[00501] As used herein, an ’’affibody" refers to a relatively small synthetic protein molecule that has high binding affinity for a target protein (e.g. for PCSK9, 1L6R, or a variant thereto). 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 weighs about 6 kDa while an antibody commonly weighs about 150 kDa), an affibody molecule works like an antibody since its binding site is approximately equivalent in surface area to the binding site of an antibody. id="p-502" id="p-502"
[00502] As used herein, "VH3-23*04" refers to a human VH domain variant comprising the polypeptide sequence of SEQ ID NO: 38. As opposed to the reference sequence, 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. As used herein, "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 VH323*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. id="p-503" id="p-503"
[00503] As used herein, "determine" or "determining" refers to ascertaining, e.g., by a quantitative or qualitative analysis. As used herein, "has been determined" can refer to ascertaining on the basis of previously obtained information or simultaneously obtained information. id="p-504" id="p-504"
[00504] In some aspects of all embodiments of the invention 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. id="p-505" id="p-505"
[00505] As used 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 embodiments, 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. id="p-506" id="p-506"
[00506] As used herein "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 variants of PCSK9 or IL6R, or PCSK9 or IL6R and an additional target. A multiplex analysis typically includes analysis of 10-50; 10-100: 1 0-1000, 105000, 10-10000 reactions in a multiplex format, such as a multiwall, an array, or a multichannel reaction. id="p-507" id="p-507"
[00507] Often 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. j00508] The term "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 abovementioned samples. The term "test sample" or "biological sample" also includes untreated or pretreated (or pre-processed) biological samples. For the analysis of nucleic acids, the biological sample should typically comprise at least one cell comprising nucleic acids. id="p-509" id="p-509"
[00509] 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). In addition, the test sample can be freshly collected or a previously collected, refrigerated, frozen or otherwise preserved sample. id="p-510" id="p-510"
[00510] In some embodiments, the test sample can be an untreated test sample. As used herein, the phrase "untreated test sample" refers to a test sample that has not had any prior sample pretreatment 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. In some embodiments, 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. After thawing, a frozen sample can be centrifuged before being subjected to methods, assays and systems described herein. In some embodiments, the test sample is a clarified test sample, for example, by centrifugation and collection of a supernatant comprising the clarified test sample. In some embodiments, 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. In some embodiments, 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. One exemplary reagent is a protease inhibitor, which is generally used to protect or maintain the stability of protein during processing. The skilled artisan is well aware of methods and processes appropriate for pre-processing of biological samples required for determination of the level of an expression product as described herein. id="p-511" id="p-511"
[00511] As used herein, "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. As used herein, "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. id="p-512" id="p-512"
[00512] As used herein, the term ’’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. id="p-513" id="p-513"
[00513] The term 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. id="p-514" id="p-514"
[00514] As used herein, 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. id="p-515" id="p-515"
[00515] As used herein "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. Non-limiting examples of next-generation sequencing methods/platforms include Massively Parallel Signature Sequencing (Lynx Therapeutics); 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 Technologies, and Helicos Biosciences. Next-generation sequencing technologies and the constraints and design parameters of associated sequencing primers are well known in the art (see, e.g. Shendure, et al., "Next-generation DNA sequencing/’ Nature, 2008, vol. 26, No. 10, 1135-1145; Mardis, "The impact of next-generation sequencing technology on genetics," Trends in Genetics, 2007, vol. 24, No. 3. pp. 133-141; Su, et al., "Next-generation sequencing and its applications in molecular diagnostics" Expert Rev Mol Diagn, 2011, 11(3):333-43: Zhang et ah, "The impact of next-generation sequencing on genomics", J Genet Genomics, 2011, 38(3):95-109; (Nyren, P. et al. Anal Biochem 208: 17175 (1993); Bentley, D. R. Curr Opin Genet Dev 16:545-52 (2006); Strausberg, R. L., et al. Drug Disc Today 13:569-77 (2008); U.S. Pat. No. 7,282,337; U.S. Pat. No. 7,279,563; U.S. Pat. No. 7,226,720; U.S. Pat. No. 7,220,549; U.S. Pat. No. 7,169,560; U.S. Pat. No. 6,818,395; U.S. Pat. No. 6.911,345; US Pub. Nos. 2006/0252077; 2007/0070349; and 20070070349; which are incorporated by referene herein in their entireties). id="p-516" id="p-516"
[00516] As used herein, ’’nucleic acid hybridization" refers to the pairing of complementary RNA and DNA strands as well as the pairing of complementary DNA single strands. In some embodiments, 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. id="p-517" id="p-517"
[00517] As used herein, 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. That is, "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. The term treatment" of a disease also includes providing relief from the symptoms or side-effects of the disease (including pal 1 iative treatment). For treatment to be effective a complete cure is not contemplated. The method can in certain aspects include cure as well. id="p-518" id="p-518"
[00518] As used herein, 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. The phrase 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. id="p-519" id="p-519"
[00519] As used herein, the term 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. id="p-520" id="p-520"
[00520] Multiple 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. id="p-521" id="p-521"
[00521] As used herein, "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. [00522| As used herein, "obtain" refers to any method of acquiring, securing, procuring, or coming into the possession of, e.g. a sample. 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. id="p-523" id="p-523"
[00523] As used herein, "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. id="p-524" id="p-524"
[00524] As used herein in reference to cholesterol levels, "maintain" refers to preventing the level from worsening (e.g. increasing). In some embodiments, 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. id="p-525" id="p-525"
[00525] In some embodiments, the subject treated according to the methods described herein has previously had their cholesterol level reduced. As used herein, "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. id="p-526" id="p-526"
[00526] An existing treatment for high cholesterol levels is the administration of a statin.
As referred to herein, a "statin" (also known as 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). Non-limiting examples of statins can include atorvastatin (LIPITOR™), fluvastatin (LESCOL™), lovastatin (MEVACOR™, ALTOCOR™), pitavastatin (LIVALO™), pravastatin (PRAVACHOL™), rosuvastatin (CRESTOR™), and simvastatin (ZOCOR™). Statins can be administered in combination with other agents, e.g. the combination of ezetimibe and simvastatin. id="p-527" id="p-527"
[00527] Some subjects are, or become, resistant to statin treatment. As used herein, "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. id="p-528" id="p-528"
[00528] Some subjects are, or become, resistant to anti-TNF alpha treatment. As used herein, "resistant to anti-TNF alpha treatment" or "reduced responsiveness to anti-TNF alpha treatment" or similar refers to a subject exhibiting a statistically significantly lower response to the administration of an anti-TNF alpha treatment 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 [00529] As used herein, the term "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 (e.g. antibodies and nucleic acid probes) are well known in the art. id="p-530" id="p-530"
[00530] In some embodiments, 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 primary labels (where the label comprises a moiety that is directly detectable or that produces a directly detectable moiety) or secondary labels (where the detectable label binds to another moiety to produce a detectable signal, e.g., as is common in immunological labeling using secondary and tertiary antibodies). The detectable label can be linked by covalent or non-covalent means to the reagent. Alternatively, 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. id="p-531" id="p-531"
[00531] In other embodiments, the detectable label can be a fluorescent compound. When the fluorescently label is exposed to light of the proper wavelength, its presence can then be detected due to fluorescence. In some embodiments, a detectable label can be a fluorescent dye molecule, or fluorophore including, but not limited to fluorescein, phycoerythrin, phycocyanin, o-phthaldehyde, fluorescamine, Cy3™, Cy5™, allophycocyanine, Texas Red, peridenin chlorophyll, cyanine, tandem conjugates such as phycoerythrin-Cy5™, green fluorescent protein, rhodamine, fluorescein isothiocyanate (FITC) and Oregon Green™, rhodamine and derivatives (e.g., Texas red and tetrarhodimine isothiocynate (TRITC)), biotin, phycoerythrin, AMCA, CyDyes™, 6carboxyfhiorescein (commonly known by the abbreviations FAM and F), 6<3Ηχ^-2’,4',7',4,7hexachlorofiuorescein (HEX), 6-carboxy-4’,5Ldichloro-2r,7’-dimethoxyfiuorescein (JOE or J), N,N,N’,N’-tetramethyl-6carboxyrhodamine (TAMRA or T), 6-carboxy-X-rhodamine (ROX or R), 5carboxyrhodamine-6G (R6G5 or G5), 6-carboxyrhodamine-6G (R6G6 or G6), and rhodamine 110; cyanine dyes, e.g. Cy3, Cy5 and Cy7 dyes; coumarins, e.g umbelliferone; benzimide dyes, e.g.
Hoechst 33258; phenanthridine dyes, e.g. Texas Red: ethidium dyes: acridine dyes; carbazole dyes; phenoxazine dyes; porphyrin dyes; polymethine dyes, e.g. cyanine dyes such as Cy3, Cy5, etc; BODIPY dyes and quinoline dyes. In some embodiments, a detectable label can be a radiolabel including, but not limited to 3H, 1251,3:)S, l4C, 32P, and 33P. In some embodiments, 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. In some embodiments, 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. In some embodiments, a detectable label can be a spectral colorimetric label including, but not limited to colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, and latex) beads. id="p-532" id="p-532"
[00532] In some embodiments, reagents can also be labeled with 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. In this 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. Such streptavidin peroxidase detection kits are commercially available, e. g. from DAKO; Carpinteria, CA. A reagent can also be detectably labeled using fluorescence emitting metals such as l52Eu, 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). id="p-533" id="p-533"
[00533] As used herein, "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. As used herein "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. id="p-534" id="p-534"
[00534] As used herein, ’’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. In some embodiments, 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 of an injection device is a needle and syringe. [00535] As used herein, 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. id="p-536" id="p-536"
[00536] As used herein, "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. id="p-537" id="p-537"
[00537] As used herein, "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. id="p-538" id="p-538"
[00538] As used herein, 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. id="p-539" id="p-539"
[00539] As used herein, "classification" of a subject, e.g., classification of the subject’s ancestry 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. In some embodiments, 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 12. [00540] The term "statistically significant or "significantly" refers to statistical significance and generally means a two standard deviation (2SD) or greater difference. id="p-541" id="p-541"
[00541] Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about." The term "about" when used in connection with percentages can mean ±1%. id="p-542" id="p-542"
[00542] As used herein the term ’’comprising or comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the method or composition, yet open to the inclusion of unspecified elements, whether essential or not. id="p-543" id="p-543"
[00543] The term consisting of' refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment. id="p-544" id="p-544"
[00544] As used herein 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 character!stic(s) of that embodiment. id="p-545" id="p-545"
[00545] The singular terms "a," an," and the" include plural referents unless context clearly indicates otherwise. Similarly, the word "or" is intended to include and" unless the context clearly indicates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The abbreviation, e.g. is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation e.g. is synonymous with the term for example. id="p-546" id="p-546"
[00546] Definitions of common terms in cell biology and molecular biology can be found in "The Merck Manual of Diagnosis and Therapy", 19th Edition, published by Merck Research Laboratories, 2006 (ISBN 0-911910-19-0); Robert S. Porter et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); Benjamin Lewin, Genes X, published by Jones & Bartlett Publishing, 2009 (ISBN-10: 0763766321); Kendrew et al. (eds.),, Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8) and Current Protocols in Protein Sciences 2009, Wiley Intersciences, Coligan ct al., eds. id="p-547" id="p-547"
[00547] Unless otherwise stated, the present invention was performed using standard procedures, as described, for example in Sambrook et al., Molecular Cloning: A Laboratory Manual (4 ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (2012); Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (1995); or Methods in Enzymology: Guide to Molecular Cloning Techniques Vol. 152, S. L. Berger and A. R. Kimmel Eds., Academic Press Inc., San Diego, USA (1987); Current Protocols in Protein Science (CPPS) (John E. Coligan, et. al., ed., John Wiley and Sons, Inc.), Current Protocols in Cell Biology (CPCB) (Juan S. Bonifacino et. al. ed., John Wiley and Sons, Inc,), and Culture of Animal Cells: A Manual of Basic Technique by R. Ian Freshney, Publisher: Wiley-Liss; 5th edition (2005), Animal Cell Culture Methods (Methods in Cell Biology, Vol. 57, Jennie P. Mather and David Barnes editors, Academic Press, 1st edition, 1998) which are all incorporated by reference herein in their entireties. [00548] Other terms are defined herein within the description of the various aspects of the invention. id="p-549" id="p-549"
[00549] All patents and other publications; including literature references, issued patents, published patent applications, and co-pending patent applications; cited throughout this application are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the technology described herein. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents. id="p-550" id="p-550"
[00550] The description of embodiments of the disclosure is not intended to be exhaustive or to limit the disclosure to the precise form disclosed. While specific embodiments of, and examples for, the disclosure are described herein for illustrative purposes, various equivalent modifications are possible within the scope of the disclosure, as those skilled in the relevant art will recognize. For example, while method steps or functions are presented in a given order, alternative embodiments may perform functions in a different order, or functions may be performed substantially concurrently. The teachings of the disclosure provided herein can be applied to other procedures or methods as appropriate. The various embodiments described herein can be combined to provide further embodiments. Aspects of the disclosure can be modified, if necessary, to employ the compositions, functions and concepts of the above references and application to provide yet further embodiments of the disclosure. Moreover, due to biological functional equivalency considerations, some changes can be made in protein structure without affecting the biological or chemical action in kind or amount. These and other changes can be made to the disclosure in light of the detailed description. All such modifications are intended to be included within the scope of the appended claims. id="p-551" id="p-551"
[00551] Specific elements of any of the foregoing embodiments can be combined or substituted for elements in other embodiments. Furthermore, while advantages associated with certain embodiments of the disclosure have been described in the context of these embodiments, other embodiments may also exhibit such advantages, and not all embodiments need necessarily exhibit such advantages to fall within the scope of the disclosure. id="p-552" id="p-552"
[00552] It will be understood that particular configurations, aspects, examples, clauses and embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine study, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims. All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The use of the word a" or "an" when used in conjunction with the term ’’comprising in the claims and/or the specification may mean one, but it is also consistent with the meaning of one or more, at least one, and one or more than one. The use of the term or in the claims is used to mean and/or unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and and/or. Throughout this application, the term about is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects. id="p-553" id="p-553"
[00553] As used in this specification and claim(s), the words comprising (and any form of comprising, such as comprise and comprises), having (and any form of having, such as have and has), including (and any form of including, such as includes and include) or ’’containing" (and any form of containing, such as ’’contains and ’’contain") are inclusive or openended and do not exclude additional, unrecited elements or method steps id="p-554" id="p-554"
[00554] Any part of this disclosure may be read in combination with any other part of the disclosure, unless otherwise apparent from the context. id="p-555" id="p-555"
[00555] All of the 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. id="p-556" id="p-556"
[00556] The present invention is described in more detail in the following non limiting Examples. [00557] Some embodiments of the technology described herein can be defined according to any of the following numbered paragraphs: 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 a. selecting a human that is 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%; and b. administering to the human an anti-TOI ligand to target the TOI in the human to treat or prevent said disease or condition.
In an alternative where the TOI is human PCSK9, 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, 1474V 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 comprises a C-terminal domain comprising said mutation in SEQ ID NO: 1.
In an alternative where the TOI is human PCSK9, 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, 1474V 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 νλ or Vk disclosed herein and wherein 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-termina! domain comprising said mutation in SEQ ID NO: 1.
In an alternative where the TOI is human PCSK9, 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, Ολ 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 Cterminal domain comprising said mutation in SEQ ID NO: 1. 2. The method of paragraph 1, wherein before step (a) the ligand has been or is determined as being capable of specifically binding to said TOI variant. 3. The method of paragraph 1 or 2, comprising determining that the human is positive for the TOI polymorphic variant, optionally wherein the step of determining comprises determining that the human is positive fora nucleotide variant encoding said TOI variant. 4. The method of paragraph 3, wherein the step of determining comprises assaying a biological sample obtained from said human for a nucleotide polymorphism encoding said TOI polymorphic variant.
. The method of paragraph 3 or 4, wherein the step of determining comprises assaying a biological sample obtained from said human for a protein corresponding to the TOI polymorphic variant. 6. The method of any preceding paragraph, wherein said frequency is less than 15%. 7. The method of any preceding paragraph, wherein said frequency is less than 10%. 8. The method of any preceding paragraph, wherein 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%. 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. selecting a human that is negative for a variant 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 that the human is negative for a TOJ variant encoded by a nucleotide sequence comprising the allele having a cumulative human allele frequency of less than 50% and/or having a total human genotype frequency of less than 50%; and b. administering to the human an anti-TOI ligand to target the TOI variant in the human and treat or prevent said disease or condition, wherein 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%.
. The method of paragraph 9, comprising determining that the human is positive for the TOI polymorphic variant, optionally wherein the 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. 11. The method of paragraph 10, wherein determining comprises assaying for the nucleotide sequence to determine the presence of said allele. 12. The method of paragraph 11, wherein the assaying comprises nucleic acid amplification. 13. The method of paragraph 11 or 12, wherein the assaying comprises hybridization, sequencing, or next generation sequencing. 14. The method of any of paragraphs 11-13, further comprising the step of obtaining a biological sample from the human.
. The method of any one of paragraphs 9-14, wherein the ligand has been or is determined as being capable of specifically binding to the most frequent TOI variant. 16. The method of any one of paragraphs 9-15, wherein the ligand has been or is determined as being substantially incapable of neutralising or inhibiting said TOI variant, 17. The method of any one of paragraphs 9-16, wherein the ligand is capable of specifically binding to the most frequent TOI variant. 18. The method of any one of paragraphs 9-17, wherein 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%. 19. The method of any preceding paragraph, wherein said TOI polymorphic variant has been or is determined as being present in at least two different human ethnic populations.
. The method of any preceding paragraph, wherein 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. 21. The method of any of the preceding paragraphs, wherein the ligand is an antibody, antibody fragment or an affibody. 22. The method of any of the preceding paragraphs, wherein 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. 23. The method of any of the preceding paragraphs, wherein 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. 24. 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.
. A kit for treating or preventing a condition or disease mediated by a target of interest as recited in any preceding paragraph, the kit 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. 26. The composition of paragraph 24 or the kit of paragraph 25, wherein the regulatory agency is FDA or EMA. 27. 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 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. 28. A method of producing an anti-human TOI antibody, the method comprising immunising a nonhuman 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 Jess than 50%, and optionally producing a TOI-binding fragment or derivative of the isolated antibody. 29. The method of paragraph 28, wherein the non-human vertebrate is a mouse or a rat.
. 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. 1. A kit for TOI genotyping a human, wherein 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 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. 32. A kit for TOI genotyping or phenotyping a human, wherein the kit comprises 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. 33. The kit of paragraph 32, wherein the allele is found in at least 2 different ethnic populations. 34. Use of 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 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%.
. Use of 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. 36. A method of targeting a Target of Interest (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 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?4, whereby a TOJ encoded by said nucleotide sequence is targeted. 37. The method of paragraph 36, 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 comprising an allele having a cumulative human allele frequency of less than 50% and/or a total human genotype frequency of less than 50%. 38. A method of Target of Interest (TOI) genotyping a nucleic acid sample of a human, the method 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%. 39. 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%. 40. 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. 41. The method of any one of paragraphs 38-40, comprising using a ligand capable of targeting a nucleic acid 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 a ligand capable of specifically binding the TOI encoded by said nucleic acid sequence to carry out said identifying step. 42. 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. 43. The diagnostic kit wherein the ligand is selected from an antibody, antibody fragment, antibody portion, affybody, oligonucleotide, modified oligonucleotide, antisense oligonucleotide, siRNA, and microRNA. 44. 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. 45, The method, ligand, composition, kit or use of any preceding paragraph, wherein 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%. 46. The method, ligand, composition, kit or use of any preceding paragraph wherein 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. 47. 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 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. 48. The method of paragraph 47, wherein the anti-TOI ligand is selected from an antibody, an antibody portion, an antibody fragment, an affibody, an antisense oligonucleotide, an siRNA, and a microRNA. id="p-558" id="p-558"
[00558] The present invention is described in more detail in the following non limiting Examples. id="p-559" id="p-559"
[00559] The invention addresses the need to treat humans having naturally-occurring rarer natural IL6R SNPs, alleles, genotypes and phenotypes (rarer protein forms). In this respect, the invention provides a method of treating or reducing the risk of an IL6R-mediated disease or condition in a human in need thereof comprising a. selecting a human comprising a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78; and b. administering to said human an antibody or antibody fragment that specifically binds an IL6R amino acid sequence encoded by said nucleotide sequence comprised by the human. id="p-560" id="p-560"
[00560] In an example, step (a) comprises selecting a human comprising an 1L6R protein encoded by the nucleotide sequence. id="p-561" id="p-561"
[00561] In an example, the antibody or antibody fragment specifically binds said 1L6R amino acid sequence. In an example, the antibody or antibody fragment binds a second IL6R protein comprising an amino acid sequence encoded by a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or VaI385Ile in SEQ ID NO: 78. In an example, the antibody comprises a VH domain derived from the recombination of a human VH gene segment, human D gene segments and a human JH segment, the VH gene segment being as defined herein. id="p-562" id="p-562"
[00562] In an example, the human has been determined to comprise the nucleotide sequence. In an example, the human has been determined to comprise an IL6R encoded by the nucleotide sequence. In an example, the method further comprises the step of determining that the human comprises the nucleotide sequence. id="p-563" id="p-563"
[00563] In an example, the determining step is performed before administration of the antibody to the human. In an example, the method further comprises the step of determining that the human comprises an IL6R encoded by the nucleotide sequence. In an example, the determining step is performed before administration of the antibody to the human. In an example, the step of determining comprises assaying a biological sample from the human for said nucleotide sequence. id="p-564" id="p-564"
[00564] Additional Tailoring of Ligands to Genotype and/or Phenotype of the Human Patient id="p-565" id="p-565"
[00565] As described herein, the present invention contemplates ligands (eg, antibodies and fragments) whose binding site specificities have been matched to one or more variant human TOIs, e.g., JL6Rs. The term "TOI" used elsewhere herein encompasses human PCSK9 and/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. For example, where the ligand comprises or consists of a human TOI, e.g, IL6Rbinding antibody or an anti-human TOI, e.g., IL6R receptor (eg, gp!30) 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. Additionally or alternatively, it is contemplated that 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. The inventor realised that it would be desirable to match the ligand to one or more corresponding variant SNPs found in the patient to be treated therapeutically and/or prophylactically. 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. id="p-566" id="p-566"
[00566] 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. For example, it has been observed that 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. For example, studies have suggested that some patients receiving HUMIRA™ (adalimumab), currently the biggest selling antibody medicine, mount a HAHA immune response against the medicine, and this may impact treatment adversely.
Reference is made to JAMA. 2011;305(14): 1460-1468. doi:10.1001/jama.2011.406: "Development of Antidrug Antibodies Against Adalimumab and Association With Disease Activity and Treatment Failure During Long-term Follow-up", GM Bartelds et al. The authors concluded that results of this study showed that development of antidrug antibodies was associated with a negative outcome of adalimumab treatment in human RA patients. It was reported that not only did patients with antiadalimumab antibodies discontinue treatment more often and earlier than patients without antiadalimumab antibodies, they also had a higher disease activity during treatment and only rarely came into remission. In addition, reportedly the data showed that two-thirds of the anti-adalimumab antibody-positive patients developed these antibodies in the first 28 weeks of treatment and that the presence of anti-adalimumab antibodies substantially influenced serum adalimumab concentrations. [00567] This HAHA theme is, therefore, a significant concern and this has been considered by the regulatory authorities. For example, the European Medicines Agency (EMA) has issued a "Guideline on immunogenicity assessment of monoclonal antibodies intended for in vivo clinical use" (available on the World Wide Web at www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2012/06/WC500128688.pdf; EMA/CHMP/BMWP/86289/2010, addendum to EMEA/CHMP/BMWP/14327/2006), which came into force on 1st December 2012. As such, it is good practice for researchers to identify and assess risk of anti-antibody drug occurrence and effects. id="p-568" id="p-568"
[00568] 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, e.g., IL6R medicines for treating, reducing the risk of, and/or preventing human IL6R-related diseases and conditions. id="p-569" id="p-569"
[00569] The inventor also considered the desirability to tailor the variation in the ligand constant region (eg, for an antibody or Fc-eontaining ligand), mindful that then the constant region being administered to the patient would be tuned to the various components, such as patient’s Fe 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 and/or inflammatory indications. In this way it is possible, therefore, to tune the effector function of the constant region (eg, Fc) to the patient more closely, to promote efficacy. For example, more efficacious drugs are desirable for better patient treatment and may provide the possibility of lowered dosing and/or dosing frequencies. id="p-570" id="p-570"
[00570] Thus, in examples of this aspect, the invention provides the following (set out as clauses):- 1. The ligand, method, use, kit or composition of the invention, wherein (i) the ligand (eg, antibody or fragment) comprises (c) a 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 (d) a constant region domain encoded by a C region gene segment; Wherein 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; and (ii) 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.
In an example, the first gene segment is any of the 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 and possible inclusion in one or more claims herein. 2. The ligand, method, use, kit or composition of clause 1, wherein 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. 3. The ligand, method, use, kit or composition of clause 2, wherein SPR is used to carry out said assay.
In an alternative, ELISA is used. 4. The ligand, method, use, kit or composition of any one of clauses 1 to 3, 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 clauses 1 to 4, 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. 6. The ligand, method, use, kit or composition of clause 5, wherein said human expresses an antibody variable domain comprising said first and second amino acid polymorphisms. 7. The ligand, method, use, kit or composition of clause 5 or 6, wherein the first and second SNPs of 20 said genome are comprised by the same antibody gene segment.
For example, 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.
For example, 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. 8. The ligand, method, use, kit or composition of any one of clauses 1 to 7, wherein each SNP is a variable region gene segment SNP. 9. The ligand, method, use, kit or composition of any one of clauses 1 to 7, wherein 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 clause 9, 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. ί. The ligand, method, use, kit or composition of any one of clauses 1 to 8, wherein each SNP is a variable domain SNP, eg, a VH domain SNP, or a Vk domain SNP, or a VZ SNP. 12. The ligand, method, use, kit or composition of any one of clauses 1 to 11, wherein said constant region domain of (b) is comprised by an antibody Fc region. 13. The ligand, method, use, kit or composition of any one of clauses 1 to 12, wherein the ligand (eg, antibody or fragment) has been determined to specifically bind one or more human TOI, e.g., IL6R, variants as disclosed herein, for example, with a KD of InM or less (eg, 100 or lOpM or less) as determined by SPR. 14. 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).
In an example, 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 I to 14), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fcfused human TOI, e.g., IL6R 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. 16. 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, e.g., IL6R, 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. 17. 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 Fcfused human TOI, e.g.. IL6R 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. 18. 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 Fcfused human TOI, e.g., IL6R 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. 19. 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 Fcfused human TOL e.g.. IL6R 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, 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 Fcfused human TOI, e.g., IL6R 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. 21. The ligand, method, use, kit or composition of any one of clauses 16 to 20, wherein said human gamma heavy chain is a human gamma-1 heavy chain. 22. The ligand, method, use, kit or composition of any one of clauses 16 to 20, wherein said human gamma heavy chain is a human gamma-2 heavy chain. 23. The ligand, method, use, kit or composition of any one of clauses 16 to 20, wherein ligand comprises a human IGHGl*01 gamma-1 heavy chain constant region. 24. The ligand, method, use, kit or composition of any one of clauses 16 to 20, wherein ligand comprises a human IGHG2*01 gamma-1 heavy chain constant region.
. The ligand, method, use, kit or composition of any one of clauses 15 to 24, wherein the human has been or is genotyped as positive for said heavy chain constant region nucleotide sequence. 26. The ligand, method, use, kit or composition of clause 23, wherein the human has been or is genotyped as positive for human IGHGl*01 nucleotide sequence. 27. The ligand, method, use, kit or composition of clause 24, wherein the human has been or is genotyped as positive for human IGHG2*01 nucleotide sequence. 28. The ligand, method, use, kit or composition of any one of clauses 16 to 24, wherein the human has been or is phenotyped as positive for said gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc. 29. The ligand, method, use, kit or composition of clause 28, (i) when dependent from clause 23, wherein the human has been or is phenotyped as positive for a human IGHGl*01 gamma heavy chain constant domain, CHI, CH2, CH3. CH4 or Fc or (ii) when dependent from clause 24. wherein the human has been phenotyped as positive for a human IGHG2*01 gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc.
. The method or use of any one of clauses 16 to 24 and 26 to 29, 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 1GHG2*O1 gamma heavy chain constant region, CHI, CH2, CH3, CH4 or Fc nucleotide sequence. 31. The method or use of any one of clauses 16 to 24 and 26 to 30, comprising phenotyping the 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 IGHGl*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. id="p-571" id="p-571"
[00571] Examples of Tailored Ligands The inventor analysed amino acid variability and distribution amongst large representative human samples. The result of the analysis for antibody gene segments is shown in Tables 4 and 9. id="p-572" id="p-572"
[00572] In a first example, the inventor identified the possibility of addressing the rarer IGH-gamma-1 SNPs 204D (observed cumulative frequency of 0.296) and 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. Thus, this example provides aspects set out in the following clauses. 32. 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 Fcfused TOI receptor) comprises a human gamma-1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 4 or a Leu corresponding to position 206 of SEQ ID NO: 4 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 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). In an example, the sample is a blood or saliva or cheek swab sample. 33. The ligand, method, use, kit or composition of clause 32,wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 81 and a Leu corresponding to position 206 of SEQ ID NO: 81. 34. The ligand, method, use, kit or composition of clause 32 or 33,wherein the genome of the human comprises a gamma-1 heavy chain constant region nucleotide sequence that encodes such an Asp and Leu or the human expresses antibodies comprising human gamma-1 constant regions comprising such an Asp and Leu.
. The ligand, method, use, kit or composition of clause 32, 33 or 34, wherein the ligand comprises a human IGHGl*01 gamma-1 heavy chain constant region, eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG 1*01. 36. The ligand, method, use, kit or composition of any one of clauses 32 to 35, wherein the 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. 37. The ligand, method, use, kit or composition of any one of clauses 32 to 36, wherein the ligand comprises a hinge region encoded by human IGHG 1*01. 38. The ligand, method, use, kit or composition of any one of clauses 32 to 37, wherein the ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 104. 39. The ligand, method, use, kit or composition of any one of clauses 32 to 38, wherein the human is of European ancestry.
As shown in Table 14, 204D and 206L are found in such humans. 40. The ligand, method, use, kit or composition of any one of clauses 32 to 39, wherein the human has been or is genotyped as positive for said Asp and/or Leu. 41. The ligand, method, use, kit or composition of any one of clauses 32 to 40, wherein the human has been or is genotyped as positive for human IGHG 1*01. 42. The ligand, method, use, kit or composition of any one of clauses 32 to 41, wherein the human has been or is phenotyped as positive for a human IGHG I *01 CH3. 43. The method or use of any one of clauses 32 to 42, comprising selecting a said human whose genome comprises a codon(s) encoding said Asp and/or Leu; comprises human IGHG 1 *01; or comprises a human IGHG1*O1 CH3. 44. The method or use of any one of clauses 32 to 43, comprising selecting a said human whose phenotype comprises said Asp and/or Leu; a human IGHG 1 *01 region ; or a human IGHG 1*01 CH3. 44a. The ligand, method, use, kit or composition of any one of clauses 32 to 44, wherein the human expresses antibodies comprising human gamma-1 constant regions comprising such an Asp and Leu. id="p-573" id="p-573"
[00573] In a second example, the inventor identified the possibility of addressing IGHgamma-2 SNPs. This included consideration of Fc region variation - in this respect, the inventor 5 focused on positions 161 and 257 which are in the Fc region. Thus, this example provides aspects set out in the following clauses. 45. The ligand, method, use, kit or composition of the invention (eg, according to any one of clauses to 31), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fc10 fused TOT 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: 83, an Asn corresponding to position 75 of SEQ ID NO: 83, a Phe corresponding to position 76 of SEQ ID NO: 83, a Val corresponding to position 161 of SEQ ID NO: 83 and an Ala corresponding to position 257 of SEQ ID NO: 83; 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. 46. The ligand, method, use, kit or composition of clause 45, wherein the ligand comprises a human 20 gamma-2 heavy chain constant region that comprises (i) a Pro corresponding to position 72 of SEQ ID NO: 83, an Asn corresponding to position 75 of SEQ ID NO: 83, a Phe corresponding to position 76 of SEQ ID NO: 83 and optionally (ii) a Val corresponding to position 161 of SEQ ID NO: 83 and/or an Ala corresponding to position 257 of SEQ ID NO: 83; and wherein the genome of the human comprises a gamma-2 heavy chain constant region nucleotide sequence that encodes 25 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 CH 1 variation. 47. The ligand, method, use, kit or composition of clause 45 or 46, wherein the ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Val corresponding to position 161 of SEQ ID NO: 83 and an Ala corresponding to position 257 of SEQ ID NO: 83 and optionally (ii) an amino acid selected from the group consisting of a Pro corresponding to position 72 of SEQ ID NO: 83, an Asn corresponding to position 75 of SEQ ID NO: 83 and a Phe corresponding to position 76 of SEQ ID NO: 83; 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. 48. The ligand, method, use, kit or composition of any one of clauses 45 to 47, wherein the 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. 49. The ligand, method, use. kit or composition of any one of clauses 45 to 48, wherein the 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. 50. The ligand, method, use, kit or composition of any one of clauses 45 to 49, wherein the ligand comprises a hinge region encoded by human IGHG2*0E 51. The ligand, method, use, kit or composition of any one of clauses 45 to 50, wherein the ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 104. 52. The ligand, method, use, kit or composition of any one of clauses 45 to 51, wherein the human is of European, African American, or European American ancestry. 53. The ligand, method, use, kit or composition of any one of clauses 45 to 52, wherein the human has been or is genotyped as positive for one, more or all of said Pro, Asn, Phe, Val and Ala. 54. The ligand, method, use, kit or composition of any one of clauses 45 to 53, wherein the human has been or is genotyped as positive for human IGHG2*01. 55. The ligand, method, use, kit or composition of any one of clauses 45 to 54, wherein the human has been or is phenotyped as positive for a human 1GHG2*O 1 CHI. 56. The ligand, method, use, kit or composition of any one of clauses 45 to 55, wherein the human has been or is phenotyped as positive for a human IGHG2*01 CH2, 57. The ligand, method, use, kit or composition of any one of clauses 45 to 56, wherein the human has been or is phenotyped as positive for a human IGHG2*01 CH3. 58. The method or use of any one of clauses 45 to 57, comprising selecting a said human whose genome comprises a codon(s) encoding one, more or all of said Pro, Asn, Phe, Val and Ala; comprises human IGHG2*0l; or comprises a human IGHG2*01 CHI, CH2 and/or CH3. 59. The method or use of 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. 60. The ligand, method, use, kit or composition of any one of clauses 45 to 59, wherein the human expresses antibodies comprising human gamma-2 constant regions comprising such a Pro, Asn, Phe, Val and Ala. [00574J In a third example, the inventor addressed human kappa constant region variation.
Thus, the present aspect of the invention also provides the following. 61. The ligand, method, use, kit or composition of the invention (eg, according to any one of clauses to 60), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fcfused TOI receptor) comprises a human kappa light chain constant region that comprises a Val corresponding to position 84 of SEQ ID NO: 93 or a Cys corresponding to position 87 of SEQ ID NO: 93; 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. 62. The ligand, method, use, kit or composition of clause 61, wherein the ligand comprises a human kappa light chain constant region that, comprises a Val corresponding to position 84 of SEQ ID NO: 93 and a Cys corresponding to position 87 of SEQ ID NO: 93. 63. The ligand, method, use, kit or composition of clause 61 or 62, wherein the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val and Cys or the human expresses antibodies comprising human kappa constant regions comprising such a Val and Cys. 64. The ligand, method, use, kit or composition of any one of clauses 61 to 63, wherein the antibody or fragment comprises a human 1GKC*O1 kappa light chain constant region. 65. The ligand, method, use, kit or composition of any one of clauses 61 to 64, wherein the ligand comprises or consists of an antibody, wherein the antibody comprises light chains that comprise SEQ ID NO: 105. 66. The ligand, method, use, kit or composition of any one of clauses 61 to 65, wherein the 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: 100). 67. The ligand, method, use, kit or composition of any one of clauses 61 to 66, wherein the human has been or is phenotyped as positive for said Val and/or Cys. 68. The ligand, method, use, kit or composition of any one of clauses 61 to 67, wherein the human has been or is genotyped as positive for human IGKC*0E 69. The ligand, method, use, kit or composition of any one of clauses 61 to 68, wherein the human has been or is phenotyped as positive for a human 1GKC*O1 domain. 70. The method or use of any one of clauses 61 to 69, comprising selecting a said human whose genome comprises a codon(s) encoding said Val and/or Cys; or comprises human IGKC*0l. 71. The method or use of any one of clauses 61 to 70, comprising selecting a said human whose phenotype comprises such a Val and/or Cys; or comprises a human IGKC*01 domain. 72. The ligand, method, use, kit or composition of any one of clauses 61 to 7], wherein the human expresses antibodies comprising human kappa constant domains comprising such a Val and Cys, eg, expresses human IGKC*01 constant domains. id="p-575" id="p-575"
[00575] In a fourth example, the inventor addressed human lambda constant region variation. Thus, this example provides aspects set out in the following clauses. 73. The ligand, method, use, kit or composition of the invention (eg, according to any one of clauses to 60), wherein the ligand (eg, comprising or consisting of an antibody or fragment or an Fcfused TOI receptor) comprises a human IGLC2*01 light chain constant region; and wherein the genome of the human comprises a human 1GLC2*O1 nucleotide sequence or the human expresses antibodies comprising human light chain IGLC2*0l constant regions. 100 74. The ligand, method, use, kit or composition of clause 73, wherein the antibody comprises light chains that comprise SEQ ID NO: 105. 75. The ligand, method, use, kit or composition of clause 73 or 74, wherein the human has been or is genotyped as positive for human IGLC2*01. 76. The ligand, method, use, kit or composition of any one of clauses 73 to 75, wherein the human has been or is phenotyped as positive for a human IGLC2*01 domain. 77. The method or use of any one of clauses 73 to 76, comprising selecting a said human whose genome comprises human 1GLC2*O1. 78. The method or use of any one of clauses 73 to 77, comprising selecting a said human whose phenotype comprises a human IGLC2*01 domain. 79. The ligand, method, use, kit or composition of any one of clauses 73 to 78, wherein the human expresses antibodies comprising human lambda IGLC2*0l constant domains. id="p-576" id="p-576"
[00576] In a fifth example, the inventor addressed human heavy chain variable region variation. Thus, this example provides aspects set out in the following clauses. 80. 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) IGHV1-18*01 and the genome ofthe human comprises a human IGHVl-18*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV 1-18*01; or (ii) 1GVH1-46*O1 and the genome of the human comprises a human IGHV1-46*O1 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV 1-46*01.
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) IGHV3-9*01 and the genome of the human comprises a human IGHV3-9*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human 1GHV3-9*O1; or (ii) 101 IGHV3-7*01and the genome of the human comprises a human IGHV3-7*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV3-7*01. 81. The ligand, method, use, kit or composition of clause 80, wherein the antibody or fragment comprises a one more or all of a CHI domain, CH2 domain, CH3 domain, hinge or Fc encoded by human IGHG2*01. 82. The ligand, method, use, kit or composition of clause 80 or 81, wherein the antibody or fragment comprises heavy chains that comprise SEQ ID NO: 104 or 106. 83. The ligand, method, use, kit or composition of any one of clauses 80 to 82, wherein the human has been or is genotyped as positive for said selected VH gene segment, positive for human IGHVl-18*01 or IGVH 1-46*01.
The ligand, method, use, kit or composition of any one of clauses 80 to 82, wherein the human has been or is genotyped as positive for said selected VH gene segment, positive for human IGHV3-9*01 or IGHV3-7*01. 84. The method or use of any of clauses 80 to 83, comprising genotyping the human as positive for said selected VH gene segment, eg, positive for human IGHV1-18*01 or IGVH I-46*01.
The method or use of any of clauses 80 to 83, comprising genotyping the human as positive for said selected VH gene segment, eg, positive for human IGHV3-9*01 or IGHV3-7*01. [00577J In a sixth example, the inventor addressed human light chain variable region variation. Thus, this example provides aspects set out in the following clauses. 85. The ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 84), 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-l*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-l*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*0t; or (iii) 1GKV1-13*O2 and the genome of the human comprises a human IGKV1- 102 13*02 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV1-13*O2.
The ligand, method, use, kit or composition of the invention (eg, according to any one of clauses 1 to 84), 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) 1GKV1-12*0 land the genome of the human comprises a human IGKV 1-12*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human JGKV1-12*01; or (ii) IGKV3-11 *01 and the genome of the human comprises a human IGKV3-11 *01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV3-ll*01. 86. The ligand, method, use, kit or composition of clause 85, wherein the antibody comprises light chains that comprise SEQ ID NO: 105 or 107. 87. The ligand, method, use, kit or composition of clause 85 or 86, wherein 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 1GKJ2*O1 (SEQ ID NO: 100) or IGKJ4*01 (SEQ ID NO: 102). 88. The ligand, method, use, kit or composition of any one of clauses 85 to 87, wherein the human has been or is genotyped as positive for said selected VL gene segment, eg, positive for human 1GKV4-1 *01,1GLV2-14*O1 or IGKV1-13*O2. 89. The method or use of 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*0) or IGKVl-13*02.
The method or use of clause 88, comprising genotyping the human as positive for said selected VL gene segment, eg, genotyping the human as positive for human IGKV1-12*01 or IGKV311*01. 90. The ligand, method, use, kit or composition of any one of clauses 1 to 89, wherein the ligand (eg, antibody or fragment) binds said TOI, e.g., human IL6R that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 with a dissociation constant (Kd) of InM or less as determined by SPR, (eg, 100, 10 or IpM or less). 103 id="p-578" id="p-578"
[00578] In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an IL6R-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 an IL6R protein that comprises a mutation Asp358Ala orVal385Ile in SEQ ID NO: 78; wherein the ligand 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 (i) a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109 and wherein said human comprises a VH gene segment encoding a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109, or the human expresses VH domains that comprise a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109; or (ii) a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111 and wherein said human comprises a VH gene segment encoding a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111 or the human expresses VH domains that comprise a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said IL6R protein comprises a mutation Asp358Ala in SEQ ID NO: 78.
In an example, said IL6R protein comprises a mutation Val385Ile in SEQ ID NO: 78. 2. A method of treating or reducing the risk of an JL6R-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 an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; wherein the ligand comprises a VH domain derived from the recombination of a human VH gene segment, a human D gene segment and a human JH segment, the human VH gene segment comprising (a) T at nucleotide number 86 shown in SEQ ID NO: 84 and wherein said human 104 comprises a VH gene segment comprising T at nucleotide number 86 shown in SEQ ID NO: 84; or (b) C at nucleotide number 272 shown in SEQ ID NO: 84 and wherein said human comprises a VH gene segment comprising T at nucleotide number 272 shown in SEQ ID NO: 84; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78. 3. The method of any preceding clause, wherein each said human VH gene segment comprises SEQ ID NO: 108. 4. The method of any preceding clause, wherein each said human VH gene segment comprises SEQ ID NO: 110.
. The method of any preceding clause, wherein said VH domain is derived from the recombination of human VH segment IGHV3-9*01, a human D gene segment and a human JH segment. 6. The method of any preceding clause, wherein said VH domain comprises the FW3 sequence of SEQ ID NO: 111. 7. The method of any preceding clause, wherein the VH gene segment comprised by said human is a germline VH gene segment. 8. The method of any preceding clause, wherein the ligand comprises a human gamma-1 heavy chain comprising said VH domain and a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises an 1GHGP01 human heavy chain constant region gene segment. 9. The method of any preceding clause, wherein the antibody or fragment comprises an IGHG1 *01 human heavy chain constant region.
. The method of any preceding clause, wherein the human is of AMR, ASN or EUR ancestry.
For example, the human is of CHB, CHS, JPT, CEU or FIN ancestry. These ancestries have very high (greater than 85%, for example greater than 90% or even 100%) cumulative frequencies for the SNP (FW3 SNP) and thus the ligand of the invention is well suited to treating or preventing a 105 human IL6R-mediated disease or condition in such a human. 11. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1. 12. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an 1L6R protein comprising said mutation Asp358Ala and/or Val385Jle in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 13. The method of any preceding clause, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val38511e and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385He, optionally, wherein the determining step is performed before administration of the antibody to the human. 14. The method of clause 13, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78.
. The method of clause 14, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 16. The method of clause 14, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a 106 multiplex format. 17. The method of clause 14 or 16, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 8. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385He in SEQ ID NO: 78. 19. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate.
. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. 21. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 22. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 23. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (1BD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic 107 juvenile idiopathic arthritis. 24. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (1BD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 26. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 27. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-579" id="p-579"
[00579] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An antiHL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein the ligand 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 (i) a CDR] comprising a Phe at position 4 shown in SEQ ID NO: 109 and wherein said human comprises a VH gene segment encoding a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109, or the human expresses VH domains that comprise a CDRI comprising a Phe at 108 position 4 shown in SEQ ID NO: 109; or (ii) a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111 and wherein said human comprises a VH gene segment encoding a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111 or the human expresses VH domains that comprise a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111; and wherein said human comprises a nucleotide sequence encoding said 1L6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. An anti-IL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein the ligand comprises a VH domain derived from the recombination of a human VH gene segment, a human D gene segment and a human JH segment, the human VH gene segment comprising (a) T at nucleotide number 86 shown in SEQ ID NO: 84 and wherein said human comprises a VH gene segment comprising T at nucleotide number 86 shown in SEQ ID NO: 84; or (b) C at nucleotide number 272 shown in SEQ ID NO: 84 and wherein said human comprises a VH gene segment comprising T at nucleotide number 272 shown in SEQ ID NO: 84; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78 3. The ligand of any preceding paragraph, wherein each said human VH gene segment comprises SEQ ID NO: 108. 4. The ligand of any preceding paragraph, wherein each said human VH gene segment comprises SEQ ID NO: 110.
. The ligand of any preceding paragraph, wherein said VH domain is derived from the recombination of human VH segment IGHV3-9*01, a human D gene segment and a human JH segment. 109 6. The ligand of any preceding paragraph, wherein said VH domain comprises the FW3 sequence of SEQ ID NO: 111. 7. The ligand of any preceding paragraph, wherein the VH gene segment comprised by said human is a germline VH gene segment. 8. The ligand of any preceding paragraph, wherein the ligand comprises a human gamma-1 heavy chain comprising said VH domain and a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises an IGHGP01 human heavy chain constant region gene segment. 9. The ligand of any preceding paragraph, wherein the antibody or fragment comprises an IGHGl*01 human heavy chain constant region.
. The ligand of any preceding paragraph, wherein the human is of AMR, ASN or EUR ancestry.
For example, the human is of CHB, CHS, JPT, CEU or FIN ancestry. 11. The ligand of any preceding paragraph, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 12. The ligand of any preceding paragraph, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385He; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385 He, optionally, wherein the determining step is performed before administration ofthe antibody to the human. 13. The ligand of paragraph 12, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 14. The ligand of paragraph 13, wherein the assaying comprises 110 contacting the biological sample with at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding an IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78.
. The ligand of paragraph 13, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 16. The ligand of paragraph 13 or 15, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 17. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 18. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 19. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an anti- 111 TNF alpha treatment.
. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 21. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory disease or condition. 22. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 23. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvFlD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 24. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versusdiost disease (GvHD). asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 112 26. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. [00580J In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an IL6R-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 an IL6R protein that comprises a mutation Asp358Ala orVa!385Ile in SEQ ID NO: 78; wherein the ligand comprises a VH domain derived from the recombination of human VH segment IGHV3-7*0I, 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; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. The method of clause 1, wherein the ligand comprises a human gamma-1 heavy chain comprising said VH domain and a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises an 1GHG1*O1 human heavy chain constant region gene segment. 3. The method of any preceding clause, wherein the antibody or fragment comprises an IGHG1 *01 human heavy chain constant region. 4. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1. 113 . The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 and/or an 1L6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 6. The method of any preceding clause, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385He and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The method of clause 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 8. The method of clause 7, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or VaI38511e in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the 1L6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The method of clause 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 114 . The method of clause 7 or 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 12. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is optionally selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 13. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. 14. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment.
. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 16. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 115 In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 17. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (1BD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736.
. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-581" id="p-581"
[00581] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-!L6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an 1L6R protein that comprises a mutation Asp358AIa or Val385Ile in SEQ ID NO: 78, wherein 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 116 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 nucleotide sequence encoding said EL6R protein comprising said 5 Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. The ligand of any paragraph 1, wherein the ligand comprises a human gamma-1 heavy chain comprising said VH domain and a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises an IGHGl*01 human heavy chain constant region gene segment. 3. The ligand of any preceding paragraph, wherein the antibody or fragment comprises an IGHGl*01 human heavy chain constant region. 4. The ligand of any preceding paragraph, wherein the human has been determined to comprise the 20 nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78.
. The ligand of any preceding paragraph, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile; and/or an 1L6R protein comprising said mutation Asp358Ala and/or Val385 He, optionally, wherein the determining step is performed before administration of the antibody to the human. 6. The ligand of paragraph 5, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 117 7. The ligand of paragraph 6, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385IJe in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ JD NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 8. The ligand of paragraph 6, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 9. The ligand of paragraph 6 or 8, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human.
. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385He in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 11. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), 118 golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 12. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment. 13. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 14. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory· disease or condition.
. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 16. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 17. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, 119 adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 19. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. |00582] In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an [L6R-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 an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; wherein the ligand comprises a Vk domain derived from the recombination of human Vk segment IGKV1-12*O1 and a human Jk segment, and wherein said human comprises a IGKV1-12*O1 Vk gene segment or the human expresses Vk domains derived from the recombination of human Vk segment IGKV1-12*01 and a human Jk segment; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2, The method of clause 1, wherein the ligand comprises a human kappa chain comprising said Vk domain and a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys shown in SEQ ID NO: 93 and wherein said human comprises an IGKC*01 human kappa chain constant region gene segment. 120 3. The method of any preceding clause, wherein the antibody or fragment comprises an IGKC*01 human kappa chain constant region. 4. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1.
. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 6. The method of any preceding clause, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or Val38511e, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The method of clause 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78. 8. The method of clause 7, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The method of clause 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid 121 hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
. The method of clause 7 or 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ilein SEQ ID NO: 78. 12. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 13. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. 14. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment.
. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 16. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic 122 juvenile idiopathic arthritis. 17. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation. Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736.
. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-583" id="p-583"
[00583] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-IL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or VaI385Ile in SEQ ID NO: 78, wherein the ligand comprises a Vk domain derived from the recombination of human Vk segment IGKV 1-12*01 and a human Jk segment, and wherein said human comprises a IGKV1-12*01 Vk gene segment or the human expresses Vk domains derived from the recombination of human Vk segment IGKV1-12*01 and a human Jk segment; and 123 wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Alaor Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an 1L6R protein that comprises a mutation Asp358Ala or Val385lie in SEQ ID NO: 78 2. The ligand of any paragraph 1, wherein the ligand comprises a human kappa chain comprising said Vk domain and a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys shown in SEQ ID NO: 93 and wherein said human comprises an IGKC*01 human kappa chain constant region gene segment. 3. The ligand of any preceding paragraph, wherein the antibody or fragment comprises an IGKC*O1 human kappa chain constant region. 4. The ligand of any preceding paragraph, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78.
. The ligand of any preceding paragraph, comprising the step of determining that the human comprises the nucleotide sequence that encodes an (L6R protein comprising said mutation Asp358Ala and/or Val385Ile; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 6. The ligand of paragraph 5, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 7. The ligand of paragraph 6, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val3851le in SEQ ID 124 NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78. 8. The ligand of paragraph 6, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 9. The ligand of paragraph 6 or 8, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human.
. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the 1L6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 11. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg. Enbrel®) treatment with or without methotrexate. 12. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment. 13. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 125 14. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory disease or condition.
. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 16. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD),. Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 17. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 19. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 126 id="p-584" id="p-584"
[00584] In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an IL6R~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 an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; wherein 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: 113 and wherein said human comprises a Vk gene segment encoding a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113, or the human expresses Vk domains that comprise a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113; or (ii) a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 and wherein said human comprises a Vk gene segment encoding a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 or the human expresses Vk domains that comprise a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385He in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. A method of treating or reducing the risk of an IL6R-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 an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; wherein the ligand comprises a Vk domain derived from the recombination of a human Vk gene segment and a human Jk segment, the human Vk gene segment comprising (a) T at nucleotide number 199 shown in SEQ ID NO: 98 and wherein said human comprises a Vk gene segment comprising T at nucleotide number 199 shown in SEQ ID NO: 98; or (b) C at nucleotide number 284 shown in SEQ ID NO: 98 and wherein said human comprises a Vk gene segment comprising T at nucleotide number 284 shown in SEQ ID NO: 98; and 127 wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78. 3. The method of any preceding clause, wherein each said human Vk gene segment comprises SEQ ID NO: 112. 4. The method of any preceding clause, wherein each said human Vk gene segment comprises SEQ ID NO: 114.
. The method of any preceding clause, wherein said Vk domain is derived from the recombination of human Vk segment 1GKV3-11*01 and a human Jk segment. 6. The method of any preceding clause, wherein said Vk domain comprises the FW3 sequence of SEQ ID NO: 115. 7. The method of any preceding clause, wherein the Vk gene segment comprised by said human is a germline Vk gene segment. 8. The method of any preceding clause, wherein the ligand comprises a human kappa chain comprising said Vk domain and a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys shown in SEQ ID NO: 93 and wherein said human comprises an IGKC*01 human kappa chain constant region gene segment.. 9. The method of any preceding clause, wherein the antibody or fragment comprises an IGKC*0l human heavy chain constant region.
. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1. 11. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or VaJ385Ile in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. . 12. The method of any preceding clause, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or 128 Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 13. The method of clause 12, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 14. The method of clause 13, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78.
. The method of clause 13, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 16. The method of clause 13 or 15. wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 17. The method of any/ preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the TL6R protein comprising said mutation Asp358Ala or Val385IIe in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 18. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment. 129 In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 19. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment.
. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 21. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 22. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 23. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 24. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative 130 colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 5 and/or SNP rs28730736. 26. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-585" id="p-585"
[00585] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-lL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein 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: 113 and wherein said human comprises a Vk gene segment encoding a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113, or the human expresses Vk domains that comprise a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: H3; or (ii) a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 and wherein said human comprises a Vk gene segment encoding a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 or the human expresses Vk domains that comprise a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115; and wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val3851le in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. An anti-IL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78, wherein the ligand comprises a Vk domain derived from the recombination of a human Vk gene segment and a human Jk segment, the human Vk gene segment comprising (a) T at nucleotide 131 number 199 shown in SEQ ID NO: 98 and wherein said human comprises a Vk gene segment comprising T at nucleotide number 199 shown in SEQ ID NO: 98; or (b) C at nucleotide number 284 shown in SEQ ID NO: 98 and wherein said human comprises a Vk gene segment comprising T at nucleotide number 284 shown in SEQ ID NO: 98; and wherein said human comprises a nucleotide sequence encoding said 1L6R protein comprising said Asp358Ala or VaI385IIe in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 3. The ligand of paragraph 1 or 2, wherein each said human VH gene segment comprises single nucleotide polymorphism rsl 82958807 and/or rsl91612627. 4. The ligand of any preceding paragraph, wherein each said human Vk gene segment comprises SEQ ID NO: 112.
. The ligand of any preceding paragraph, wherein each said human Vk gene segment comprises SEQ ID NO: 114. 6. The ligand of any preceding paragraph, wherein said Vk domain is derived from the recombination of human Vk segment IGKV3-11 *01 and a human Jk segment. 7. The ligand of any preceding paragraph, wherein said Vk domain comprises the FW3 sequence of SEQ ID NO: 115. 8. The ligand of any preceding paragraph, wherein the Vk gene segment comprised by said human is a germline Vk gene segment. 9. The ligand of any preceding paragraph, wherein the ligand comprises a human kappa chain comprising said Vk domain and a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys shown in SEQ ID NO: 93 and wherein said human comprises an IGKC*01 human kappa chain constant region gene segment..
. The ligand of any preceding paragraph, wherein the antibody or fragment comprises an IGKC*01 human heavy chain constant region. 132 11. The ligand of any preceding paragraph, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or VaI38511e in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 12. The ligand of any preceding paragraph, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 13. The ligand of paragraph 12, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or VaJ385Ile in SEQ ID NO: 78. 14. The ligand of paragraph 14, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ue in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358AIa and/or Val385Ile in SEQ ID NO: 78.
. The ligand of paragraph 13, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 133 16. The ligand of paragraph 13 or 14, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 17. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 18. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cinizia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 19. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment.
. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 21. The ligand of any preceding paragraph, w herein said disease or condition is an inflammatory disease or condition. 22. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 134 23. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 24. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 26. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-586" id="p-586"
[00586] In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an IL6R-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 an IL6R protein that comprises a mutation A$p358Ala or Val385Ile in SEQ ID NO: 78, wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHG1*O1 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: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand, antibody or antibody fragment has been determined to specifically 135 bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 2. The method of clause 1, wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81. 3. The method of any preceding clause, wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHG 1 *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said IL6R protein comprising said mutation Asp358Ala or Val3851Ie in SEQ ID NO: 78. 4. The method of clause 1,2 or 3, wherein the antibody or fragment comprises an IGHG I *01 human heavy chain constant region.
. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause I. 6. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358A!a and/or Val385Ile in SEQ ID NO: 78 and/or an 1L6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78. 7. fhe method of any one of clauses 1 to 6, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385He, optionally, wherein the determining step is performed before administration of the antibody to the human. 8. The method of clause 7, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The method of clause 8, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense 136 sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385IIe in SEQ ID NO: 78.
. The method of clause 8, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 11. The method of clause 8 or 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 12. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358AIa or Val385He in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 13. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
Jn any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 14. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment.
. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 137 16. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 17. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 18. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 21. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 138 id="p-587" id="p-587"
[00587] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-lL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHGl*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: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. The ligand of paragraph 1, wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 8L 3. The ligand of any preceding paragraph, wherein said ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHGl*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 4. The ligand of paragraph 1,2 or 3, wherein the ligand comprises an IGHG1 *01 human heavy chain constant region.
. The ligand of any preceding paragraph, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 6. The ligand of any one of paragraphs 1 to 5, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Vai385Ile; and/or an IL6R protein comprising said mutation Asp358Ala and/or 139 Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The ligand of paragraph 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 8. The ligand of paragraph 7, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The ligand of paragraph 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
. The ligand of paragraph 7 or 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 140 12. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate, 13. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment. 14. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment.
. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory disease or condition. 16. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (FBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 17. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel 141 disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736.
. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-588" id="p-588"
[00588] In an embodiment of the invention, there are provided methods as per the following clanses:- 1. A method of treating or reducing the risk of an IL6R-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 an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein 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: 83, an Asn at position 75 shown in SEQ ID NO: 83, a Phe at position 76 shown in SEQ ID NO: 83, a Val at position 161 shown in SEQ ID NO: 83 and an Ala at position 257 shown in SEQ ID NO: 83 and wherein said human comprises (i) an 1GHG2*OI 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: 83, Asn at position 75 shown in SEQ ID NO: 83, Phe at position 76 shown in SEQ ID NO: 83, Val at position 161 shown in SEQ ID NO: 83 or Ala at position 257 shown in SEQ ID NO: 83 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385lie in SEQ ID NO: 78. 2. The method of clause 1, wherein the ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Pro at position 72 shown in SEQ ID NO: 83, an Asn at position 75 shown in SEQ ID NO: 83, a Phe at position 76 shown in SEQ ID NO: 83 and optionally (ii) a Val at position 161 shown in SEQ ID NO: 83 and/or an Ala at position 257 shown in SEQ ID NO: 83; 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). 142 In an example, said ligand, antibody or antibody fragment has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78. 3. The method of any preceding clause, wherein the ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Val at position 161 shown in SEQ ID NO: 83 and an Ala at position 257 shown in SEQ ID NO: 83 and optionally (ii) an amino acid selected from the group consisting of a Pro at position 72 shown in SEQ ID NO: 83, an Asn at position 75 shown in SEQ ID NO: 83 and a Phe at position 76 shown in SEQ ID NO: 83; 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). 4. The method of clause 1,2 or 3, wherein the antibody or fragment comprises an IGHG2*01 human heavy chain constant region.
. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1. 6. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78. 7. The method of any one of clauses 1 to 6, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 8. The method of clause 7, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The method of clause 8, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ue in SEQ ID NO: 78 or comprising an antisense 143 sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78.
. The method of clause 8, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 11. The method of clause 8 or 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 12. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385lle in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 13. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 14. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment.
. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 144 16. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 17. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 18. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 21. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 145 id="p-589" id="p-589"
[00589] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-IL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358AIa or Val385Ile in SEQ ID NO: 78, wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHGl*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: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. The ligand of paragraph 1, wherein the ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Pro at position 72 shown in SEQ ID NO: 83, an Asn at position 75 shown in SEQ ID NO: 83, a Phe at position 76 shown in SEQ ID NO: 83 and optionally (ii) a Val at position 161 shown in SEQ ID NO: 83 and/or an Ala at position 257 shown in SEQ ID NO:83; 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). 3. The ligand of any preceding paragraph, wherein the ligand comprises a human gamma-2 heavy chain constant region that comprises (i) a Val at position 161 shown in SEQ ID NO: 83 and an Ala at position 257 shown in SEQ ID NO: 83 and optionally (ii) an amino acid selected from the group consisting of a Pro at position 72 shown in SEQ ID NO: 83, an Asn at position 75 shown in SEQ ID NO: 83 and a Phe at position 76 shown in SEQ ID NO: 83; 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). 4. The ligand of paragraph 1, 2 or 3, wherein the ligand comprises an IGHG2*01 human heavy chain constant region. 146 . The ligand of any preceding paragraph, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ue in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 6. The ligand of any one of paragraphs 1 to 5, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385He; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The ligand of paragraph 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 8. The ligand of paragraph 7, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The ligand of paragraph 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 147 . The ligand of paragraph 7 or 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 12. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 13. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment. 14. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment.
. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory disease or condition. 16. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 148 17. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736.
. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-590" id="p-590"
[00590] In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an IL6R-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 an 1L6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and wherein said human comprises (i) an IGKCl*01 human heavy 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: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ue in SEQ ID NO: 78.
In an example, said ligand, antibody or antibody fragment has been determined to specifically 149 bind an 1L6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 2. The method of clause 1, wherein the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys at position 87 shown in SEQ ID NO: 93. 3. The method of any preceding clause, wherein the ligand comprises a human kappa constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys at position 87 shown in SEQ ID NO: 93 and wherein said human comprises (i) an IGKC*01 human kappa chain constant region gene segment and (ii) a nucleotide sequence encoding said IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 4. The method of clause 1,2 or 3, wherein the ligand comprises an IGKC1 *01 human kappa chain constant region.
. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1. 6. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 7. The method of any one of clauses I to 6, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385He and/or an 1L6R protein comprising said mutation Asp358Ala and/or Val385lie, optionally, wherein the determining step is performed before administration of the antibody to the human. 8. The method of clause 7, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The method of clause 8, wherein 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 an 1L6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78 or comprising an antisense 150 sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78.
. The method of clause 8, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 11. The method of clause 8 or 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 12. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 13. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 14. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment.
. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 151 16. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 17. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 18. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 21. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 152 id="p-591" id="p-591"
[00591] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-IL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and wherein said human comprises (i) an IGKC1 *01 human heavy 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: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val38511e in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 2. The ligand of paragraph 1, wherein the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys at position 87 shown in SEQ ID NO: 93. 3. The ligand of any preceding paragraph, wherein the ligand comprises a human kappa constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 and a Cys at position 87 shown in SEQ ID NO: 93 and wherein said human comprises (i) an IGKC*0l human kappa chain constant region gene segment and (ii) a nucleotide sequence encoding said 1L6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 4. The ligand of paragraph 1,2 or 3, wherein the ligand comprises an JGKC1 *01 human kappa chain constant region.
. The ligand of any preceding paragraph, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val3851le in SEQ ID NO: 78. 6. The ligand of any one of paragraphs I to 5, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation 153 Asp358Ala and/or Val385Ile; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The ligand of paragraph 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a 1L6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 8. The ligand of paragraph 7, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The ligand of paragraph 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
. The ligand of paragraph 7 or 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or 154 Val385Ile in SEQ ID NO: 78. 12. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 13. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment. 14. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment.
. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory disease or condition. 16. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 17. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 155 18. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratoiy distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 19. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736.
. The ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. (00592] In an embodiment of the invention, there are provided methods as per the following clauses:- 1. A method of treating or reducing the risk of an lL6R-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 an IL6R protein that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78, wherein the ligand comprises a human IGLC1 *01 lambda chain constant region and wherein said human comprises (i) a human IGLCI *01 lambda chain constant region gene segment, or the human expresses antibodies comprising human IGLCI *01 lambda chain constant regions and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78.
In an example, said ligand, antibody or antibody fragment has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 2. The method of any preceding clause comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of clause 1. 3. The method of any preceding clause, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 156 4. The method of any one of clauses 1 to 3, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358A)a and/or Val385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human.
. The method of clause 4, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78. 6. The method of clause 5, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 7. The method of clause 5, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 8. The method of clause 5 or 7, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 9. The method of any preceding clause, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or 157 Val385Ile in SEQ ID NO: 78.
. The method of any preceding clause, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate. 11. The method of any preceding clause, wherein said human is receiving or has received an antiTNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. 12. The method of any preceding clause, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 13. The method of any preceding clause, wherein said disease or condition is an inflammatory disease or condition. 14. The method of any preceding clause, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis.
. The method of any preceding clause, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 158 16. The method of any preceding clause, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 17. The method of any preceding clause, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 22. The method of any preceding clause, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-593" id="p-593"
[00593] In an embodiment of the invention, there are provided methods as per the following paragraphs:- 1. An anti-IL6R ligand (eg, an antibody or fragment) for treating or reducing the risk of an IL6Rmediated disease or condition in a human in need thereof, wherein the ligand specifically binds an 1L6R protein that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78, wherein the ligand comprises a human IGLC1 *01 lambda chain constant region and wherein said human comprises (i) a human IGLCl*01 lambda chain constant region gene segment, or the human expresses antibodies comprising human IGLC1 *01 lambda chain constant regions and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385He in SEQ ID NO: 78.
In an example, said ligand has been determined to specifically bind an IL6R protein that comprises a mutation Asp358Ala or Val3 85Ile in SEQ ID NO: 78 2. The ligand of paragraph 1, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78; and/or an IL6R protein comprising said mutation Asp358Ala and/or Val3851Ie in SEQ ID NO: 78. 3. The ligand of paragraph 1 or 2, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile; and/or an 1L6R protein comprising said mutation Asp358Ala and/or Val385IIe, 159 optionally, wherein the determining step is performed before administration of the antibody to the human. 4. The ligand of paragraph 3, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a 1L6R protein comprising said mutation Asp358Ala and/or Val385IIe in SEQ ID NO: 78.
. The ligand of paragraph 4, wherein 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 an 1L6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the 1L6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 6. The ligand of paragraph 4, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 7. The ligand of paragraph 4 or 6, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 8. The ligand of any preceding paragraph, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 160 9. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is or has been further determined to be substantially resistant an anti-TNF alpha treatment, In any embodiment herein "an anti-TNF alpha treatment" is selected from the group consisting of an infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), and etanercept (eg, Enbrel®) treatment with or without methotrexate.
. The ligand of any preceding paragraph, wherein said ligand is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an antiTNF alpha treatment. 11. The ligand of any preceding paragraph, wherein said ligand is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 12. The ligand of any preceding paragraph, wherein said disease or condition is an inflammatory disease or condition. 13. The ligand of any preceding paragraph, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versushost disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 14. The ligand of any preceding paragraph, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
. The ligand of any preceding paragraph, wherein said ligand treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel 161 disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 16. The ligand of any preceding paragraph, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 17. Phe ligand of any preceding paragraph, wherein said ligand is for intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-594" id="p-594"
[00594] Regimens id="p-595" id="p-595"
[00595] A: The invention further provides the following regimens, ligands and kits. 1. A method for treating a human IL6R-mediated disease or condition in a human by targeting a rare variant human IL6R, the method comprising administering to the human a ligand (eg, an antibody or fragment) that has been determined to specifically bind to said lL6Rvariant; wherein the human expresses said IL6R variant or the genome of the human comprises a nucleotide sequence encoding said IL6R variant; wherein said human is treated for said disease or condition.
The variant IL6R can, for example, be an IL6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. In an example, the IL6R comprises a mutation Asp358Ala in SEQ ID NO: 78. In an example, the IL6R comprises a mutation Val385Ile in SEQ ID NO: 78.
In an example, the IL6R is a a human soluble IL6R.
In an example, the IL6R is a a human cell-suface IL6R.
In an embodiment, determination of said specific binding is by reference to binding assay data, eg, as determined using SPR or ELISA. Determination may, for example, be by reference to information in a printed publication, eg, with knowledge of data presented in the present or another patent application or in a journal article. Once armed with such knowledge (eg, in the absence of further testing of binding), the skilled person is able - by direction of the present invention - to treat a relevant human whose genotype or phenotype matches the binding specificity of the ligand. 162 The antibody or fragment can be according to any configuration, example, embodiment, aspect, clause or paragraph herein.
In an embodiment, the method comprises, before said administering, selecting a human comprising said nucleotide sequence encoding the IL6R, wherein the human is said human in clause 1 (eg, la). 2. The method of clause 1, comprising before said administering the step of determining that the ligand specifically binds to said IL6R, eg, using SPR or ELISA. 3. The method of clause 1 or 2, wherein the specific binding to said IL6R is binding with a dissociation constant (Kd) of InM or less, eg, 100, 10 or IpM or less. 4. The method of any of clauses 1 to 3 (eg, clause la), wherein said disease or condition is an inflammatory disease or condition.
. The method of clause 4 (eg, when dependent from clause la), wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
In any embodiment herein, the arthritis is moderate to severe rheumatoid arthritis or Systemic juvenile idiopathic arthritis. 6. The method of any one of clauses 1 to 5 (eg, when dependent from clause la), wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R comprising a mutation Asp358Ala or Val38511e in SEQ ID NO: 78 and/or a IL6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 7. The method of any one of clauses 1 to 6 (eg, when dependent from clause la), comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78 and/or a IL6R protein comprising a mutation Asp358Ala or Val38511e in SEQ ID NO: 78, optionally, wherein the 163 determining step is performed before administration of the antibody to the human. 8. The method of clause 7, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the IL6R that encodes an IL6R comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 9. The method of clause 8, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the . The method of clause 8 or 9, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 11. The method of any one of clauses 1 to 10, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment. 12. The method of any one of clauses 1 to 11, wherein said human is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. 13. The method of clauses II or 12, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said anti-TNF alpha treatment. 14. The method of any one of clauses 8 to 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human.
. The method of any one of clauses 1 to 14, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or 164 Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 16. The method of any one of clauses 1 to 1 5, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 17. The method of any one of clauses 1 to 16, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 18. The method of any one of clauses 1 to 17, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 19. The method of any one of clauses I to 18, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation.
. A ligand (eg, an antibody or fragment) for use in the method of any one of clauses 1 to 19, wherein the ligand specifically binds the IL6R. 21. A kit comprising the ligand of clause 20 and instructions for carrying out the method of any one of clauses 1 to 19. 165 id="p-596" id="p-596"
[00596] B: The invention further provides the following regimens, ligands and kits. 1. A method of treating or reducing the risk of an IL6R-mediated disease or condition in a human in need thereof, the method comprising:- a. Carrying out an initial treatment of said human for an initial treatment period by administering an anti-human IL6R ligand (eg, an antibody or fragment) to said human, wherein (i) the ligand has been determined to specifically bind to an 1L6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78; (ii) the human expresses said IL6R or the genome of the human comprises a nucleotide sequence encoding said JL6R and (iii) the human has received or is receiving an anti-TNF alpha treatment for treating or reducing the risk of an IL6R-mediated disease or condition; wherein the initial treatment comprises the administration of a single or multiple doses of the ligand to the human; b. Determining to (i) terminate anti-TNF alpha treatment (ii) keep the human off anti-TNF alpha treatment; or (iii) reduce anti-TNF alpha treatment after said initial treatment period; and c. Continuing to administer the ligand to said patient after said time period has expired, thereby treating or reducing the risk of an lL6R-mediated disease or condition in said human.
In an embodiment, determination of said specific binding is by reference to binding assay data, eg, as determined using SPR or ELISA. Determination may, for example, be by reference to information in a printed publication, eg, with knowledge of data presented in the present or another patent application or in a journal article. Once armed with such knowledge (eg, in the absence of further testing of binding), the skilled person is able - by direction of the present invention - to treat a relevant human whose genotype or phenotype matches the binding specificity of the ligand.
The antibody or fragment can be according to any configuration, example, embodiment, aspect, clause or paragraph herein.
A pharmaceutically-effective amount of said ligand is administered.
In an embodiment, the method comprises, before said administering, selecting a human comprising said nucleotide sequence encoding the IL6R, wherein the human is said human recited in clause 1.
In an example, the initial treatment period is 7 days, 14 days, 21 days, 28 days, a month, two months, three months, four months, five months, six months, seven months, eight months, nine months or a 166 year. 2. The method of clause 1, wherein the human has or is suffering from an anti-TNF alpha treatmentassociated side effect.
An example of a side effect is selected from the group consisting of hepatitis B infection in a carrier of the virus, an allergic reaction, a nervous system condition, a blood condition, heart failure, an immune reaction (eg, lupus-like syndrome), a liver condition, and new or worsening psoriasis. In an example, the treatment is adalimumab treatment. 3. The method of clause 1 or 2, comprising, before said initial treatment, the step of determining that the human has or is suffering from an anti-TNF alpha treatment-associated side effect. 4. The method of clause 2 or 3, comprising, after step (b) (eg, during step (c)) determining that the side effect has lessened.
. The method of any one of clauses 1 to 4, wherein said human is over 40 years of age (eg, 50 or over, 55 or over, 60 or over, 65 or over, or 70 or over). 6. The method of any one of clauses 1 to 5, wherein step (c) comprises determining to increase the doses of said ligand to be administered after said initial treatment period and administering said increased doses to said human. 7. The method of any one of clauses 1 to 6, wherein step (b) comprises determining that the human is intolerant or refactoiy to treatment by an anti-TNF alpha treatment. 8. The method of any one of clauses 1 to 7, wherein the initial treatment comprises the administration of infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), or etanercept (eg, Enbrel®) treatment, with or without methotrexate, to the human in addition to the ligand. 9. The method of any one of clauses 1 to 8, wherein step (b) comprises terminating or reducing infliximab (eg, Remicade®), adalimumab (eg, Humira®), certolizumab pegol (eg, Cimzia®), golimumab (eg, Simponi®), or etanercept (eg, Enbrel®) treatment during step (c). 167 . The method of any one of clauses 1 to 9, comprising increasing (ie, increasing compared to the initial treatment dose) the ligand dose during step (c). 11. The method of any one of clauses 1 to 10, wherein the human has received high dose anti-TNF alpha treatment prior to the initial treatment, and wherein step (c) comprises administering a lower (eg, a medium or low) dose anti-TNF alpha treatment in addition to said ligand.
The skilled person is familiar with the meaning of high, medium and low dose treatments (and how to determine according to each patient, eg, the patient’s body mass). 12. The method of any one of clauses 1 to 10, wherein the human has received medium dose antiTNF alpha treatment prior to the initial treatment, and wherein step (c) comprises administering a lower (eg, a low) dose statin treatment or no anti-TNF alpha in addition to said ligand. 13. The method of any one of clauses 1 to 10, wherein the human has received low dose anti-TNF alpha treatment prior to the initial treatment, and wherein step (c) comprises administering no anti-TNF alpha in addition to said ligand. 14. The method of any one of clauses 1 to 13, comprising, before the initial treatment, the step of determining that the ligand specifically binds to said IL6R, eg, using SPR or ELISA.
. The method of any one of clauses 1 to 14, wherein the specific binding to said 1L6R is binding with a dissociation constant (Kd) of InM or less, eg, 100, 10 or IpM or less. 16. The method of any of clauses 1 to 15, wherein the human is at risk of or suffering from a condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 17. The method of clause 16, wherein step (c) treats or reduces the risk of said condition in the human. 18. The method of any one of clauses 1 to 17, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R comprising a mutation Asp358Ala or Val385Ile in 168 SEQ ID NO: 78 and/or an IL6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 19. The method of any one of clauses 1 to 18, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R comprising a mutation Asp358Ala or Val385lie in SEQ ID NO: 78 and/or an IL6R protein comprising a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein the determining step is performed before administration of the antibody to the human.
. The method of clause 19, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the IL6R that comprises a mutation Asp358Ala or Val385He in SEQ ID NO: 78. 21. The method of clause 20, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ue in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala and/or Val385lie in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the 1L6R protein comprising said mutation Asp358Ala and/or Val38511e in SEQ ID NO: 78. 22. The method of clause 20 or 21, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 23. The method of any one of clauses 1 to 22, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha (eg, adalimubab, infliximab or etanercept) treatment. 169 24. The method of any one of clauses 20 to 22, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human.
. The method of any one of clauses 1 to 24, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Aia or Val385Ile in SEQ ID NO: 78. 26. The method of any one of clauses I to 25, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease. 27. The method of any one of clauses 1 to 26, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 28. The method of any one of clauses 1 to 27, wherein said ligand (eg, antibody or antibody fragment) is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 29. A ligand (eg, an antibody or fragment) for use in the method of any one of clauses 1 to 28, wherein the ligand specifically binds the IL6R.
. A kit comprising the ligand of clause 29 and instructions for carrying out the method of any one of clauses 1 to 28. [00 597J In an example of any aspect of the invention, the ligand (eg, antibody or fragment, eg, sarilumab) is administered to the human at a two-weekly dose of from 75 to 200mg (eg, from 150 to 1200mg administered once or twice over a two-week period, eg, 150mg or 200mg once a week or once every other week). In an example, the ligand is for such administration to the human. id="p-598" id="p-598"
[00598] 170 id="p-599" id="p-599"
[00599] rsl2083537 id="p-600" id="p-600"
[00600] With reference to any configuration, aspect, embodiment or example of the invention described herein, in an embodiment the human comprises an IL6R nucleotide sequence comprising mutation 2913A>G (SEQ ID NO: 79) (ie, wherein the human comprises SNP rsl 2083537). In an example, the human has been genotyped for said mutation. In an example, the method comprises genotyping the human for said mutation (eg, prior to the administration of the ligand). In an example, the human is suffering from or at risk of suffering from asthma. In an example, the ligand or method is for treating, preventing, or reducing the risk of asthma in the human. In an example, the ligand or method treats or reduces the risk of asthma in the human.
J00601] Thus, in an alternative to any configuration, aspect, embodiment or example of the invention herein, instead of the phrase "wherein said human comprises a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78", it can instead be read "wherein said human comprises a nucleotide sequence encoding an IL6R protein, wherein the nucleotide sequence comprises a mutation 2913A>G in SEQ ID NO: 79" or "wherein said human comprises a nucleotide sequence encoding an IL6R protein, wherein the nucleotide sequence comprises SNP rs 12083 53 7" or "wherein the genome of said human comprises SNP rs 12083537". Additionally, in an example, said IL6R protein comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. id="p-602" id="p-602"
[00602] Some embodiments of the technology described herein can be defined according to any of the following numbered paragraphs: 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 a. Administering to the human an anti-TOI ligand to target a TOI variant in the human and treat or prevent said disease or condition, wherein the TOI in said human is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence having a total human genotype frequency of less than 50%; wherein b. Before step (a) said human has been or is genotyped as positive for said nucleotide sequence or phenotyped as positive for said.TOI variant. 2. The method of paragraph I, wherein before step (a) the ligand has been or is determined as being capable of binding to said TOI variant. 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 171 a. Administering to the human an anti-TOl ligand to target a TOI variant in the human and treat or prevent said disease or condition, wherein the TOI in said human is encoded by a nucleotide sequence having a cumulative human allele frequency of less than 50% and/or wherein the TOI in said human is encoded by a nucleotide sequence having a total human genotype frequency of less than 50%; wherein b. Before step (a) the ligand has been or is determined as capable of binding to said TOI variant. 4. The method of paragraph 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 method of paragraph 3 or 4, wherein said human has been or is genotyped as positive for said variant nucleotide sequence before step (a). 6. The method of any preceding paragraph, wherein the human has been or is phenotyped as positive for said TOI variant before step (a). 7. The method of any preceding paragraph, wherein said frequency is less than 10 or 15%. 8. The method of any preceding paragraph, 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% and/or having a total human genotype frequency of less than 50%. 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 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% and/or having a total human genotype frequency of more than 50%; wherein b. Before 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%.
. The method of paragraph 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. 172 11. The method of paragraph 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. 12. 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). 13. The method of any one of paragraphs 9 to 12, wherein the ligand is capable of binding to the most frequent TOI variant. 14. The method of any one of paragraphs 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%.
. The method of any preceding paragraph, 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. 16. The method of any preceding paragraph, wherein 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. 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. 18. The ligand of paragraph 17, wherein the ligand has been or is determined as capable of binding the human TOI encoded by said nucleotide sequence. 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 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.
. The ligand of any one of paragraphs 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%.
The ligand of any one of paragraphs 17 to 19, wherein the human has been or is genotyped as positive for said TOI nucleotide sequence having a total human genotype frequency of less than 50%. 21. The ligand of any one of paragraphs 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%. 22. The ligand of any one of paragraphs 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 173 than 50% and/or having a total human genotype frequency of less than 50%; optionally wherein 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% and/or having a total human genotype frequency of more than 50%. 23. The ligand of any one of paragraphs 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%. 24. The ligand of any one of paragraphs 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.
. The ligand of paragraph 24, wherein the ligand is an antibody or antibody fragment. 26. The ligand of any one of paragraphs 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. 27. The ligand of any one of paragraphs 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. 28. 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. 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 174 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, the method comprising immunising a nonhuman 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. 31. 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. 32. A kit for TOI genotyping a human, wherein 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 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. 33. A kit 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. 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 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%.
. 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. 36. 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 175 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. 37. The method of paragraph 36, 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%. 38. 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%. 39. 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%. 40. 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. 41. 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. 42. 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. 43. 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. 44. The method, ligand, composition, kit or use of any preceding paragraph, wherein the TOI is encoded by a nucleotide sequence having a cumulative human allele frequency from I to 10% and/or a total human genotype frequency from 1 to about 15% or from 1 to 15%. 45. The method, ligand, composition, kit or use of any preceding paragraph wherein 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. 46. The ligand, method, use, kit or composition of any preceding paragraph, wherein (i) the ligand (eg, antibody or fragment) comprises 176 (e) a 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 (f) a constant region domain encoded by a C region gene segment; Wherein 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; and (ii) 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. 47. The ligand, method, use, kit or composition of paragraph 46, wherein 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. 48. The ligand, method, use, kit or composition of paragraph 47, wherein SPR is used to cany out said assay. 49. 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). 50. 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. 1. The ligand, method, use, kit or composition of paragraph 50, wherein said human expresses an antibody variable domain comprising said first and second amino acid polymorphisms. 52. The ligand, method, use, kit or composition of paragraph 50 or 51, wherein the first and second SNPs of said genome are comprised by the same antibody gene segment. 53. The ligand, method, use, kit or composition of any one of paragraphs 46 to 52, wherein each SNP is a variable region gene segment SNP. 54. The ligand, method, use, kit or composition of any one of paragraphs 46 to 52, wherein 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. 177 55. 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. 56. The ligand, method, use, kit or composition of any one of paragraphs 46 to 53, wherein each SNP is a variable domain SNP, eg, a VH domain SNP, ora Vk domain SNP, or a νλ SNP. 57. The ligand, method, use, kit or composition of any one of paragraphs 46 to 56, wherein said constant region domain of (b) is comprised by an antibody Fc region. 58. The ligand, method, use, kit or composition of any one of paragraphs 46 to 57, wherein the ligand (eg, antibody or fragment) 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 lOpM or less) as determined by SPR. 59. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 58), 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). 60. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 59), 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. 61. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 60), 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 CH 1 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. 62. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 61), 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 178 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. 63. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 62), wherein, 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. 64. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 63), 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. 65. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of paragraphs 46 to 64), 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. 66. The ligand, method, use, kit or composition of any one of paragraphs 61 to 65, wherein said human gamma heavy chain is a human gamma-1 heavy chain. 67. The ligand, method, use, kit or composition of any one of paragraphs 61 to 65, wherein said human gamma heavy chain is a human gamma-2 heavy chain. 68. The ligand, method, use, kit or composition of any one of paragraphs 61 to 65, wherein ligand comprises a human IGHGl*01 gamma-1 heavy chain constant region. 69. The ligand, method, use, kit or composition of any one of paragraphs 61 to 65, wherein ligand comprises a human 1GHG2*O1 gamma-1 heavy chain constant region. 70. The ligand, method, use, kit or composition of any one of paragraphs 60 to 69, wherein the human has been or is genotyped as positive for said heavy chain constant region nucleotide sequence. 71. The ligand, method, use, kit or composition of paragraph 68, wherein the human has been or is genotyped as positive for human IGHGl*01 nucleotide sequence. 72. The ligand, method, use, kit or composition of paragraph 69, wherein the human has been or is genotyped as positive for human IGHG2*01 nucleotide sequence. 73. The ligand, method, use, kit or composition of any one of paragraphs 61 to 69, wherein the human has been or is phenotyped as positive for said gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc. 179 74. The ligand, method, use, kit or composition of paragraph 63, (i) when dependent from clause 23, wherein the human has been or is phenotyped as positive for a human IGHG1 *01 gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc or (ii) when dependent from clause 24, wherein the human has been phenotyped as positive for a human IGHG2*01 gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc. 75. The method or use of 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, CFI4 or Fc nucleotide sequence. 76. The method or use of any one of paragraphs 61 to 69 and 71 to 75, comprising phenotyping the 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 1GHG2*O1 gamma heavy chain constant domain, CHI, CH2, CH3, CH4 or Fc. 77. The ligand, method, use, kit or composition of any preceding paragraph (eg, according to any one of clauses 46 to 76), 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. 78. The ligand, method, use, kit or composition of paragraph 77,wherein the ligand comprises a 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. 79. The ligand, method, use, kit or composition of paragraph 77 or 78,wherein the genome of the human comprises a gamma-1 heavy chain constant region nucleotide sequence that encodes such an Asp and Leu or the human expresses antibodies comprising human gamma-1 constant regions comprising such an Asp and Leu. 80. The ligand, method, use, kit or composition of paragraph 77, 78 or 79, wherein the ligand comprises a human IGHGl*01 gamma-1 heavy chain constant region, eg, an Fc, CHI, CH2 and/or CH3 domain encoded by human IGHG1 *01. 81. The ligand, method, use, kit or composition of any one of paragraphs 77 to 80, wherein the genome of the human comprises a human IGHG1*O1 nucleotide sequence or the human expresses 180 antibodies comprising human constant domains encoded by a human IGHG 1*01 nucleotide sequence. 82. The ligand, method, use, kit or composition of any one of paragraphs 77 to 81, wherein the ligand comprises a hinge region encoded by human IGHGl*01. 83. The ligand, method, use, kit or composition of any one of paragraphs 77 to 82, wherein the ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 61. 84. The ligand, method, use, kit or composition of any one of paragraphs 77 to 83, wherein the human is of European ancestry. 85. The ligand, method, use, kit or composition of any one of paragraphs 77 to 84, wherein the human has been or is genotyped as positive for said Asp and/or Leu. 86. The ligand, method, use, kit or composition of any one of paragraphs 77 to 85, wherein the human has been or is genotyped as positive for human IGHG) *01. 87. The ligand, method, use, kit or composition of any one of paragraphs 77 to 86, wherein the human has been or is phenotyped as positive for a human IGHG1*OJ CH3. 88. The method or use of any one of paragraphs 77 to 87, comprising selecting a said human whose genome comprises a codon(s) encoding said Asp and/or Leu; comprises human IGHG 1 *01; or comprises a human IGHGl*01 CH3. 89. The method or use of any one of paragraphs 77 to 88, comprising selecting a said human whose phenotype comprises said Asp and/or Leu; a human IGHG 1*01 region ; or a human IGHG 1*01 CH3. 90. The ligand, method, use, kit or composition of any preceding paragraph, 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. 91. The ligand, method, use, kit or composition of paragraph 90, wherein 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 181 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). 92. The ligand, method, use, kit or composition of paragraph 90 or 91, wherein the 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). 93. The ligand, method, use, kit or composition of any one of paragraph 90 to 92, wherein the 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. 94. The ligand, method, use, kit or composition of any one of paragraphs 90 to 93, wherein the 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. 95. The ligand, method, use, kit or composition of any one of paragraphs 90 to 94, wherein the ligand comprises a hinge region encoded by human IGHG2*01. 96. The ligand, method, use, kit or composition of any one of paragraphs 90 to 95, wherein the ligand comprises or consists of an antibody, wherein the antibody comprises heavy chains that comprise SEQ ID NO: 63 or 65. 97. The ligand, method, use, kit or composition of any one of paragraphs 90 to 96, wherein the human is of European, African American, or European American ancestry. 98. The ligand, method, use, kit or composition of any one of paragraphs 90 to 97, wherein the human has been or is genotyped as positive for one, more or all of said Pro, Asn, Phe, Val and Ala. 99. The ligand, method, use, kit or composition of any one of paragraphs 90 to 98, wherein the human has been or is genotyped as positive for human IGHG2*01. 100. The ligand, method, use, kit or composition of any one of paragraphs 90 to 99, wherein the human has been or is phenotyped as positive for a human IGHG2*01 CH 1. 101. The ligand, method, use, kit or composition of any one of paragraphs 90 to 100, wherein the human has been or is phenotyped as positive for a human IGHG2*01 CH2. 102. The ligand, method, use, kit or composition of any one of paragraphs 90 to 101, wherein the human has been or is phenotyped as positive for a human 1GHG2*O] CH3. 182 103. The method or use of any one of paragraphs 90 to 102, comprising selecting a said human whose genome comprises a codon(s) encoding one, more or all of said Pro, Asn, Phe, Val and Ala; comprises human IGHG2*01; or comprises a human IGHG2*01 CHI, CH2 and/or CH3. 104. The method or use of any one of paragraphs 90 to 103, 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. 105. The ligand, method, use, kit or composition of any one of paragraphs 90 to 104, wherein the human expresses antibodies comprising human gamma-2 constant regions comprising such a Pro, Asn, Phe, Val and Ala. 106. The ligand, method, use, kit or composition of any preceding paragraph, 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. 107. The ligand, method, use, kit or composition of paragraph 106, wherein the 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. 108. The ligand, method, use, kit or composition of paragraph 106 or 107, wherein the genome of the human comprises a kappa light chain constant region nucleotide sequence that encodes such a Val and Cys or the human expresses antibodies comprising human kappa constant regions comprising such a Val and Cys. 109. The ligand, method, use, kit or composition of any one of paragraphs 106 to 108, wherein the antibody or fragment comprises a human IGKC^Ol kappa light chain constant region. 110. The ligand, method, use, kit or composition of any one of paragraphs 106 to 109, wherein the ligand comprises or consists of an antibody, wherein the antibody comprises light chains that comprise SEQ ID NO: 62 or 66.
HE The ligand, method, use, kit or composition of any one of paragraphs 106 to 110, wherein the 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 1GKJ2*O1 (SEQ ID NO: 57). 112. The ligand, method, use, kit or composition of any one of paragraphs 106 to 111, wherein the human has been or is phenotyped as positive for said Val and/or Cys. 113. The ligand, method, use, kit or composition of any one of paragraphs 106 to 112, wherein the human has been or is genotyped as positive for human IGKC*01. 183 114. The ligand, method, use, kit or composition of any one of paragraphs 106 to 113, wherein the human has been or is phenotyped as positive for a human IGKC*01 domain. 115. The method or use of any one of paragraphs 106 to 114, comprising selecting a said human whose genome comprises a codon(s) encoding said Val and/or Cys; or comprises human IGKC*01. 116. The method or use of any one of paragraphs 106 to 115, comprising selecting a said human whose phenotype comprises such a Val and/or Cys; or comprises a human IGKC*01 domain. 7. The ligand, method, use, kit or composition of any one of paragraphs 106 to 116, wherein the human expresses antibodies comprising human kappa constant domains comprising such a Val and Cys, eg, expresses human IGKC*01 constant domains. 118. The ligand, method, use, kit or composition of any preceding paragraph, 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. 119. The ligand, method, use, kit or composition of paragraph 1 18, wherein the antibody comprises light chains that comprise SEQ ID NO: 64. 120. The ligand, method, use, kit or composition of paragraph 118 or 119, wherein the human has been or is genotyped as positive for human IGLC2*01. 121. The ligand, method, use, kit or composition of any one of paragraphs 118 to 120, wherein the human has been or is phenotyped as positive for a human IGLC2*01 domain. 122. The method or use of any one of paragraphs 118 to 212, comprising selecting a said human whose genome comprises human IGLC2*01. 123. The method or use of any one of clauses 73 to 77, comprising selecting a said human whose phenotype comprises a human IGLC2*01 domain. 124. The ligand, method, use, kit or composition of any one of paragraphs 108 to 123, wherein the human expresses antibodies comprising human lambda IGLC2*01 constant domains. 125. The ligand, method, use, kit or composition of any preceding paragraph, 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 IGHV118*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHVl-18*01; or (ii) IGVH1-46*O1 and the genome of the human comprises a human IGHV 1-46*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV 1-46*01. 184 126. The ligand, method, use, kit or composition of paragraph 125, wherein the antibody or fragment comprises a one more or all of a CHI domain, CH2 domain, CH3 domain, hinge or Fc encoded by human IGHG2*01. 127. The ligand, method, use, kit or composition of paragraph 125 or 126, wherein the antibody or fragment comprises heavy chains that comprise SEQ ID NO: 63 or 65. 128. The ligand, method, use, kit or composition of any one of paragraphs 125 to 127, wherein the human has been or is genotyped as positive for said selected VH gene segment, positive for human IGHV 1-18*01 or IGVH1 -46*01. 129. The method or use of any of paragraphs 125 to 128, comprising genotyping the human as positive for said selected VH gene segment, eg, positive for human IGHV1-18*01 or IGVH 1-46*01. 130. The ligand, method, use, kit or composition any preceding paragraph, 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) IGKV41*01 and the genome of the human comprises a human IGKV4-I*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV4J *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*O2 and the genome of the human comprises a human IGKV1-13*02 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKVL13*02. 131. The ligand, method, use, kit or composition of paragraph 130, wherein the antibody comprises light chains that comprise SEQ ID NO: 62, 64 or 66. 132. The ligand, method, use, kit or composition of paragraph 130 or 131, wherein 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. 133. The ligand, method, use, kit or composition of any one of paragraphs 130 to 132, wherein the human has been or is genotyped as positive for said selected VL gene segment, eg, positive for human IGKV4-l*01, IGLV2-14*01 or IGKV1-13*O2. 134. The method or use of paragraph 133, 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 IGKVl-13*02. 135. The ligand, method, use, kit or composition of any preceding paragraph, wherein the ligand (eg, antibody or fragment) binds said human TOI with a dissociation constant (Kd) of InM or less as determined by SPR, (eg, 100, 10 or IpM or less). 185 136. The ligand, method, use, kit or composition of any preceding paragraph, wherein the TOI is human PCSK9 or human IL-6R.
EXAMPLES id="p-603" id="p-603"
[00603] Example 1; Rare PCSK9 Variants id="p-604" id="p-604"
[00604] Proprotein convertase subtilisin kexin type 9 (PCSK9) is a serine protease involved in regulating the levels of the low density lipoprotein receptor (LDLR) protein (Horton et al., 2007; Seidah and Prat, 2007). In vitro experiments have shown that adding PCSK9 to HepG2 cells lowers the levels of cell surface LDLR (Benjannet et al., 2004; Lagace et al., 2006; Maxwell et al., 2005; Park et al., 2004). Experiments with 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 coimmunofluoresce with the LDLR throughout the endosomal pathway (Lagace et al., 2006). id="p-605" id="p-605"
[00605] 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. Crystal and NMR structures of different domains from mouse furin and PCI reveal subtilisin-like pro- and catalytic domains, and a P domain directly C-terminal to the catalytic domain (Henrich et al., 2003; Tangrea et al., 2002). Based on the amino acid sequence similarity within this subfamily, all seven members are predicted to have similar structures (Henrich et al., 2005). SKI-1/S1P and PCSK9 are classified in subfamily S8A. Sequence comparisons with these proteins also suggest the presence of subtilisin-like pro- and catalytic domains (Sakai et aL, 1998; Seidah et al., 2003; Seidah et al., 1999). In these proteins the amino acid sequence C-terminal to the catalytic domain is more variable and does not suggest the presence of a P domain. id="p-606" id="p-606"
[00606] 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. Following this initial cleavage reaction, the pro-domain remains bound to the catalytic domain where it then acts as an inhibitor of catalytic activity (Fu et aL, 2000). When conditions are correct, 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. 186 id="p-607" id="p-607"
[00607] Autocatalysis of the PCSK9 zymogen occurs between Gin 152 and Seri 53 (VFAQ^SIP (SEQ ID NO: 67)) (Naureckiene et al., 2003), and has been shown to be required for its secretion from cells (Seidah et al., 2003). A second autocatalytic event at a site within PCSK9's pro-domain has not been observed. Purified PCSK9 is made up of two species that can be separated by non-reducing SDS-PAGE; the pro-domain at 17 Kd, and the catalytic plus C-terminal domains at 65 Kd. PCSK9 has not been isolated without its inhibitory pro-domain, and measurements of PCSK9’s catalytic activity have been variable (Naureckiene et al., 2003; Seidah et aL, 2003). id="p-608" id="p-608"
[00608] In certain embodiments, 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, NARCL 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. The term "PCSK9" denotes both the proprotein and the product generated following autocatalysis of the proprotein. When only the autocatalyzed product is being referred to (such as for an antigen binding protein or ligand that binds to the cleaved PCSK9), the protein can be referred to as the "mature," "cleaved", "processed" or "active" PCSK9. When only the inactive form is being referred to, the protein can be referred to as the "inactive", "pro-form", or "unprocessed" form of PCSK9. The term 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. IA and IB of US20120093818A1; which is incorporated by reference herein in its entirety). id="p-609" id="p-609"
[00609] 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. id="p-610" id="p-610"
[00610] Anti-PCSK9 ligands (eg, antibodies and anti-sense RNA) 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. In this way, the inventor arrived at insight into the natural occurrences and distributions of rarer human PCSK9 forms that can serve as useful targets (at the protein or nucleic acid level) for human treatment, prophylaxis 187 and diagnosis pertinent to diseases and conditions mediated or associated with PCSK9 activity. This particularly provides for tailored therapies, prophylaxis and diagnosis in humans that are devoid of the common PCSK9 gene or protein (ie, the form a or a’ as used in US20120093818A1 and US20110065902A1 to generate antibodies). id="p-611" id="p-611"
[00611] The skilled person will know that SNPs or other changes that translate into amino acid variation can cause variability in activity and/or conformation 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 more effectively tailor medicines and diagnosis of patients. The invention, therefore, provides for tailored pharmaceuticals and testing that specifically addresses rarer PCSK9 polymorphic variant forms. Such forms or "alleles" (at the nucleotide level), in many of the examples determined by the inventor, 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. id="p-612" id="p-612"
[00612] Furthermore, the inventor surprisingly realised that the rarer natural forms, although present in humans at much lower frequencies than the common form, nevertheless are represented in multiple and ethnically-diverse human populations and usually with many human examples per represented ethnic population. Thus, the inventor realised that targeting such rarer forms would provide for effective treatment, prophylaxis or diagnosis across many human ethnic populations, thereby extending the utility of the present invention. id="p-613" id="p-613"
[00613] With this realisation, 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. In this way, the patient receives drugs and ligands that are tailored to their needs as determined by the patient's genetic or phenotypic makeup. Hand-in-hand with this, 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. id="p-614" id="p-614"
[00614] In developing this thinking, 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%; 188 • PCSK9 variants found in many different human ethnic populations (using the standard categorisation of the 1000 Genomes Project, which is an accepted standard in the art; see Table 4 below); and ♦ PCSK9 variants found in many individuals distributed across such many different ethnic populations. id="p-615" id="p-615"
[00615] On the basis of these criteria, the inventor identified the variants listed in Table 1 below (excluding form rz). id="p-616" id="p-616"
[00616] 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), 10 as opposed to silent variations that do not alter amino acid residues in the target protein. 189 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% (a) Amino acid variability, population distributions and frequencies 0.64815 Cum Freq6 0.0855 0.0729 i 0.0292 0.0221 0.0149 । 8900Ό 0.0045 0.0041 0.0032 0.4506 (0.8457) Hom Freq4 (Het + Hom freq5) 0.009 (0.162) 0.0081 (0.1377) 0.009 (0.0324) (0.0441) (0.0225) (0.0135) (600’0) (Τ800Ό) (0.0063) 0.3951 Het Freq3 0.153 0.1296 0.0234 o o 0.0225 1 0.0135 CD O O 0 0.0081 0.0063 14 No. Unique Pops2 12 12 cn CO CO LD 939 No. Individs1 180 153 49 cn ΓΝ i 15 | io ASW,YRI,GBR, TSI,CLM,CHB, LWK,CHS,MXL, J PT, PUR, IBS, FIN, CEU Human Populations . 5 vf IXL co 00 > "k ϋ > CX CL D > 5 Η H § % LC UO -a — < 1- X Li- I x £ 2 S CC 5 X 2 > 5 <-> Lu § " < 2 iw'Nid'sai 'Hnd'ldr'lATD 'ISra8£)'MSV LWK,ASW,YRI, CLM I LWK,ASW,YRI >ί iZ) < § Σ> LU CJ U_ Co H Ctl D CL CHS, ASW; J PT, PUR,CHB 670E Amino Acid Position & Variation 670G X X X 619Q 619P X 4741 474V X X X X X 443A 443T X X 425N 425S X 53A 53V X X 46R 46L X Form a Variant Form M- u Q. E OJ St ‘ro CT (zzmpTempText 190 £ ο <υ Ε ό ο. ο I ο -α ω όϋ <υ -η Ε 3 <ν -C 0) CL .— CO -Ο Ε 3 C ‘ο <3 Ο C Ε < C ο ΙΛ Ο Ο. (zzmpTempText 191 (b) Nucleotide Sequence Variations of Selected Alleles < ________Nucleotide Position1__ 1:55529187 ______ Non-Synonymous Nucleotide Variation2 __1 CD \ ___Variant ID3 __________________________________________~J □Ί m o vn k_ Corresponding Amino Acid Variation __________ | 67OG X X X "x" in a box indicates that a variant allele comprises the non-synonymous nucleotide variation indicated in the 5th row. < 1:55527222 u CM CM LD co 00 CM v> 619P X < 1:55524237 rs562556 474V X X X X X o 1:55523855 < rs28362263 443T X X < 1:55523802 o rs28362261 425S X o 1:55505668 h— Q oo LD CO oo Ln rH r-i tn 1______53V_______I X X o 1:55505647 H rsll591147 I 1. 46L_______I X Allele a Variant Allele H— w k. Q. E ω £ 'ra ET allele a): and 3. NCBI dbSNP reference number (NCBI dbSNP Build 138 released on Apr 25, 2013). 192 IX no co CD CO o cd id lu co CD Cd c 00 (ϋ cr CD CD Σ cr LU 0 (a) Human PCSK9 Form a Amino Acid Sequence (SEQ ID NO:1) - Pro-form with Signal Sequence P 0 cn S 0) CD in id rd Id rf) ijD LL hd Cd Cd kJ Q Cd Oi Ιό c CD s CD Cli id Q X id c? ο pt, g td cn Oi cn EH Cd E £ 00 (D ‘5 CD 05 £ co Φ Cl CD CD co E E CD 2 oo CD rb no cu CO CO no on CD CD Q. ex uO o* > CH Q Cd rd QD CO .2? co E V) CD ω CD > 00 CD 73 k00 co cb co co Ί GO (U CD bO a CD d u. 00 QO ¢0 CD 00 £ > & (D (O CO Ql (O CO cr iZ LU LU 1/1 CO CO a-*’ $ s1 CD (D C| (Z) C/) LU LU 0 LU (Z I— 0 □J LU tsi LU s a IP S' CZ) CA Φ V) CXJ II JH <υ H 193 C4 cr O E o cd C ύ al ω cu cu □ E o 4= <υ c CD P cr o w E <2 I = « s -C (Λ O uu O ω < o CM LC cr t-H ID O r-ID AVDNTCWRSR DVSTTGSTSEEAVTAVAICCRSR H LAQASQE LQ 194 CJ ID id ID A 0 0 A A O u CJ ID rd rd 0 rd 0 co (c) Human PCSK9 Allele a Nucleotide Sequence (SEQ ID NO: 28) - Encoding Pro-form1' Plus Signal cd ID ID ID cd CD ID CD ID U ID CD in ID CD CD £ 0 A 0 O rd 0 0 rd 0 U rd A 0 Cj rd 0 8 CD δ CD CD ΰ CD O 0 D 0 d u A rd U rd O o rd 0 0 O ri» 0 0 rd 0 O rd 0 0 rd 0 0 0 A 0 A U 0 rd U U U rd rd 0 A u A C A rd TO TO no no no TO r;ij rd 0 0 A 0 0 A rd 0 rd o u r-< 0 Q 0 rd rd 0 0 0 d 0 ω 0 0 rd 0 0 A U 0 0 0 o 0 0 0 A 0 rd A 0 0 0 0 A 0 0 s 0 A U 0 rd 0 u 0 0 rd U 0 0 A O u E-l rd TO £ CD no no u no TO no no TO TO no no to u TO O no ro 0 rd 0 U U 0 A A o A 0 A A U rd 0 0 0 rd 0 0 rd 0 rd A 0 rd 0 to TO no no TO no TO u TO TO & TO TO CJ TO no TO TO no to no CO no no no no TO no no TO no no u CD no no TO TO CD TO no TO no no no no no cj CJ TO no no no CD CJ TO no no to co 8 no TO no no co no TO o TO £ CJ no co no CO re no no co no no no CD CD no co no TO no CD CJ to co no no no co u no no no co no co CD HO TO no no no no no bo no no no υ CD TO no no u no CJ no no co u no cj no TO CJ no TO no TO TO no np no u no no no TO no no no u no TO CJ tb no no no co u no bO TO CJ no no no no no TO no no tJ no TO no CD co TO TO no no £ TO no TO no no no no TO ω TO no no CJ no TO no no CJ co CJ no no no co TO £ TO CD no no no ϋ no co u to no no no no no no no no CD no CJ no no no CJ no CD CTO UO no co TO to & no CD TO ϊΜ no no gP no no no no no TO no no no no ti TO no 4-1 no no no TO TO no TO TO TO co TO no no no no Ei no CD TO TO tb no TO no no TO no co TO u no no TO no no TO (J TO no no no u TO u no TO ύ no TO no CJ no co t u no no no C TO TO u CD no no co no no no no TO no CD no no no &0 no no TO TO TO no TO no CD no TO o no no CD no CD S’ CJ no no TO u t no TO no no no TO no HO tuO no CD no TO no TO no £ TO cj no no no no tv no no no no TO CJ TO U TO CJ TO no Φ u C TO D σ o> (zzmpTempText 195 UQ N42.5S AAT to AG AWT G CC to ACC CO cj u cj CD bO £ bO ba ω uo ro CD col «J bO ro tuO cv ¢0 ro u ra CO 0 (J 0 < 0 CJ h— 0 l·CJ CJ <£ 0 CJ < CJ CJ < u CJ CJ < (J CJ CJ CJ CJ u CJ l·CJ < (J CJ CJ < CJ CJ < (J CJ CJ CJ CJ CJ CJ CJ CJ < CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ o CJ l·— < CJ CJ CJ CJ CJ < CJ CJ < CJ < Q.
CJ CJ CJ CJ CJ CJ < 0 CJ CJ CJ CJ CJ CJ < CJ CJ CJ CJ U CJ CJ 5 CJ < CJ CJ < CJ CJ q CJ u CJ CJ o CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CJ CD CD H CJ CD CI CD o o UJ CJ < u CD u CJ CJ CJ CJ < CJ CJ CD CJ CJ CD CD CD CD CJ CJ CJ 0 CJ 0 0 CD CD CJ u 0 < 0 0 0 < Ch kO •YD I O .5 σ3 P o X3 cj < 0 0 < μ0 CO .E 45 o o G 0> Q rO 3 σo Q 3 II C3 O Um ω £ o UPPER CASE = nucleotide sequence encoding C-terminal domain (nucleotides 1347-2076) Underlined = allelic variations from allele a in other sequences (aa residue number changes and codon changes shown) The pro-form is encoded by nucleotide sequence from nucleotide 91 to (and including) nucleotide 2076. The mature form is encoded by nucleotide sequence from nucleotide 457 to (and including) nucleotide 2076. 196 id="p-617" id="p-617"
[00617] Variant Allele Nucleotide Sequences Thus, (i) The nucleotide sequence of allele/is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele / comprises a GTC codon instead of an ATC codon at the position labelled 1474V" in SEQ ID NO: 28; (ii) The nucleotide sequence of allele c is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele c comprises a GGG codon instead of an GAG codon at the position labelled E670G in SEQ ID NO: 28; (iii) The nucleotide sequence of allele r is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele r comprises a GTC codon instead of an ATC codon at the position labelled I474V in SEQ ID NO: 28; and a GGG codon instead of an GAG codon at the position labelled E670G in SEQ ID NO: 28; (iv) The nucleotide sequence of allele p is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele p comprises a GTC codon instead of a GCC codon at the position labelled A53 V in SEQ ID NO: 28; and a GTC codon instead of an ATC codon at the position labelled I474V in SEQ ID NO: 28; (v) The nucleotide sequence of allele m is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele m comprises a ACC codon instead of a GCC codon at the position labelled A443T in SEQ ID NO: 28; (vi) The nucleotide sequence of allele e is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele e comprises a AGT codon instead of an AAT codon at the position labelled N425S in SEQ ID NO: 28; and a GTC codon instead of an ATC codon at the position labelled 1474V in SEQ ID NO: 28; (vii) The nucleotide sequence of allele h is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele h comprises a ACC codon instead of a GCC codon at the position labelled "A443T in SEQ ID NO: 28; and a CCG codon instead of a CAG codon at the position labelled Q619P in SEQ ID NO: 28; (viii) The nucleotide sequence of allele aj is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele aj comprises a CTT codon instead of an CGT codon at the position labelled R46L in SEQ ID NO: 28; and a GTC codon instead of an ATC codon at the position labelled I474V in SEQ ID NO: 28; and (ix) The nucleotide sequence of allele q is identical to SEQ ID NO: 28 except that the nucleotide sequence of allele q comprises a GTC codon instead of a GCC codon at the position labelled A53V in SEQ ID NO: 28; and a GGG codon instead of an GAG codon at the position labelled E670G in SEQ ID NO: 28. id="p-618" id="p-618"
[00618] Variant Pro-Form Amino Acid Sequences (Numbering is as per SEQ ID NO: 1 recited above) 197 (A) The amino acid sequence of form/is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form f comprises a valine at position 474; (B) The amino acid sequence of form c is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form c comprises a glycine at position 670; (C) The amino acid sequence of form r is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form r comprises a valine at position 474 and a glycine at position 670; (D) The amino acid sequence of form p is identical the amino acid sequence from amino acid number 3 1 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form p comprises a valine at position 53 and a valine at position 474; (E) The amino acid sequence of form m is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form m comprises a threonine at position 443; (F) The amino acid sequence of form e is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form e comprises a serine at position 425 and a valine at position 474; (G) The amino acid sequence of form h is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form h comprises a threonine at position 443 and a proline at position 619; (H) The amino acid sequence of form aj is identical to the amino acid sequence from amino acid number 31 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form aj comprises a leucine at position 46 and a valine at position 474; and (I) The amino acid sequence of form q is identical to the amino acid sequence from amino acid number 3 1 to (and including) amino acid number 692 of SEQ ID NO: 1 except that the amino acid sequence of form q comprises a valine at position 53 and a glycine at position 670. id="p-619" id="p-619"
[00619] Variant Mature Form Amino Acid Sequences (Numbering is as per SEQ ID NO: 1 recited above) (Af ) The amino acid sequence of form/is identical to SEQ ID NO: 2 except that the amino acid sequence of form/comprises a valine at position 474; (B’ ) The amino acid sequence of form c is identical to SEQ ID NO: 2 except that the amino acid sequence of form c comprises a glycine at position 670; (C) The amino acid sequence of form r is identical to SEQ ID NO: 2 except that the amino acid sequence of form r comprises a valine at position 474 and a glycine at position 670; (D’ ) The amino acid sequence of form p is identical to SEQ ID NO: 2 except that the amino acid sequence of form p comprises a valine at position 474; 198 (E1 ) The amino acid sequence of form m is identical to SEQ ID NO: 2 except that the amino acid sequence of form m comprises a threonine at position 443; (F) The amino acid sequence of form e is identical to SEQ ID NO: 2 except that the amino acid sequence of form e comprises a serine at position 425 and a valine at position 474; (G1) The amino acid sequence of form h is identical to SEQ ID NO: 2 except that the amino acid sequence of form h comprises a threonine at position 443 and a proline at position 619; (H’) The amino acid sequence of form aj is identical to SEQ ID NO: 2 except that the amino acid sequence of form aj comprises valine at position 474; and (Γ) The amino acid sequence of form q is identical to SEQ ID NO: 2 except that the amino acid sequence of form q comprises a glycine at position 670. id="p-620" id="p-620"
[00620] The mature form of p is identical to the mature form of/and aj. |00621] The mature form of c is identical to the mature form of q. id="p-622" id="p-622"
[00622] Further sequence analysis and 3D in silico modelling (see Figure 1) revealed that selected variants also fulfilled the following selection criteria:- • PCSK9 variants whose variant amino acid residues (versus the common form of human PCSK9) are found in the mature form of the target (ie, outside the pro-domain); and • PCSK9 variants whose variant amino acid residues (versus the common form of human PCSK9) are surface-exposed on the target, which the inventor saw as contributing to determining the topography of the target and potentially contributing to how and where ligand binding on the target occurs. id="p-623" id="p-623"
[00623] As shown in Figure 1, identified positions 425, 443, 474, 619 and 670 (found in the selected variants of the invention) are all surface-exposed and outside of the pro-domain. Variant positions 425 and 443 are surface-exposed on the catalytic domain, while variant positions 474, 619 and 670 are surface-exposed on the C-terminal domain. id="p-624" id="p-624"
[00624] In a first example, the invention addresses the need to treat humans having naturallyoccurring rarer natural PCSK9 alleles, genotypes and phenotypes (rarer protein forms). In this respect, the invention provides the following aspects: id="p-625" id="p-625"
[00625] In a First Aspect: An anti-human PCSK9 ligand for use in a method of treating and/or preventing a PCSK9-mediated disease or condition in a human whose genome comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37, wherein the method comprises administering the ligand to the human. id="p-626" id="p-626"
[00626] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. 199 id="p-627" id="p-627"
[00627] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-628" id="p-628"
[00628] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). |00629| In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 3 1, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 3 1,32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-630" id="p-630"
[00630] In an example, the nucleotide sequence is SEQ ID NO: 29. id="p-631" id="p-631"
[00631] In an example, the nucleotide sequence is SEQ ID NO: 30. [00632| In an example, the nucleotide sequence is SEQ ID NO: 31. id="p-633" id="p-633"
[00633] In an example, the nucleotide sequence is SEQ ID NO: 32. id="p-634" id="p-634"
[00634] In an example, the nucleotide sequence is SEQ ID NO: 33. id="p-635" id="p-635"
[00635] In an example, the nucleotide sequence is SEQ ID NO: 34. id="p-636" id="p-636"
[00636] In an example, the nucleotide sequence is SEQ ID NO: 35. id="p-637" id="p-637"
[00637] In an example, the nucleotide sequence is SEQ ID NO: 36. id="p-638" id="p-638"
[00638] In an example, the nucleotide sequence is SEQ ID NO: 37. id="p-639" id="p-639"
[00639] In a Second Aspect: The ligand of aspect 1, wherein the ligand has been or is determined as capable of binding a human PCSK9 selected from the group consisting forms f c, r, p, m, et h, aj and q. id="p-640" id="p-640"
[00640] In an example of any aspect, the ligand binds (or has been determined to bind) two, three, four or more human PCSK9 selected from the group consisting forms f c, r. p, e, h aj and q. [00641] In an example of any aspect, the ligand comprises a protein domain that specifically binds to PCSK9, eg, a human PCSK9 selected from the group consisting forms f c, r, p, m, e, h, aj and q. [00642] The term "specifically binds," or the like, means that a ligand, eg, an antibody or antigenbinding 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 1 xlO-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 PCSK9 may, however, exhibit cross-reactivity to other antigens such as a PCSK9 molecule from another specie. Moreover, multi-specific antibodies (e.g., bispecifics) that bind to human PCSK9 and one or more additional antigens are nonetheless considered antibodies that "specifically bind" PCSK9, as used herein. 200 id="p-643" id="p-643"
[00643] In an example of any aspect, the ligand comprises or consists of a protein that mimics the EGFA domain of the LDL receptor and specifically binds to PCSK9, eg, a human PCSK9 selected from the group consisting forms/ c, r, p, m, e, h, aj and q. id="p-644" id="p-644"
[00644] In an example of any aspect, the ligand antagonises PCSK9, eg, a human PCSK9 selected from the group consisting forms f c, r, p, m, e, h, aj and q. id="p-645" id="p-645"
[00645] In an example of any aspect, the method comprises (before administering the ligand) the step of determining that the ligand is capable of binding a human PCSK9 selected from the group consisting forms f, c, r, p, m, e, h, aj and q. [00646| In an example of any aspect, binding is determined by SPR. In an example of any aspect, binding is determined by ELISA. id="p-647" id="p-647"
[00647] In an example of any aspect, said forms are the mature forms. id="p-648" id="p-648"
[00648] In an example of any aspect, said forms are the pro-forms. id="p-649" id="p-649"
[00649] In a Third Aspect: A ligand that binds a human PCSK9 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 4-27 for use in a method comprising the step of using the ligand to target said PCSK9 in a human to treat and/or prevent a disease or condition mediated by PCSK9, the method comprising administering the ligand to the human. id="p-650" id="p-650"
[00650] In an example, the disease or condition is mediated by a human PCSK9 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 4-27. . id="p-651" id="p-651"
[00651] In an example, the amino acid sequence selected from the group consisting of SEQ ID NOs: 4-23, 26 and 27; or selected from the group consisting of SEQ ID NOs: 4-14 and 18-27; or selected from the group consisting of SEQ ID NOs: 4-14, 18-23, 26 and 27. These are naturallyoccurring sequences that do not comprise 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-652" id="p-652"
[00652] In an example, the amino acid sequence is SEQ ID NO: 18, 19 or 20, that comprises a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-653" id="p-653"
[00653] In an example, the amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 11, 12, 26 and 27, that comprise 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). id="p-654" id="p-654"
[00654] In an example, the amino acid sequence selected from the group consisting of SEQ ID NOs: 10-14 and 18-27; or selected from the group consisting of SEQ ID NOs: 10-14, 18-23, 26 and 27. These are sequences that have a naturally-occurring combination of differences from SEQ ID NOs: 1-3 (form a) and which meet the criteria set out above. id="p-655" id="p-655"
[00655] In an example, the amino acid sequence is SEQ ID NO:4. id="p-656" id="p-656"
[00656] In an example, the amino acid sequence is SEQ ID NO:5. id="p-657" id="p-657"
[00657] In an example, the amino acid sequence is SEQ ID NO:6. id="p-658" id="p-658"
[00658] In an example, the amino acid sequence is SEQ ID NO:7. id="p-659" id="p-659"
[00659] In an example, the amino acid sequence is SEQ ID NO:8. 201 id="p-660" id="p-660"
[00660] In an example, the amino acid sequence is SEQ ID NO: 9. id="p-661" id="p-661"
[00661] In an example, the amino acid sequence is SEQ ID NO: 10. id="p-662" id="p-662"
[00662] In an example, the amino acid sequence is SEQ ID NO: 11. id="p-663" id="p-663"
[00663] In an example, the amino acid sequence is SEQ ID NO: 12. id="p-664" id="p-664"
[00664] In an example, the amino acid sequence is SEQ ID NO: 13. id="p-665" id="p-665"
[00665] In an example, the amino acid sequence is SEQ ID NO: 14. id="p-666" id="p-666"
[00666] In an example, the amino acid sequence is SEQ ID NO: 15. id="p-667" id="p-667"
[00667] In an example, the amino acid sequence is SEQ ID NO: 16. id="p-668" id="p-668"
[00668] In an example, the amino acid sequence is SEQ ID NO: 17. id="p-669" id="p-669"
[00669] In an example, the amino acid sequence is SEQ ID NO: 18. id="p-670" id="p-670"
[00670] In an example, the amino acid sequence is SEQ ID NO: 19. id="p-671" id="p-671"
[00671] In an example, the amino acid sequence is SEQ ID NO: 20. id="p-672" id="p-672"
[00672] In an example, the amino acid sequence is SEQ ID NO: 21. id="p-673" id="p-673"
[00673] In an example, the amino acid sequence is SEQ ID NO: 22. id="p-674" id="p-674"
[00674] In an example, the amino acid sequence is SEQ ID NO: 23. id="p-675" id="p-675"
[00675] In an example, the amino acid sequence is SEQ ID NO: 24. id="p-676" id="p-676"
[00676] In an example, the amino acid sequence is SEQ ID NO: 25. |00677] In an example, the amino acid sequence is SEQ ID NO: 26. 100678] In an example, the amino acid sequence is SEQ ID NO: 27. (00679] In a Fourth Aspect: The ligand of aspect 3, wherein the genome of the human comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37. id="p-680" id="p-680"
[00680] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-681" id="p-681"
[00681] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-682" id="p-682"
[00682] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et <7/2005). id="p-683" id="p-683"
[00683] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-684" id="p-684"
[00684] In an example, the nucleotide sequence is SEQ ID NO: 29 [00685] In an example, the nucleotide sequence is SEQ ID NO: 30. 202 id="p-686" id="p-686"
[00686] In an example, the nucleotide sequence is SEQ ID NO: 31. id="p-687" id="p-687"
[00687] In an example, the nucleotide sequence is SEQ ID NO: 32. id="p-688" id="p-688"
[00688] In an example, the nucleotide sequence is SEQ ID NO: 33 id="p-689" id="p-689"
[00689] In an example, the nucleotide sequence is SEQ ID NO: 34 id="p-690" id="p-690"
[00690] In an example, the nucleotide sequence is SEQ ID NO: 35. id="p-691" id="p-691"
[00691] In an example, the nucleotide sequence is SEQ ID NO: 36. id="p-692" id="p-692"
[00692] In an example, the nucleotide sequence is SEQ ID NO: 37. id="p-693" id="p-693"
[00693] In a Fifth Aspect: The ligand of any preceding aspect, wherein the human has been or is genotyped as positive for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof. id="p-694" id="p-694"
[00694] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-695" id="p-695"
[00695] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S. which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-696" id="p-696"
[00696] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). id="p-697" id="p-697"
[00697] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31,32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32. 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-698" id="p-698"
[00698] In an example, the nucleotide sequence is SEQ ID NO:29. id="p-699" id="p-699"
[00699] In an example, the nucleotide sequence is SEQ ID NO:30. id="p-700" id="p-700"
[00700] In an example, the nucleotide sequence is SEQ ID NO:31. id="p-701" id="p-701"
[00701] In an example, the nucleotide sequence is SEQ ID NO:32. id="p-702" id="p-702"
[00702] In an example, the nucleotide sequence is SEQ ID NO:33. id="p-703" id="p-703"
[00703] In an example, the nucleotide sequence is SEQ ID NO:34. (00704] In an example, the nucleotide sequence is SEQ ID NO:35. id="p-705" id="p-705"
[00705] In an example, the nucleotide sequence is SEQ ID NO:36. id="p-706" id="p-706"
[00706] In an example, the nucleotide sequence is SEQ ID NO:37. id="p-707" id="p-707"
[00707] In a Sixth Aspect: The ligand of any preceding aspect, wherein the human has been or is phenotyped as positive for a human PCSK9 selected from the group consisting of forms f, c, r, p, m, e, h, aj and q or at least the catalytic or C-terminal domain thereof. id="p-708" id="p-708"
[00708] In an example, said forms are the mature forms. 203 id="p-709" id="p-709"
[00709] In an example, said forms are the pro-forms. id="p-710" id="p-710"
[00710] In a Seventh Aspect: The ligand of any preceding aspect, wherein the method comprises genotyping the human as positive for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof. id="p-711" id="p-711"
[00711] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-712" id="p-712"
[00712] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-713" id="p-713"
[00713] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et a/2005). id="p-714" id="p-714"
[00714] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31,32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-715" id="p-715"
[00715] In an example, the nucleotide sequence is SEQ ID NO: 29. id="p-716" id="p-716"
[00716] In an example, the nucleotide sequence is SEQ ID NO: 30. id="p-717" id="p-717"
[00717] In an example, the nucleotide sequence is SEQ ID NO: 31. id="p-718" id="p-718"
[00718] In an example, the nucleotide sequence is SEQ ID NO: 32. id="p-719" id="p-719"
[00719] In an example, the nucleotide sequence is SEQ ID NO: 33. id="p-720" id="p-720"
[00720] In an example, the nucleotide sequence is SEQ ID NO: 34. id="p-721" id="p-721"
[00721] In an example, the nucleotide sequence is SEQ ID NO: 35. id="p-722" id="p-722"
[00722] In an example, the nucleotide sequence is SEQ ID NO: 36. id="p-723" id="p-723"
[00723] In an example, the nucleotide sequence is SEQ ID NO: 37. id="p-724" id="p-724"
[00724] In an Eighth Aspect: The ligand of any preceding aspect, wherein the method comprises phenotyping the human has positive for a human PCSK9 selected from the group consisting of forms / c, r, p, m, e, h, aj and q or at least the catalytic or C-terminal domain thereof. id="p-725" id="p-725"
[00725] In an example, said forms are the mature forms. id="p-726" id="p-726"
[00726] In an example, said forms are the pro-forms. id="p-727" id="p-727"
[00727] In a Ninth Aspect: The ligand of any preceding aspect, wherein the human has been or is genotyped as heterozygous for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof; optionally wherein the human has been or is genotyped as comprising the nucleotide sequence of SEQ 204 ID NO: 28 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof and a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof. id="p-728" id="p-728"
[00728] "Heterozygous" here means that in the human’s genotype one allele comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof and other allele can be any PCSK9 (eg, form a, a' or an allele comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain-or C-terminal domain-encoding sequence thereof). id="p-729" id="p-729"
[00729] In an example, the method comprises (before administering the ligand) genotyping the human as heterozygous for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof; optionally also genotyping the human as comprising the nucleotide sequence of SEQ ID NO: 28 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof and a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or Cterminal domain-encoding sequence thereof. id="p-730" id="p-730"
[00730] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. id="p-731" id="p-731"
[00731] These are naturally-occurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-732" id="p-732"
[00732] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-733" id="p-733"
[00733] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et «/2005). id="p-734" id="p-734"
[00734] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31,32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-735" id="p-735"
[00735] In an example, the nucleotide sequence is SEQ ID NO:29. id="p-736" id="p-736"
[00736] In an example, the nucleotide sequence is SEQ ID NO:30. id="p-737" id="p-737"
[00737] In an example, the nucleotide sequence is SEQ ID NO:3L id="p-738" id="p-738"
[00738] In an example, the nucleotide sequence is SEQ ID NO:32. id="p-739" id="p-739"
[00739] In an example, the nucleotide sequence is SEQ ID NO:33. id="p-740" id="p-740"
[00740] In an example, the nucleotide sequence is SEQ ID NO:34. 205 id="p-741" id="p-741"
[00741] In an example, the nucleotide sequence is SEQ ID NO:35. id="p-742" id="p-742"
[00742] In an example, the nucleotide sequence is SEQ ID NO:36. id="p-743" id="p-743"
[00743] In an example, the nucleotide sequence is SEQ ID NO:37. id="p-744" id="p-744"
[00744] In a Tenth Aspect: The ligand of any one of aspects 1 to 9, wherein the genome of the human has been or is genotyped as homozygous for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof. id="p-745" id="p-745"
[00745] "Homozygous" here means that in the human’s genotype each allele comprises the same nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof. id="p-746" id="p-746"
[00746] In an example, the method comprises genotyping the human as homozygous for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C-terminal domain-encoding sequence thereof. id="p-747" id="p-747"
[00747] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-748" id="p-748"
[00748] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-749" id="p-749"
[00749] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). id="p-750" id="p-750"
[00750] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a} and which meet the criteria set out above. id="p-751" id="p-751"
[00751] In an example, the nucleotide sequence is SEQ ID NO: 29. id="p-752" id="p-752"
[00752] In an example, the nucleotide sequence is SEQ ID NO: 30. id="p-753" id="p-753"
[00753] In an example, the nucleotide sequence is SEQ ID NO: 31. id="p-754" id="p-754"
[00754] In an example, the nucleotide sequence is SEQ ID NO: 32. id="p-755" id="p-755"
[00755] In an example, the nucleotide sequence is SEQ ID NO: 33. id="p-756" id="p-756"
[00756] In an example, the nucleotide sequence is SEQ ID NO: 34. id="p-757" id="p-757"
[00757] In an example, the nucleotide sequence is SEQ ID NO: 35. id="p-758" id="p-758"
[00758] In an example, the nucleotide sequence is SEQ ID NO: 36. id="p-759" id="p-759"
[00759] In an example, the nucleotide sequence is SEQ ID NO: 37. 206 id="p-760" id="p-760"
[00760] In an Eleventh Aspect: The ligand of any preceding aspect, wherein the ligand comprises an antibody binding site that binds a human PCSK9 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 4-27 and optionally has been or is determined as capable of such binding. id="p-761" id="p-761"
[00761] In an example, the method comprises (before administering the ligand) the step of determining that the ligand is capable of binding to said human PCSK9. [00762| In an example, the binding is specific binding. In an example, the ligand binds (or has been determined as binding) to the PCSK9 with an affinity (Kd) of ImM, ΙΟΟηΜ, lOnM or InM or less. In an embodiment, the affinity is no less than 10, 100 or 1000 fM. [00763| In an example, binding or affinity is determined by SPR or ELISA. id="p-764" id="p-764"
[00764] In an example, the disease or condition is mediated by a human PCSK9 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 4-27. id="p-765" id="p-765"
[00765] In an example, the amino acid sequence selected from the group consisting of SEQ ID NOs: 4-23, 26 and 27; or selected from the group consisting of SEQ ID NOs: 4-14 and 18-27; or selected from the group consisting of SEQ ID NOs: 4-14, 18-23, 26 and 27. These are naturallyoccurring sequences that do not comprise 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-766" id="p-766"
[00766] In an example, the amino acid sequence is SEQ ID NO: 18, 19 or 20, that comprises a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-767" id="p-767"
[00767] In an example, the amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 11, 12, 26 and 27, that comprise 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). id="p-768" id="p-768"
[00768] In an example, the amino acid sequence selected from the group consisting of SEQ ID NOs: 10-14and 18-27; or selected from the group consisting of SEQ ID NOs: 10-14, 18-23,26 and 27. These are sequences that have a naturally-occurring combination of differences from SEQ ID NOs: 1 -3 (form a) and which meet the criteria set out above id="p-769" id="p-769"
[00769] In an example, the amino acid sequence is SEQ ID NO: 4. id="p-770" id="p-770"
[00770] In an example, the amino acid sequence is SEQ ID NO: 5. id="p-771" id="p-771"
[00771] In an example, the amino acid sequence is SEQ ID NO: 6. id="p-772" id="p-772"
[00772] In an example, the amino acid sequence is SEQ ID NO: 7. id="p-773" id="p-773"
[00773] In an example, the amino acid sequence is SEQ ID NO: 8. id="p-774" id="p-774"
[00774] In an example, the amino acid sequence is SEQ ID NO: 9. id="p-775" id="p-775"
[00775] In an example, the amino acid sequence is SEQ ID NO: 10. id="p-776" id="p-776"
[00776] In an example, the amino acid sequence is SEQ ID NO: 11. id="p-777" id="p-777"
[00777] In an example, the amino acid sequence is SEQ ID NO: 12. id="p-778" id="p-778"
[00778] In an example, the amino acid sequence is SEQ ID NO: 13. id="p-779" id="p-779"
[00779] In an example, the amino acid sequence is SEQ ID NO: 14. 207 id="p-780" id="p-780"
[00780] In an example, the amino acid sequence is SEQ ID NO: 15. id="p-781" id="p-781"
[00781] In an example, the amino acid sequence is SEQ ID NO: 16. id="p-782" id="p-782"
[00782] In an example, the amino acid sequence is SEQ ID NO: 17. id="p-783" id="p-783"
[00783] In an example, the amino acid sequence is SEQ ID NO: 1 8. id="p-784" id="p-784"
[00784] In an example, the amino acid sequence is SEQ ID NO: 19. id="p-785" id="p-785"
[00785] In an example, the amino acid sequence is SEQ ID NO: 20. {00786] In an example, the amino acid sequence is SEQ ID NO: 21. id="p-787" id="p-787"
[00787] In an example, the amino acid sequence is SEQ ID NO: 22. f00788] In an example, the amino acid sequence is SEQ ED NO: 23. id="p-789" id="p-789"
[00789] In an example, the amino acid sequence is SEQ ID NO: 24. id="p-790" id="p-790"
[00790] In an example, the amino acid sequence is SEQ ID NO: 25. id="p-791" id="p-791"
[00791] In an example, the amino acid sequence is SEQ ID NO: 26. id="p-792" id="p-792"
[00792] In an example, the amino acid sequence is SEQ ID NO: 27. id="p-793" id="p-793"
[00793] In a Twelfth Aspect: The ligand of aspect 11, wherein the ligand is an antibody or antibody fragment. For example, the antibody or antibody fragment is a PCSK9 antagonist, eg, neutralises PCSK9. id="p-794" id="p-794"
[00794] Examples of such antibodies are disclosed, for instance, in WO 2008/057457, WO2008/057458, WO 2008/057459, WO 2008/063382, WO 2008/133647, WO 2009/100297, WO 2009/100318, WO 201 1/037791, WO 201 1/053759, WO 201 1/053783, WO 2008/125623, WO 2011/072263, WO 2009/055783, WO 2010/029513, WO 2011/11 1007, WO 2010/077854, the disclosures and sequences of such antibodies being incorporated herein for use in the invention in their entireties by reference. One specific example is AMG 145 (Amgen), LY3015014 (Eli Lilly) or aliiOcumab. Advantageously, the ligand is or comprises alirocumab. Alternatively, the ligand is or comprises evolocumab. |00795] In an example, the ligand is SAR236553/REGN727 (Sanofi Aventis/Regeneron) or a PCSK9-binding derivative thereof. id="p-796" id="p-796"
[00796] In an example, the ligand comprises or consists of a neutralizing antibody that binds to the PCSK9, wherein the antibody binds to PCSK9 and reduces the likelihood that PCSK9 binds to LDLR. id="p-797" id="p-797"
[00797] The ligand of aspect 11, wherein the ligand is a PCSK9 antagonist, eg, neutralises PCSK9. id="p-798" id="p-798"
[00798] In an example of any aspect of the invention, the ligand comprises or consists a ligand selected from evolocumab, lD05-IgG2 (Merck & Co.), ALN-PCS02 (Alnylam), RN316 (PfizerRinat), LY3015014 (Eli Lilly) and alirocumab (SAR236553/REGN727; Sanofi Aventis/Regeneron). [00799] In Thirteenth Aspect: The ligand of any one of aspects 1 to 10, wherein (i) the ligand comprises a sequence of contiguous nucleotides that specifically hybridises to a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or C 208 terminal domain-encoding sequence thereof, or specifically hybridises to an antisense sequence or an RNA transcript of said sequence, wherein said sequence of contiguous nucleotides hybridises to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridises to an antisense sequence or an RNA transcript thereof respectively; and/or (ii) the ligand comprises a sequence of at least 10 contiguous nucleotides of a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or is an antisense sequence or RNA version of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28. id="p-800" id="p-800"
[00800] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-801" id="p-801"
[00801] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-802" id="p-802"
[00802] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005).
J00803] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. |00804] In an example, the nucleotide sequence is SEQ ID NO:29. 100805] In an example, the nucleotide sequence is SEQ ID NO:30. id="p-806" id="p-806"
[00806] In an example, the nucleotide sequence is SEQ ID NO:31. id="p-807" id="p-807"
[00807] In an example, the nucleotide sequence is SEQ ID NO:32. (00808] In an example, the nucleotide sequence is SEQ ID NO:33. id="p-809" id="p-809"
[00809] In an example, the nucleotide sequence is SEQ ID NO.34. id="p-810" id="p-810"
[00810] In an example, the nucleotide sequence is SEQ ID NO:35. id="p-811" id="p-811"
[00811] In an example, the nucleotide sequence is SEQ ID NO:36. id="p-812" id="p-812"
[00812] In an example, the nucleotide sequence is SEQ ID NO:37. id="p-813" id="p-813"
[00813] In an embodiment, the ligand comprises at least 10, 11,12, 13, 14, 15,20, 25,30,35,40, 45, 50 or 100 contiguous nucleotides of said nucleotide sequence. id="p-814" id="p-814"
[00814] In a Fourteenth Aspect: The ligand of any preceding aspect, wherein said disease or condition is hyperlipidaemia, hypercholesterolaemia (eg, familial hypercholesterolaemia), heart attack, stroke, coronary heart disease, atherosclerosis or a cardiovascular disease or condition. 209 id="p-815" id="p-815"
[00815] The ligand of any preceding aspect, wherein the disease or condition is hypercholesterolemia, hyperlipidemia, hypercholesterolemia, dyslipidemia, cholestatic liver disease, nephrotic syndrome, hypothyroidism, obesity, atherosclerosis or a cardiovascular disease. id="p-816" id="p-816"
[00816] In an example, said disease or condition is hypercholesterolaemia. The term "hypercholesterolaemia," as used herein, refers to a condition in which cholesterol levels are elevated above a desired level. In some embodiments, this denotes that serum cholesterol levels are elevated. In some embodiments, the desired level takes into account various "risk factors" that are known to one of skill in the art (and are described or referenced in US20120093818). id="p-817" id="p-817"
[00817] The ligand of any preceding aspect, wherein the human is identified as heterozygous for Familial Hypercholesterolemia, statin intolerant, statin uncontrolled, or at risk for developing hypercholesterolemia, dyslipidemia, cholestatic liver disease, nephrotic syndrome, hypothyroidism, obesity, atherosclerosis or a cardiovascular disease. id="p-818" id="p-818"
[00818] In a Fifteenth Aspect: The ligand of any preceding aspect, wherein said disease or condition is associated with elevated LDL cholesterol. id="p-819" id="p-819"
[00819] Cholesterol levels are measured in milligrams (mg) of cholesterol per deciliter (dL) of blood in the United States and some other countries. Canada and most European countries measure cholesterol in millimoles (mmol) per liter (L) of blood. Below are general guideline ideal ranges and elevated ranges.
Total cholesterol Total cholesterol* (U.S. and some other countries) (Canada and most of Europe) Below 200 mg/dL Below 5.2 mmol/L Ideal 200-239 mg/dL 5.2-6.2 mmol/L Borderline high 240 mg/dL and above Above 6.2 mmol/L High LDL cholesterol LDL cholesterol* (U.S. and some other countries) (Canada and most ofEurope) 100-129 mg/dL 2.6-3.3 mmol/L Ideal 130-159 mg/dL 3.4-4.1 mmol/L Borderline high 160-189 mg/dL 4.1-4.9 mmol/L High 190 mg/dL and above Above 4.9 mmol/L Very high ^Canadian and European guidelines differ slightly from U.S. guidelines. These conversions are based on U.S. guidelines. id="p-820" id="p-820"
[00820] Elevated LDL cholesterol is, therefore, 160 mg/dL or above (4.1 mmol/L or above). id="p-821" id="p-821"
[00821] In a Sixteenth Aspect: The ligand of any preceding aspect, wherein the ligand inhibits human PCSK9 binding to human LDL receptor and optionally has been or is determined as capable of such inhibition. 210 id="p-822" id="p-822"
[00822] In an example, the method comprises (before administering the ligand) determining that the ligand is capable of such inhibition. id="p-823" id="p-823"
[00823] Inhibition determination is eg, inhibition in a blood or serum sample, at rtp, at ph7, at 37 degrees centigrade and/or under the physiological conditions of a human body. id="p-824" id="p-824"
[00824] In a Seventeeth Aspect: The ligand of any preceding aspect, wherein the human is resistant or substantially resistant to statin (eg, avorstatin and/or fluvastatin) treatment of said disease or condition. id="p-825" id="p-825"
[00825] In an Eighteenth Aspect: The ligand of any preceding aspect, wherein the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human (i) whose genome comprises SEQ ID NO: 29 and wherein the human is of ASW,YRI,GBR,TSL CLM,LWK,MXL,JPT,PUR,IBS,FIN or CEU ancestry; or (ii) whose genome comprises SEQ ID NO: 30 and wherein the human is of ASW,YRI,GBR,TSI,CLM, CHB,LWK,CHS,JPT,PUR,FIN or CEU ancestry; or (iii) whose genome comprises SEQ ID NO: 32 and wherein the human is of ASW,GBR,TSI,CLM, JPT,PUR,IBS,FIN or CEU ancestry; or (iv) whose genome comprises SEQ ID NO: 33 and wherein the human is of LWK,ASW,YRI or CLM ancestry; or (v) whose genome comprises SEQ ID NO: 34 and wherein the human is of LWK,ASW or YRI ancestry; or (vi) whose genome comprises SEQ ID NO: 35 and wherein the human is of PUR,TSI,FIN or CEU ancestry; or (vii) whose genome comprises SEQ ID NO: 36 and wherein the human is of LWK,ASW or YRI ancestry; or (viii) whose genome comprises SEQ ID NO: 37 and wherein the human is of CHS,ASW,JPT,PUR or CHB ancestry. |00826] In a Ninteenth Aspect: The ligand of any preceding aspect, wherein the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human (i) that expresses PCSK9 form f and wherein the human is of ASW,YRI,GBR,TSI,CLM,LWK,MXL,JPT,PUR,IBS,FIN or CEU ancestry; or (ii) that expresses PCSK9 form c and wherein the human is of ASW,YRI,GBR,TSI,CLM,CHB,LWK,CHS,JPT,PUR,FIN or CEU ancestry; or (iii) that expresses PCSK9 formp and wherein the human is of ASW,GBR,TSI,CLM,JPT,PUR,IBS,FIN or CEU ancestry; or (iv) that expresses PCSK9 form m and wherein the human is of LWK,ASW,YRI or CLM ancestry; or (v) that expresses PCSK9 form e and wherein the human is of LWK,ASW or YRI ancestry; or (vi) that expresses PCSK9 form h and wherein the human is of PUR,TSI,FIN or CEU ancestry; or (vii) that expresses PCSK9 form aj and wherein the human is of LWK,ASW or YRI ancestry; or 211 (viii) that expresses PCSK9 form q and wherein the human is of CHS,ASW,JPT,PUR or CHB ancestry. id="p-827" id="p-827"
[00827] In an example, said forms are the mature forms. id="p-828" id="p-828"
[00828] In an example, said forms are the pro-forms. |00829] In a Twentieth Aspect: A pharmaceutical composition or kit for treating and/or preventing a PCSK9-mediated condition or disease (eg, as recited in aspect 14 or 15), the composition or kit comprising a ligand of any preceding aspect and optionally a statin (eg, cerovastatin, atorvastatin, simvastatin, pitavastin, rosuvastatin, fluvastatin, lovastatin or pravastatin); and optionally in combination with a label or instructions for use to treat and/or prevent said disease or condition in a human (eg, covering treatment of a human as recited in aspect 18 or 19); optionally wherein the label or instructions comprise a marketing authorisation number (eg, an FDA or EMA authorisation number); optionally wherein the label or instructions comprise directions to administer alirocumab or evolocumab to said human; optionally wherein the kit comprises an IV or injection device that comprises the ligand (and, eg, also a statin). id="p-830" id="p-830"
[00830] In a Twenty-first Aspect: A method of producing an anti-human PCSK9 antibody binding site, the method comprising obtaining a plurality of anti-PCSK9 antibody binding sites, screening the antibody binding sites for binding to a human PCSK9 selected from the group consisting of forms/ c, r, p, mt e, h, aj and q or a catalytic or C-terminal domain or a peptide thereof that comprises amino acid variation from the corresponding sequence of SEQ ID NO: 1, 2 or 3 and isolating an antibody binding site that binds in the screening step, and optionally producing a form / c, r, p, m, e, h, aj or q PCSK9-binding fragment or derivative of the isolated antibody. id="p-831" id="p-831"
[00831] In an example, said forms are the mature forms. id="p-832" id="p-832"
[00832] In an example, said forms are the pro-forms. id="p-833" id="p-833"
[00833] In an example of this and the next aspect, the plurality of binding sites comprises or consists of a plurality of 4-chain antibodies or fragments thereof, eg, dAbs, Fabs or scFvs. Suitable methods for producing pluralities of binding sites for screening include phage display (producing a phage display library of antibody binding sites), ribosome display (producing a ribosome display library of antibody binding sites), yeast display (producing a yeast display library of antibody binding sites), or immunisation of a non-human vertebrate (eg, a rodent, eg, a mouse or rat, eg, a Velocimouse™, Kymouse™, Xenomouse™, Aliva Mouse™, HuMab Mouse™, Omnimouse™, Omnirat™ or MeMo Mouse™) with a PCSK9 epitope and isolation of a repertoire of antibodyproducing cells (eg, a B-cell, plasma cell or plasmablast repertoire) and/or a repertoire of isolated antibodies. id="p-834" id="p-834"
[00834] In an example, the method comprises selecting one or more antibody binding sites that each specifically binds to a human PCSK9 epitope comprising amino acid variation from the corresponding sequence of SEQ ID NO: 1, 2 or 3. 212 id="p-835" id="p-835"
[00835] In a Twenty-second Aspect: A method of producing an anti-human PCSK9 antibody, the method comprising immunising a non-human vertebrate (eg, a mouse or a rat) with a human PCSK9 comprising an amino acid sequence selected from the group consisting of the amino acid sequences of forms / c, r, p, m, h, aj and q or a catalytic or C-terminal domain or a peptide thereof that comprises amino acid variation from the corresponding sequence of SEQ ID NO: 1,2 or 3 and isolating an antibody that binds a human PCSK9 comprising selected from the group consisting of forms f c, r, p, m, e, h, aj and q or a catalytic or C-terminal domain or a peptide thereof that comprises amino acid variation from the corresponding sequence of SEQ ID NO: 1,2 or 3, and optionally producing a form f c, r, pt m, e, h, aj or q PCSK9-binding fragment or derivative of the isolated antibody. [00836} In an example, said forms are the mature forms. id="p-837" id="p-837"
[00837] In an example, said forms are the pro-forms. id="p-838" id="p-838"
[00838] In a Twenty-third Aspect: The method of aspect 21 or 22, 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. id="p-839" id="p-839"
[00839] For example, the method comprises isolating a cell (eg, B-cell, plasmablast, plasma cell or memory cell) comprising the nucleic acid, wherein the cell is obtained from a non-human vertebrate that has been immunised with the PCSK9 epitope. id="p-840" id="p-840"
[00840] In a Twenty-fourth Aspect: A kit for PCSK9 genotyping a human, wherein the kit comprises a nucleic acid (i) comprising a sequence of 10 or more (eg, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100 or more) contiguous nucleotides that specifically hybridises to a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or at least the catalytic domain- or Cterminal domain-encoding sequence thereof, or specifically hybridises to an antisense sequence or an RNA transcript of said sequence, wherein said sequence of contiguous nucleotides hybridises to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridises to an antisense sequence or an RNA transcript thereof; and/or (ii) comprising a sequence of at least 10 or more (eg, 10, 15,20,30,40,50, 60,70, 80, 90, 100 or more) nucleotides of a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or comprising an antisense sequence or RNA version of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28. id="p-841" id="p-841"
[00841] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. 213 id="p-842" id="p-842"
[00842] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-843" id="p-843"
[00843] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). (00844] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. {00845] In an example, the nucleotide sequence is SEQ ID NO:29. 100846] In an example, the nucleotide sequence is SEQ ID NO:30. id="p-847" id="p-847"
[00847] In an example, the nucleotide sequence is SEQ ID NO:31. id="p-848" id="p-848"
[00848] In an example, the nucleotide sequence is SEQ ID NO:32. id="p-849" id="p-849"
[00849] In an example, the nucleotide sequence is SEQ ID NO:33. id="p-850" id="p-850"
[00850] In an example, the nucleotide sequence is SEQ ID NO:34. id="p-851" id="p-851"
[00851] In an example, the nucleotide sequence is SEQ ID NO:35. id="p-852" id="p-852"
[00852] In an example, the nucleotide sequence is SEQ ID NO:36. id="p-853" id="p-853"
[00853] In an example, the nucleotide sequence is SEQ ID NO:37. id="p-854" id="p-854"
[00854] In a Twenty-fifth Aspect: A kit for PCSK9 genotyping or phenotyping a human, wherein the kit comprises a ligand according to any one of aspects 1 to 19 or an antibody, fragment or derivative produced by the method of any one of aspects 21 to 23. f00855] In a Twenty-sixth Aspect: Use of an anti-PCSK9 ligand that binds a human PCSK9 selected from the group consisting of forms/ c, r, p, m, e, h, aj and q in the manufacture of a medicament for treating and/or preventing a PCSK9-mediated disease or condition in a human whose genome comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37. optionally for treating and/or preventing a PCSK9-mediated disease or condition in a human as recited in aspect 18 or 19. id="p-856" id="p-856"
[00856] In an example, said forms are the mature forms. (00857] In an example, said forms are the pro-forms. id="p-858" id="p-858"
[00858] In a Twenty-seventh Aspect: Use of an anti-PCSK9 ligand that binds a human PCSK9 selected from the group consisting of forms/ c, r, p, m, e, h, aj and q in the manufacture of a medicament for targeting said PCSK9 in a human to treat and/or prevent a disease or condition mediated by PCSK9, optionally for targeting PCSK9 in a human as recited in aspect 18 or 19. id="p-859" id="p-859"
[00859] In an example, said forms are the mature forms. id="p-860" id="p-860"
[00860] In an example, said forms are the pro-forms. id="p-861" id="p-861"
[00861] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or 214 selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-862" id="p-862"
[00862] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-863" id="p-863"
[00863] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). id="p-864" id="p-864"
[00864] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31,32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-865" id="p-865"
[00865] In an example, the nucleotide sequence is SEQ ID NO:29. id="p-866" id="p-866"
[00866] In an example, the nucleotide sequence is SEQ ID NO:30. id="p-867" id="p-867"
[00867] In an example, the nucleotide sequence is SEQ ID NO:31. id="p-868" id="p-868"
[00868] In an example, the nucleotide sequence is SEQ ID NO:32. id="p-869" id="p-869"
[00869] In an example, the nucleotide sequence is SEQ ID NO:33. id="p-870" id="p-870"
[00870] In an example, the nucleotide sequence is SEQ ID NO:34. id="p-871" id="p-871"
[00871] In an example, the nucleotide sequence is SEQ ID NO:35. id="p-872" id="p-872"
[00872] In an example, the nucleotide sequence is SEQ ID NO:36. id="p-873" id="p-873"
[00873] In an example, the nucleotide sequence is SEQ ID NO:37. id="p-874" id="p-874"
[00874] The ligand can be any anti-PCSK9 ligand disclosed herein. id="p-875" id="p-875"
[00875] In a Twenty-eight Aspect: The use of aspect 26 or 27, wherein the ligand, human. disease or condition is according to any one of aspects 1 io 19. id="p-876" id="p-876"
[00876] In a Twenty-ninth Aspect: A method of targeting a PCSK9 for treating and/or preventing a PCSK9-mediated disease or condition in a human, the method comprising administering an anti-PCSK9 ligand to a human comprising a nucleotide sequence selected from the group consisting SEQ ID NOs: 29-37, whereby a PCSK9 encoded by said nucleotide sequence is targeted. id="p-877" id="p-877"
[00877] The ligand can be any anti-PCSK9 ligand disclosed herein. id="p-878" id="p-878"
[00878] In a Thirtieth Aspect: The method of aspect 29, wherein the method comprises targeting a human PCSK9 selected from the group consisting of forms f c, r, p, mt e, h, aj and q with said ligand to treat and/or prevent said disease or condition in said human. id="p-879" id="p-879"
[00879] In an example, said forms are the mature forms, id="p-880" id="p-880"
[00880] In an example, said forms are the pro-forms. id="p-881" id="p-881"
[00881] In a Thirty-first Aspect: A method of treating and/or preventing a disease or condition mediated by PCSK9 in a human, the method comprising targeting a human PCSK9 selected from the 215 group consisting of forms/ c, r, p, m, e, h, aj and q by administering to the human a ligand that binds said PCSK9 thereby treating and/or preventing said disease or condition in the human. id="p-882" id="p-882"
[00882] In an example, said forms are the mature forms. id="p-883" id="p-883"
[00883] In an example, said forms are the pro-forms. id="p-884" id="p-884"
[00884] The ligand can be any anti-PCSK9 ligand disclosed herein. id="p-885" id="p-885"
[00885] In a Thirty-second Aspect: The method of aspect 3 I, wherein the genome of the human comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37. id="p-886" id="p-886"
[00886] In an example, the nucleotide sequence is selected from the group consisting of SEQ ID NOs: 29-35 and 37; or selected from the group consisting of SEQ ID NOs: 29-32 and 34-37; or selected from the group consisting of SEQ ID NOs: 29-32, 34, 35 and 37. These are naturallyoccurring allele (haplotype) sequences that do not encode 46L and which meet the criteria set out above. These groups comprise variants that are associated with elevated LDL-C. id="p-887" id="p-887"
[00887] In an example, the nucleotide sequence is SEQ ID NO: 34, that encodes a 425S, which is associated with elevated LDL-C (Pisciotta et al 2006). id="p-888" id="p-888"
[00888] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31 and 37, that encode 670G which is a marker for severity of coronary atherosclerosis (Chen et al 2005). |00889] In an example, the nucleotide sequence selected from the group consisting of SEQ ID NOs: 31, 32, 34, 35, 36 and 37; or selected from the group consisting of SEQ ID NOs: 31,32, 34, 35 and 37. These are allele (haplotype) sequences that have a naturally-occurring combination of differences from SEQ ID NO: 28 (form a) and which meet the criteria set out above. id="p-890" id="p-890"
[00890] In an example, the nucleotide sequence is SEQ ID NO: 29. |00891] In an example, the nucleotide sequence is SEQ ID NO: 30. id="p-892" id="p-892"
[00892] In an example, the nucleotide sequence is SEQ ID NO: 3 1. id="p-893" id="p-893"
[00893] In an example, the nucleotide sequence is SEQ ID NO: 32. id="p-894" id="p-894"
[00894] In an example, the nucleotide sequence is SEQ ID NO: 33. id="p-895" id="p-895"
[00895] In an example, the nucleotide sequence is SEQ ID NO: 34. id="p-896" id="p-896"
[00896] In an example, the nucleotide sequence is SEQ ID NO: 35. id="p-897" id="p-897"
[00897] In an example, the nucleotide sequence is SEQ ID NO: 36. id="p-898" id="p-898"
[00898] In an example, the nucleotide sequence is SEQ ID NO: 37. id="p-899" id="p-899"
[00899] In a Thirty-third Aspect: The method of any one of aspects 29 to 32, wherein the human has been or is genotyped as positive for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domain-encoding sequence thereof. id="p-900" id="p-900"
[00900] In a Thirty-fourth Aspect: The method of any one of aspects 29 to 33, wherein the human has been or is phenotyped as positive for a human PCSK9 selected from the group consisting of forms f c, r, p, m, e, h, aj and q. id="p-901" id="p-901"
[00901] In an example, said forms are the mature forms. 216 id="p-902" id="p-902"
[00902] In an example, said forms are the pro-forms. id="p-903" id="p-903"
[00903] In a Thirty-fifth Aspect: The method of any one of aspects 29 to 34, wherein the method comprises genotyping the human as positive for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domain-encoding sequence thereof. [00904] In a Thirty-sixth Aspect: The method of any one of aspects 29 to 35, wherein the method comprises phenotyping the human as positive for a human PCSK9 sequence selected from the group consisting of forms / c, r, p, m, e, A, aj and q, [00905] In an example, said forms are the mature forms. id="p-906" id="p-906"
[00906] In an example, said forms are the pro-forms. id="p-907" id="p-907"
[00907] In a Thirty-seventh Aspect: The method of any one of aspects 29 to 36, wherein the human has been or is genotyped as heterozygous for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domain-encoding sequence thereof; optionally wherein the human has been or is genotyped as comprising the nucleotide sequence of SEQ ID NO: 28 or the catalytic- or C-terminal domain-encoding sequence thereof and a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domain-encoding sequence thereof. id="p-908" id="p-908"
[00908] In a Thirty-eighth Aspect: The method of any one of aspects 29 to 37, wherein the genome of the human has been or is genotyped as homozygous for a nucleotide sequence selected from the group consisting of of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domain-encoding sequence thereof. id="p-909" id="p-909"
[00909] In a Thirty-ninth Aspect: The method of any one of aspects 29 to 38, wherein the method comprises genotyping the human for a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domain-encoding sequence thereof before administering the ligand to the human, wherein the ligand is determined to be capable of binding to a PCSK9 encoded by said selected sequence. id="p-910" id="p-910"
[00910] In a Fortieth Aspect: The method of any one of aspects 29 to 39, wherein the ligand, human, disease or condition is according to any one of aspects 1 to 19. id="p-911" id="p-911"
[00911] In a Forty-first Aspect: A method according to any one of aspects 29 to 40 for treating and/or preventing a condition or disease as recited in aspect 14 or 15, the method comprising administering said ligand and a statin (eg, cerovastatin, atorvastatin, simvastatin, pitavastin, rosuvastatin, fluvastatin, lovastatin or pravastatin) to the human. id="p-912" id="p-912"
[00912] In a Forty-second Aspect: The method of aspect 41, wherein the ligand and statin are administered separately. id="p-913" id="p-913"
[00913] In a Forty-third Aspect: The method of aspect 41, wherein the ligand and statin are administered simultaneously. (00914] In a Forty-fourth Aspect: The method of any one of aspects 29 to 43, wherein the ligand is administered by subcutaneous injection. 217 id="p-915" id="p-915"
[00915] In a Forty-fifth Aspect: A method of PCSK9 genotyping a nucleic acid sample of a human, the method comprising identifying in the sample the presence of a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or the catalytic- or C-terminal domainencoding sequence thereof. id="p-916" id="p-916"
[00916] In a Forty-sixth Aspect: A method of PCSK9 typing a protein sample of a human, the method comprising identifying in the sample the presence of a human PCSK9 selected from the group consisting of forms / c, r, p, m, e, h, aj and q. id="p-917" id="p-917"
[00917] In an example, said forms are the mature forms. id="p-918" id="p-918"
[00918] In an example, said forms are the pro-forms. id="p-919" id="p-919"
[00919] In a Forty-seventh Aspect: The method of aspect 45 or 46, comprising obtaining a sample of serum, blood, faeces, hair, tissue, cells, urine or saliva from a human, whereby the nucleic acid or protein sample is obtained and used in the step of identifying said sequence. id="p-920" id="p-920"
[00920] In a Forty-eighth Aspect: The method of any one of aspects 45 to 47, comprising using a ligand according to any one of aspects 1 to 19 to carry out said identifying step. id="p-921" id="p-921"
[00921] In a Forty-ninth Aspect: A method of treating and/or preventing in a human patient a cardiovascular disease or condition, or a disease or condition that is associated with elevated LDL cholesterol (eg, hypercholesterolaemia), wherein the patient is receiving or has previously received statin treatment for said disease or condition, the method comprising typing the patient using a method of any one of aspects 45 to 48 and administering a ligand according to one of aspects 1 to 19 whereby the human is treated or said disease or condition is prevented; optionally also reducing or stopping statin treatment. id="p-922" id="p-922"
[00922] In an example, said reducing or stopping comprises reducing the dose and/or dosing frequency of statin. id="p-923" id="p-923"
[00923] In a Fiftieth Aspect: A diagnostic, therapeutic or prophylactic kit comprising a ligand that is capable of binding to or has been or is determined as capable of binding to an amino acid sequence selected from SEQ ID NOs: 4-27 and instructions for carrying out the method of any one of aspects 46 to 49 and/or a label or instructions indicating or covering administration of the ligand to a human as defined in any one of aspects 1 to 19. id="p-924" id="p-924"
[00924] In a Fifty-first Aspect: A diagnostic, therapeutic or prophylactic kit comprising a nucleic acid probe comprising a nucleotide sequence that specifically hybridises to a nucleotide sequence selected from the group consisting of SEQ ID NOs: 29-37 or an antisense sequence or RNA transcript thereof and instructions for carrying out the method of aspect 45, 47 or 48. id="p-925" id="p-925"
[00925] In examples of the present invention, the ligand specifically binds to human PCSK9, eg, one or more of the rare PCSK.9 variants disclosed herein (eg, one, two, three, more or all mature forms/ c, r, p, m, e, h, aj and q) and optionally also the a and/or a ’ form. For example, the ligand specifically binds to.mature form f and/or c as well as form a. Determination of such binding can be performed by any antibody binding test as known in the art, eg, by surface plasmon resonance. 218 Binding to each such form is, for example, respectively with aKd of at least ImM, lOOnM, InM, JOOpM, lOpM or JpM. id="p-926" id="p-926"
[00926] In an example, the ligand binds form a and a PCSK9 selected from the group consisting of forms/ c, r, p, m, e, h, aj and q, wherein the ligand binding to said selected form is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-927" id="p-927"
[00927] In an example, the ligand binds form a and form/ wherein the ligand binding to form/is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-928" id="p-928"
[00928] In an example, the ligand binds form a and form c, wherein the ligand binding to form c is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% ofthe Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-929" id="p-929"
[00929] In an example, the ligand binds form a and form r, wherein the ligand binding to form r is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-930" id="p-930"
[00930] In an example, the ligand binds form a and form p, wherein the ligand binding to formp is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-931" id="p-931"
[00931] In an example, the ligand binds form a and form m, wherein the ligand binding to form m is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-932" id="p-932"
[00932] In an example, the ligand binds form a and form e, wherein the ligand binding to form e is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% ofthe Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-933" id="p-933"
[00933] In an example, the ligand binds form a and form A, wherein the ligand binding to form h is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-934" id="p-934"
[00934] In an example, the ligand binds form a and form aj, wherein the ligand binding to form aj is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% ofthe Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-935" id="p-935"
[00935] In an example, the ligand binds form a and form q, wherein the ligand binding to form q is with a Kd (determined by SPR) that is at least 60, 70, 80, 90 or 95% of the Kd for binding to form a. In an embodiment, both forms are mature forms. In an embodiment, both forms are pro-forms. id="p-936" id="p-936"
[00936] In examples of the present invention, the ligand neutralises human PCSK9, eg, one or more of the rare PCSK9 variants disclosed herein (eg, one, two, three, more or all mature forms/ c, r, p, m, e, h, aj and q) and optionally also the a and/or a' form. For example, the ligand neutralises mature form / and/or c as well as form a. Determination of neutralisation can be performed, for 219 example, by any neutralisation assay method disclosed in US20I20093818A1 (Amgen, Inc) or US20110065902A1 (Regeneron Pharmaceuticals, Inc). Ligands of the invention that bind or target PCSK9 are useful, for example, for therapeutic and prophylactic applications disclosed in US20120093818A1 and US20110065902A1, these specific disclosures being incorporated herein by reference for use in the present invention and for possible inclusion in claims herein. id="p-937" id="p-937"
[00937] In embodiments where the ligand is used for therapeutic applications, an antigen binding protein can inhibit, interfere with or modulate one or more biological activities of a PCSK9 (eg, one or more of the rare variants disclosed herein and optionally also the a and/or a ’ form). In one embodiment, ligand binds specifically to human PCSK9 (eg, one or more of the rare variants disclosed herein and optionally also the a and/or a' form) and/or substantially inhibits binding of human PCSK9 (eg, said one or more of the rare variants disclosed herein and optionally also the a and/or a form) to LDLR by at least 20%, eg, 20%-40%, 40-60%, 60-80%, 80-85%, or more (for example, by measuring binding in an in vitro competitive binding assay). In an example, the ligand is an antibody. id="p-938" id="p-938"
[00938] In an embodiment, the ligand has a Kd of less (binding more tightly) than 10"7, 10 s, 10-9, 10-10, 10-11, 10"12, 1013 M for binding to one, two or more of the rare variants disclosed herein and optionally also the a and/or a1 form. In an example, Kd is determined using SPR. id="p-939" id="p-939"
[00939] In an embodiment, the ligand has an IC50 for blocking the binding of LDLR to one or more of the rare PCSK9 variants disclosed herein (and optionally also the a and/or a' form) of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM. id="p-940" id="p-940"
[00940] In an embodiment, the ligand has an IC50 for blocking the binding of LDLR to the a and/or a' form of PCSK9 that is no more than 1000, 100, 90, 80, 70, 60, 50, 40, 30, 20 or 10-fold more (ie, more inhibitory) than the IC50 for blocking the binding of LDLR to one or more of the rare PCSK9 variants disclosed herein (eg, one or more PCSK9 proteins comprising a sequence selected from SEQ ID NOs: 4 to 27). Additionally or alternatively, for example, the ligand has an IC50 for blocking the binding of LDLR to (i) the a and/or a; form of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM, eg, in the range of ImM to IpM (eg, ImM to ΙΟΟρΜ; lOnM to lOOpM; InM to lOpM; or lOOpM to IpM) and (ii) one or more PCSK9 proteins comprising a sequence selected from SEQ ID NOs: 4 to 27 of less than 1 microM, 1000 nM to 100 nM, 100 nM to 10 nM, 10 nM to 1 nM, 1000 pM to 500 pM, 500 pM to 200 pM, less than 200 pM, 200 pM to 150 pM, 200 pM to 100 pM, 100 pM to 10 pM, 10 pM to 1 pM, eg, in the range of ImM to lpM(eg, ImM to lOOpM; lOnMto lOOpM; InM to ΙΟρΜ; or lOOpM to IpM). [00941] In an embodiment, the ligand binds to the a and/or a ’ form of PCSK9 with a binding affinity (Kd) that is greater than up to 10%, greater than up to 20%, greater than up to 40%, greater 220 than up to 50%, greater than up to 55%, greater than up to 60%, greater than up to 65%, greater than up to 70%, greater than up to 75%, greater than up to 80%, greater than up to 85%, greater than up to 90%, greater than up to 95% or greater than up to 100% (ie, is double) relative to binding to a PCSK9 comprising a sequence selected from SEQ ID NOs: 4 to 27. Such binding measurements can be made using a variety of binding assays known in the art, eg, using surface plasmon resonance (SPR), such as by Biacore™ or using the ProteOn XPR36™ (Bio-Rad®), or using KinExA® (Sapidyne Instruments, Inc). id="p-942" id="p-942"
[00942] In one embodiment, the surface plasmon resonance (SPR) is carried out at 25°C. In another embodiment, the SPR is carried out at 37°C. id="p-943" id="p-943"
[00943] In one embodiment, 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)). id="p-944" id="p-944"
[00944] In one embodiment, the SPR is carried out at a physiological salt level, eg, 150mM NaCl. id="p-945" id="p-945"
[00945] In one embodiment, 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. id="p-946" id="p-946"
[00946] In one example, 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. In one example, the SPR is carried out at 25QC or 37°C in HBS-EP. HBS-EP is available from Teknova Inc (California; catalogue number H8022). id="p-947" id="p-947"
[00947] In an example, the affinity of the ligand which is an antibody is determined using SPR by 1. Coupling anti-mouse (or other relevant vertebrate) IgG (eg, Biacore BR-1008-38) to a biosensor chip (eg, GLM chip) such as by primary amine coupling; 2. Exposing the anti-mouse IgG (vertebrate antibody) to a test IgG antibody to capture test antibody on the chip; 3. Passing the test antigen over the chip’s capture surface at 1024nM, 256nM, 64nM, !6nM, 4nM with a OnM (i.e. buffer alone); and 4. And determining the affinity of binding of test antibody to test antigen using surface plasmon resonance, eg, under an SPR condition discussed above (eg, at 25°C in physiological buffer). SPR can be carried out using any standard SPR apparatus, such as by BiacoreTM or using the ProteOn XPR36TM (Bio-Rad®). id="p-948" id="p-948"
[00948] Regeneration of the capture surface can be carried out with lOmM glycine at pHl .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 XPR36™ analysis software. id="p-949" id="p-949"
[00949] In an embodiment, assaying or testing of a ligand of the invention is carried out at or substantially at pH7 (eg, for in vitro tests and assays/ and at or substantially at rtp. 221 id="p-950" id="p-950"
[00950] One example of an IgG2 heavy chain constant domain of an anti-PCSK9 antibody of the present invention has the amino acid sequence as shown in SEQ ID NO: 154, FIG. 3KK of US20120093818AI, which sequence is incorporated herein by reference. 100951] One example of an lgG4 heavy chain constant domain of an anti-PCSK9 antibody of the 5 present invention has the amino acid sequence as shown in SEQ ID NO: 155, FIG. 3KK of US20120093818A1, which sequence is incorporated herein by reference. id="p-952" id="p-952"
[00952] One example of a kappa light chain constant domain of an anti-PCSK9 antibody has the amino acid sequence as shown in SEQ ID NO: 157, FIG. 3KK which sequence is incorporated herein by reference. id="p-953" id="p-953"
[00953] One example of a lambda light chain constant domain of an anti-PCSK9 antibody has the amino acid sequence as shown in SEQ ID NO: 156, FIG. 3KK of US20120093818A1, which sequence is incorporated herein by reference. id="p-954" id="p-954"
[00954] In examples of the present invention, the ligand binds mature PCSK9, eg, a mature form of one or more of the rare variants disclosed herein and optionally also the a and/or a3 form. |00955] In examples of the present invention, the ligand binds the catalytic domain of PCSK9, eg, of a mature form of one or more of the rare variants disclosed herein and optionally also the a and/or a 3 form. id="p-956" id="p-956"
[00956] In examples of the present invention, the ligand binds the prodomain of PCSK9, eg, of a mature form of one or more of the rare variants disclosed herein and optionally also the a and/or a ’ 20 form. id="p-957" id="p-957"
[00957] In some embodiments, the ligand binds to the V domain of PCSK9, eg, of a mature form of one or more of the rare variants disclosed herein and optionally also the a and/or a3 form. In some embodiments, the ligand binds to the V domain of PCSK9 (eg, of a mature form of one or more of the rare variants disclosed herein and optionally also the a and/or a3 form) and prevents (or reduces, eg, 25 by at least 10%) PCSK9 from binding to LDLR. In some embodiments, the ligand binds to the V domain of PCSK9 (eg, of a mature form of one or more of the rare variants disclosed herein and optionally also the a and/or a3 form), and while it does not prevent (or reduce) the binding of PCSK9 to LDLR, the ligand prevents or reduces (eg, by at least 10%) the adverse activities mediated through PCSK9 on LDLR. id="p-958" id="p-958"
[00958] In examples of the present invention, the ligand is or comprises a fully human antibody.
In an example, the ligand comprises human variable regions or humanised variable regions. id="p-959" id="p-959"
[00959] In an example, the ligand of the invention specifically binds to an epitope of a human PCSK9 selected from the group consisting of forms/ c, r, p, m, e, h, aj and q, wherein the epitope comprises at least one amino acid that is not found in form a. For example, the amino acid is selected 35 from the group consisting of 46L, 53V, 425S, 443T, 474V, 619P and 670G (numbering as used in SEQ ID NO: 1). For example, the amino acid is selected from the group consisting of 425S, 443T, 474V, 619P and 670G (numbering as used in SEQ ID NO:1). For example, the amino acid is selected 222 from the group consisting of 425S and 443T (numbering as used in SEQ ID NO: 1). For example, the amino acid is selected from the group consisting of 474V, 619P and 670G (numbering as used in SEQ ID NO: 1). In an example, the PCSK9 form is the mature form. In an example, the PCSK9 form is the pro-form. In an example, the ligand also specifically binds to form a and/or a'. In an embodiment, the ligand specifically binds to an epitope of form/PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form c PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form r PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form p PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form m PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form e PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form h PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form aj PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to an epitope of form q PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. id="p-960" id="p-960"
[00960] In an embodiment, ligand binds specifically to the pro-domain of a human PCSK9 selected from the group consisting of forms/ ct r, p, m, e, h, aj and q, In an example, the ligand also specifically binds to the pro-domain of form a and/or a*. In an embodiment, the ligand specifically binds to the pro-domain of form /PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form c PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form r PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form p PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the prodomain of form m PCSK93 wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form e PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form h PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form aj PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the pro-domain of form q PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. 223 id="p-961" id="p-961"
[00961] In an embodiment, ligand binds specifically to the catalytic domain of a human PCSK9 selected from the group consisting of forms/ c> r, p, m, e, h, aj and q. In an example, the ligand also specifically binds to the catalytic domain of form a and/or af. In an embodiment, the ligand specifically binds to the catalytic domain of form/PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form c PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form r PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form p PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form m PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form e PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form h PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form qj PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the catalytic domain of form q PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. id="p-962" id="p-962"
[00962] In an embodiment, ligand binds specifically to the C-terminal domain of a human PCSK9 selected from the group consisting of forms/ c, r, p, m, e, h, aj and q. In an example, the ligand also specifically binds to the C-terminal domain of form a and/or a'. In an embodiment, the ligand specifically binds to the C-terminal domain of form/PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Cterminal domain of form c PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the C-terminal domain of form r PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the C-terminal domain of form p PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the C-terminal domain of form m PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Cterminal domain of form e PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the C-terminal domain of form h PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the C-terminal domain of form aj PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand 224 specifically binds to the C-terminal domain of form q PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. id="p-963" id="p-963"
[00963] In an embodiment, ligand binds specifically to the substrate-binding groove of a human PCSK9 selected from the group consisting of forms/ c, r, pt m, e, h, aj and q (see Cunningham et aL, Nat Struct Mol Biol. 2007 May; 14(5):413-9. Epub 2007 Apr 15, "Structural and biophysical studies of PCSK9 and its mutants linked to familial hypercholesterolemia", incorporated herein in its entirety by reference). In an example, the ligand also specifically binds to the substrate-binding groove of form a and/or a1. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form/PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form c PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form r PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form p PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form m PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substratebinding groove of form e PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form h PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form aj PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. In an embodiment, the ligand specifically binds to the Substrate-binding groove of form q PCSK9, wherein the epitope comprises at least one amino acid that is not found in form a. id="p-964" id="p-964"
[00964] Reference is made to US20120093818A1 (Amgen, Inc), the entire disclosure of which is incorporated herein. This patent application discloses relevant ligands for use in the present invention, as well as examples and methods of producing and testing ligands that can be used with reference to the present invention. id="p-965" id="p-965"
[00965] In an example, the ligand is or comprises an antibody disclosed in Table 2 of US20120093818A1 (Amgen, Inc) or is a PCSK9-binding derivative thereof. id="p-966" id="p-966"
[00966] In an embodiment, the PCSK9-binding ligand ofthe invention is selected from the antigen binding proteins disclosed in US20120093818A1 (Amgen, Inc), eg, in paragraphs [0009] to [0014] and [0058] to [0063] of US20120093818A1; all ofthese disclosures (including the sequences of such proteins) are incorporated herein by reference as though explicitly recited herein and for possible inclusion in one or more claims or for use in the present invention. id="p-967" id="p-967"
[00967] In this paragraph SEQ ID NOs are those as appearing in US20120093818A1 (Amgen, Inc) and these sequences are incorporated herein by reference as though explicitly recited herein and 225 for possible inclusion in one or more claims or for use in the present invention. In some aspects, the ligand of the invention comprises an isolated antigen binding protein that binds PCSK9 comprising: A) one or more heavy chain complementary determining regions (CDRHs) selected from the group consisting of: (i) a CDRH1 from a CDRH1 in a sequence selected from the group consisting of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, and 60; (ii) a CDRH2 from a CDRH2 in a sequence selected from the group consisting of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, and 60; (iii) a CDRH3 from a CDRH3 in a sequence selected from the group consisting of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91, 64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, and 60; and (iv) a CDRH of (i), (ii), and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 4 amino acids: B) one or more light chain complementaiy determining regions (CDRLs) selected from the group consisting of: (i) a CDRL1 from a CDRL1 in a sequence selected from the group consisting of SEQ ID NO: 5,7,9, 10, 12, 13, 15, 16, 17, 18, 19, 20,21,22,23,24, 26,28, 30,31,32,33,35,36,37,38, 39, 40, 42, 44, and 46; (ii) a CDRL2 from a CDRL2 in a sequence selected from the group consisting of SEQ ID NO: 5,7,9, 10, 12, 13, 15, 16, 17, 18, 19, 20,21,22,23, 24, 26,28,30,31,32,33,35,36, 37, 38, 39, 40,42, 44, and 46; (iii) a CDRL3 from a CDRL3 in a sequence selected from the group consisting of SEQ ID NO: 5,7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20,21,22, 23,24, 26, 28,30,31,32, 33, 35, 36, 37, 38, 39, 40, 42, 44, and 46; and (iv) a CDRL of (i), (ii) and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 4 amino acids; or C) one or more heavy chain CDRHs of A) and one or more light chain CDRLs of B). In some embodiments, the isolated antigen binding protein comprises at least one CDRH of A) and at least one CDRL of B). In some embodiments, the isolated antigen binding protein comprises at least two CDRH of A) and at least two CDRL of B). In some embodiments, the isolated antigen binding protein comprises said CDRH1, CDRH2, CDRH3, CDRL1, CDRL2 and CDRL3. In some embodiments, the CDRH of A) is selected from at least one of the group consisting of: (i) a CDRH1 amino acid sequence selected from the CDRH1 in a sequence selected from the group consisting of SEQ ID NO: 67. 79, 89, and 49; (ii) a CDRH2 amino acid sequence selected from the CDRH2 in a sequence selected from the group consisting of SEQ ID NO: 67, 79, 89, and 49; (iii) a CDRH3 amino acid sequence selected from the CDRH3 in a sequence selected from the group consisting of SEQ ID NO: 67, 79, 89, and 49; and (iv) a CDRH of (i), (ii) and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 2 amino acids. In addition, the CDRL of B) is selected from at least one of the group consisting of: (i) a CDRL1 amino acid sequence selected from the CDRLl in a sequence selected from the group consisting of SEQ ID NO: 12, 35, 32, and 23; (ii) a CDRL2 amino acid sequence selected from the CDRL2 in a sequence selected from the group consisting of SEQ ID NO: 12, 35, 32, and 23; (iii) a CDRL3 amino acid sequence selected from the CDRL3 in a sequence selected from the group consisting of SEQ ID NO: 12, 35, 32, and 23; and (iv) a CDRL of (i), (ii) and (iii) that contains 226 one or more amino acid substitutions, deletions or insertions of no more than 2 amino acids; or C) one or more heavy chain CDRHs of A) and one or more light chain CDRLs of B. In some embodiments, the CDRH of A) is selected from at least one of the group consisting of: (i) a CDRH1 amino acid sequence of the CDRH1 amino acid sequence in SEQ ID NO: 67; (ii) a CDRH2 amino acid sequence of the CDRH2 amino acid sequence in SEQ ID NO: 67: (iii) a CDRH3 amino acid sequence of the CDRH3 amino acid sequence in SEQ ID NO: 67; and (iv) a CDRH of (i), (ii) and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 2 amino acids; said CDRL of B) is selected from at least one of the group consisting of: (i) a CDRL1 amino acid sequence of the CDRL I amino acid sequence in SEQ ID NO: 12; (ii) a CDRL2 amino acid sequence of the CDRL2 amino acid sequence in SEQ ID NO: 12; (iii) a CDRL3 amino acid sequence of the CDRL3 amino acid sequence in SEQ ID NO: 12; and (iv) a CDRL of (i), (ii) and (iii) that contains one or more amino acid substitutions, deletions or insertions of no more than 2 amino acids; or C) one or more heavy chain CDRHs of A) and one or more light chain CDRLs of B). In some embodiments, the antigen binding protein comprises A) a CDRH1 of the CDRH1 sequence in SEQ ID NO: 67, a CDRH2 of the CDRH2 sequence in SEQ ID NO: 67, and a CDRH3 of the CDRH3 sequence in SEQ ID NO: 67, and B) a CDRL1 of the CDRL1 sequence in SEQ ID NO: 12, a CDRL2 of the CDRL2 sequence in SEQ ID NO: 12, and a CDRL3 of the CDRL3 sequence in SEQ ID NO: 12. In some embodiments, the antigen binding protein comprises a heavy chain variable region (VH) having at least 80% sequence identity with an amino acid sequence selected from the group consisting of SEQ ID NO: 74, 85, 71, 72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91,64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, and 60, and/or a light chain variable region (VL) having at least 80% sequence identity with an amino acid sequence selected from the group consisting of SEQ ID NO: 5, 7, 9. 10, 12, 13, 15, 16, 17, 18, 19, 20,21, 22, 23,24, 26, 28, 30, 31, 32,33, 35,36, 37, 38,39,40,42, 44, and 46. In some embodiments, the VH has at least 90% sequence identity with an amino acid sequence selected from the group consisting of SEQ ID NO: 74, 85, 71,72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91,64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, and 60, and/or the VL has at least 90% sequence identity with an amino acid sequence selected from the group consisting of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20,21, 22,23,24, 26,28,30,31, 32, 33,35,36,37,38, 39,40,42, 44, and 46. In some embodiments, the VH is selected from the group consisting of SEQ ID NO: 74, 85, 71,72, 67, 87, 58, 52, 51, 53, 48, 54, 55, 56, 49, 57, 50, 91,64, 62, 89, 65, 79, 80, 76, 77, 78, 83, 69, 81, and 60, and/or the VL is selected from the group consisting of SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19, 20, 21,22, 23, 24, 26, 28, 30, 31, 32, 33, 35, 36, 37, 38, 39, 40,42, 44, and 46. [00968] In an example of any aspect of the invention, the PCSK9-targeting or binding ligand comprises or consists of AMG145 or 31H4, 16F12, 11F1, 8A3 or 21B12 disclosed in US20120093818A1 (Amgen, Inc) or an antibody comprising the variable domains of AMG 145, 31H4, 16F12, 11F1, 8 A3 or 21B12, the disclosures of which (including sequences) are incorporated herein by reference as though explicitly recited herein and for possible inclusion in one or more 227 claims or for use in the present invention. Preferably, the PCSK9-targeting or binding ligand comprises or consists of AMG145. id="p-969" id="p-969"
[00969] In an example, the AMG 145 or other ligand of the invention is glycosylated, eg, has human glycosylation (eg, produced by a CHO, Cos or Hek293 cell). In an example, the ligand of the invention is produced in CHO. id="p-970" id="p-970"
[00970] Reference is made to US20I10065902AI (Regeneron Pharmaceuticals, Inc), the entire disclosure of which is incorporated herein. This patent application discloses relevant ligands for use in the present invention, as well as examples and methods of producing and testing ligands and determining medical efficacy that can be used with reference to the present invention. id="p-971" id="p-971"
[00971] Reference is made to the following PCT applications, the entire disclosures of which are incorporated herein. These disclose relevant ligands for use in the present invention, as well as examples and methods of producing and testing ligands and determining medical efficacy that can be used with reference to the present invention.
WO2008057457 WO2008057458 WO2008057459 WO2008063382 WO2008133647 WO2009100297 WO2009100318 WO2011037791 WO2011053759 WO2011053783 WO2008125623 WO2011072263 WO2009055783 WO2010029513 WO20I1111007 WO2010077854 id="p-972" id="p-972"
[00972] Antibody ligands to PCSK9 are described in, for example, WO 2008/057457, WO 2008/057458, WO 2008/057459, WO 2008/063382, WO 2008/125623, and US 2008/0008697: each of which is incorporated by reference herein in its entirety.. id="p-973" id="p-973"
[00973] In an example, the ligand is or comprises an antibody disclosed in the Examples of US20110065902A1 (eg, 316P or 300N) or is a PCSK9-binding derivative thereof. All of these disclosures (including the sequences of such proteins and corresponding nucleotide sequences) are incorporated herein by reference as though explicitly recited herein and for possible inclusion in one or more claims or for use in the present invention. In an embodiment, the ligand is or comprises the 228 variable domains of antibody 316P or 300N disclosed in US20110065902Al or is (or comprises) such antibody or a PCSK9-binding derivative thereof. id="p-974" id="p-974"
[00974] In an embodiment, the ligand is or comprises the variable domains of antibody alirocumab or SAR236553/REGN727 (Sanofi Aventis/Regeneron) or is (or comprises) such antibody or a PCSK9-binding derivative thereof. In an example, the alirocumab is glycosylated, eg, has human glycosylation (eg, produced by a CHO, Cos or Hek293 cell). Preferably, the ligand is alirocumab or SAR236553/REGN727. id="p-975" id="p-975"
[00975] In an embodiment, the ligand is or comprises the variable domains of antibody evolocumab or or is (or comprises) such antibody or a PCSK9-binding derivative thereof. In an example, the antibody is glycosylated, eg, has human glycosylation (eg, produced by a CHO, Cos or Hek293 cell). Preferably, the ligand is evolocumab. id="p-976" id="p-976"
[00976] In an embodiment, the ligand is selected from evolocumab, lD05-IgG2 (Merck & Co.), ALN-PCS02 (Alnylam), RN316 (Pfizer-Rinat) and alirocumab. [00977) In an embodiment, the ligand is selected from the following (sequences and definitions as per US2011/0065902, incorporated herein by reference):- 1. An antibody or antigen-binding fragment thereof which specifically binds hPCSK.9, wherein the antibody or antigen-binding fragment comprises the heavy and light chain CDRs of a HCVR/LCVR amino acid sequence pair having SEQ ID NOs: 218/226. 2. The antibody or antigen-binding fragment of concept 1 comprising heavy and light chain CDR amino acid sequences having SEQ ID NOs: 220, 222, 224, 228, 230 and 232. 3. The antibody or antigen-binding fragment of concept 2 comprising an HCVR having the amino acid sequence of SEQ ID NO: 218 and an LCVR having the amino acid sequence of SEQ ID NO: 226. 4. An antibody or antigen-binding fragment thereof which binds to the same epitope on hPCSK9 as an antibody comprising heavy and light chain CDR amino acid sequences having SEQ ID NOs: 220, 222, 224, 228, 230 and 232.
. An antibody or antigen-binding fragment thereof which competes for binding to hPCSK9 with an antibody comprising heavy and light chain CDR amino acid sequences having SEQ ID NOs: 220, 222, 224, 228, 230 and 232. id="p-978" id="p-978"
[00978] In an embodiment, the ligand is selected from the following (sequences and definitions as per US2012/0093818, incorporated herein by reference):- 1. An isolated neutralizing antigen binding protein that binds to a PCSK9 protein comprising the amino acid sequence of SEQ ID NO: 1, wherein the neutralizing antigen binding protein decreases the LDLR lowering effect of PCSK9 on LDLR, wherein the antigen binding protein comprises a light chain comprising an amino acid sequence of SEQ ID NO: 46, and wherein the antigen binding protein comprises a heavy chain comprising an amino acid sequence of SEQ ID NO: 60. 2. The isolated neutralizing antigen binding protein of concept 2, wherein the antigen binding protein 229 is a LDLR non-competitive neutralizing antigen binding protein. 3. The isolated neutralizing antigen binding protein of concept 2, wherein the antigen binding protein is a LDLR competitive neutralizing antigen binding protein. 4. An antigen binding protein that selectively binds to PCSK9, wherein said antigen binding protein binds to PCSK9 with a Kd that is less than 100 pM.
. An antigen binding protein that binds to a PCSK 9 protein of SEQ ID NO: 303 in a first manner, wherein the antigen binding protein binds to a variant of PCSK9 in a second manner, wherein said PCSK9 variant has at least one point mutation at a position selected from the group consisting of: 207, 208, 185, 181,439,513,538,539, 132, 351,390,413,582, 162, 164, 167, 123, 129,311,313,337, 519, 521, and 554 of SEQ ID NO: 303, wherein the first manner comprises a first EC50, a first Bmax, or a first EC50 and a first Bmax, wherein the second manner comprises a second EC50, a second Bmax, or a second EC50 and a second Bmax, and wherein a value for the first manner is different from a value for the second manner, and wherein the antigen binding protein comprises a light chain comprising an amino acid sequence of SEQ ID NO: 46, and wherein the antigen binding protein comprises a heavy chain comprising an amino acid sequence of SEQ ID NO: 60. 6. The antigen binding protein of concept 6, wherein the first manner comprises a first Bmax, wherein the second manner comprises a second Bmax that is different from the first Bmax, and wherein said PCSK9 variant has at least one point mutation selected from the group consisting of: D162R, R164E, E167R, S123R, E129R, A3 UR, D313R, D337R, R519E, H521R, and Q554R. 7. The antigen binding protein of concept 6, wherein the antigen binding protein binds to PCSK9 at a location that overlaps with a location that LDLR binds to PCSK9. 8. A method of making an antigen binding protein that binds to a PCSK9 protein comprising the amino acid sequence of SEQ ID NO: 1. wherein the antigen binding protein decreases the LDLR lowering effect of PCSK9 on LDLR, said method comprising:providing a host cell comprising a nucleic acid sequence that encodes the antigen binding protein; andmaintaining the host cell under conditions in which the antigen binding protein is expressed, wherein the antigen binding protein comprises a light chain comprising an amino acid sequence of SEQ ID NO: 46, and wherein the antigen binding protein comprises a heavy chain comprising an amino acid sequence of SEQ ID NO: 60. 9. A method for treating or preventing a condition associated with elevated serum cholesterol levels in a subject, said method comprising administering to a subject in need thereof an effective amount of an isolated neutralizing antigen binding protein simultaneously or sequentially with an agent that elevates the availability of LDLR protein, wherein the isolated antigen binding protein binds to a PCSK9 protein comprising the amino acid sequence of SEQ ID NO: 1, wherein the neutralizing antigen binding protein decreases the LDLR lowering effect of PCSK9 on LDLR, wherein the antigen binding protein comprises a light chain comprising an amino acid sequence of SEQ ID NO: 46, and wherein the antigen binding protein comprises a heavy chain comprising an amino acid sequence of 230 SEQ ID NO: 60.
. The method of concept 10, wherein the agent that elevates the availability of LDLR protein comprises a statin. 11. An antigen binding protein that binds to PCSK9, wherein when the antigen binding protein is bound to PCSK9, the antibody is positioned 8 angstroms or less from at least one of the following residues of PCSK9: S153, S188,1189, Q190, SI91, D192, RI94, E197, G198, R199, V20O, D224, R237, D238, K243, S373, D374, S376, T377, F379,1154, T187, H193, E195,1196, M20L V202, C223, T228, S235, G236, A239, G244, M247,1369, S372, C375, or C378, wherein the antigen binding protein comprises a light chain comprising an amino acid sequence of SEQ ID NO: 46, and wherein the antigen binding protein comprises a heavy chain comprising an amino acid sequence of SEQ ID NO: 60. id="p-979" id="p-979"
[00979] The ligand can be used for the treatment, therapy, prophylaxis and/or diagnosis of one or more diseases or conditions or susceptibility thereto, wherein such diseases or conditions comprise those disclosed in US20120093818A1 (Amgen, Inc) and US20110065902A1 (Regeneron Pharmaceuticals, Inc), eg, a disease or condition disclosed in paragraphs [0375] to [0383] of US201200938I8A1, which disclosure is incorporated herein by reference in its entirety for inclusion in one more claims herein. id="p-980" id="p-980"
[00980] The ligand can be administered to a human characterised as described in US20120093818AI (Amgen, Inc) or US20110065902A1. id="p-981" id="p-981"
[00981] The ligand can be administered in a form or combination disclosed in US20120093818A1 (Amgen, Inc) or US20110065902A1, which disclosure is incorporated herein by reference. For example, the ligand with a drug, excipient, diluent or carrier as described in US20120093 818A1 (Amgen, Inc) or US20110065902A1 (eg, as disclose in paragraphs [0384] to [0412] of US20120093818A1), which disclosure is incorporated herein by reference, and the present invention also relates to the corresponding pharmaceutical compositions comprising the combination of a ligand of the invention and such a further agent. . id="p-982" id="p-982"
[00982] The ligand can be used in a method of diagnosis as set out in US20120093 818A1 (Amgen, Inc) or US20110065902A1, eg, in paragraphs [0413] to [0415] of US2012009381 8A1 which disclosure is incorporated herein by reference. id="p-983" id="p-983"
[00983] Diagnostic Applications id="p-984" id="p-984"
[00984] In some embodiments, the ligand of the invention is a diagnostic tool. The ligand can be used to assay the amount of PCSK9 present in a sample and/or subject. As will be appreciated by one of skill in the art, such ligands need not be neutralizing ligands. In some embodiments, the diagnostic ligand is not a neutralizing ligand. In some embodiments, the diagnostic ligand binds to a different epitope than a neutralizing ligand binds to. In some embodiments, the two ligands do not compete with one another. 231 id="p-985" id="p-985"
[00985] In some embodiments, the ligands of the invention are used or provided in an assay kit and/or method for the detection of PCSK9 in mammalian tissues or cells in order to screen/diagnose for a disease or disorder associated with changes in levels of PCSK9. The kit comprises a ligand that binds PCSK9 and means for indicating the binding of the ligand with PCSK9, if present, and optionally PCSK9 protein levels. Various means for indicating the presence of a ligand can be used. For example, fluorophores, other molecular probes, or enzymes can be linked to the ligand and the presence of the ligand can be observed in a variety of ways. The method for screening for such disorders can involve the use of the kit, or simply the use of one of the disclosed ligands and the determination of whether the ligand binds to PCSK9 in a sample. As will be appreciated by one of skill in the art, high or elevated levels of PCSK9 will result in larger amounts of the ligand binding to PCSK9 in the sample. Thus, degree of ligand binding can be used to determine how much PCSK9 is in a sample. Subjects or samples with an amount of PCSK9 that is greater than a predetermined amount (e.g., an amount or range that a person without a PCSK9 related disorder would have) can be characterized as having a PCSK9 mediated disorder. In some embodiments, the invention provides a method wherein the ligand is administered to a subject taking a statin, in order to determine if the statin has increased the amount of PCSK9 in the subject. id="p-986" id="p-986"
[00986] In some embodiments, the ligand is a non-neutralizing ligand and is used to determine the amount of PCSK9 in a subject receiving an ABP and/or statin treatment. id="p-987" id="p-987"
[00987] In some embodiments, the ligand of the invention can specifically bind human PCSK9 (eg, one, two or more rare variant forms disclosed herein) and is characterized by at least one of: (i) capable of reducing serum total cholesterol at least about 25-35% and sustaining the reduction over at least a 24 day period relative to a predose level; (ii) capable of reducing serum LDL cholesterol at least about 65-80% and sustaining the reduction over at least a 24 day period relative to a predose level; (iii) capable of reducing serum LDL cholesterol at least about 40-70% and sustaining the reduction over at least a 60 or 90 day period relative to a predose level; (iv) capable of reducing serum triglyceride at least about 25-40% relative to predose level; (v) does not reduce serum HDL cholesterol or reduces serum HDL cholesterol no more than 5% relative to predose level. In some embodiments, an isolated nucleic acid molecule is provided and it encodes the ligand. In some embodiments an expression vector is provided and comprises the nucleic acid molecule. In some embodiments, a pharmaceutical composition is provided and it can comprise the ligand and a pharmaceutically acceptable carrier. In some embodiments, a method is provided for treating a disease or condition which is ameliorated, improved, inhibited or prevented with a PCSK9 antagonist ligand of the invention. The method can comprise administering a therapeutic amount of the pharmaceutical composition or ligand to a subject in need thereof. In some embodiments, the subject is a human subject suffering from hypercholesterolemia, hyperlipidemia, indicated for LDL apheresis, identified as heterozygous for Familial Hypercholesterolemia, statin intolerant, statin uncontrolled, at risk for developing hypercholesterolemia, dyslipidemia, cholestatic liver disease, nephrotic syndrome, 232 hypothyroidism, obesity, atherosclerosis and cardiovascular diseases. In some embodiments, a method of providing a treatment or therapy is provided to a subject. In some embodiments, the method comprises reducing serum cholesterol at least about 40-70% over at least 60 to 90 days. In some embodiments, a method of receiving treatment or therapy is provided, the method can comprise receiving a ligand thereof at a frequency of once every 60 to 90 days. id="p-988" id="p-988"
[00988] In one aspect, the invention provides a ligand of the invention which is or comprises an human antibody or antigen-binding fragment of a human antibody that specifically binds and inhibits human proprotein convertase subtilisin/kexin type 9 (hPCSK9, eg, one, two or more rare variant forms disclosed herein and optionally form a and/or form a’), characterized by the ability to reduce serum LDL cholesterol in a human by 40-80% over a 24, 60 or 90 day period relative to predose levels, with little or no reduction in serum HDL cholesterol and/or with little or no measurable effect on liver function, as determined by ALT and AST measurements. id="p-989" id="p-989"
[00989] In one embodiment, the ligand of the invention comprises an antibody or antigen-binding fragment of an antibody that specifically binds hPCSK9 and is characterized by at least one of: (i) capable of reducing serum total cholesterol at least about 25-35% and sustaining the reduction over at least a 24 day period relative to a predose level, preferably the reduction in serum total cholesterol is at least about 30-40%; (ii) capable of reducing serum LDL cholesterol at least about 65-80% and sustaining the reduction over at least a 24 day period relative to a predose level; (iii) capable of reducing serum triglyceride at least about 25-40% relative to predose level; (iv) does not reduce serum HDL cholesterol or reduces serum HDL cholesterol no more than 5% relative to predose level. id="p-990" id="p-990"
[00990] See US2011/0065902 for definitions of these terms and optional features, the disclosure of which is incorporated herein by reference in its entirety. id="p-991" id="p-991"
[00991] In one embodiment, the invention comprises an antibody or antigen-binding fragment of an antibody that specifically binds hPCSK9 and is characterized by at least one of: (i) capable of reducing serum LDL cholesterol at least about 40-70% and sustaining the reduction over at least a 60 or 90 day period relative to a predose level; (ii) capable of reducing serum triglyceride at least about 25-40% relative to predose level; (iii) does not reduce serum HDL cholesterol or reduces serum HDL cholesterol no more than 5% relative to predose level. id="p-992" id="p-992"
[00992] In one embodiment, the antibody or antigen-binding fragment is characterized as exhibiting an enhanced binding affinity (KD) for hPCSK9 at pH 5.5 relative io the KD at pH 7.4, as measured by plasmon surface resonance. In a specific embodiment, the antibody or fragment thereof exhibits at least a 20-fold, at least a 40-fold or at least a 50-fold enhanced affinity for PCSK9 at an acidic pH relative to a neutral pH, as measured by surface plasmon resonance. 233 id="p-993" id="p-993"
[00993] In one embodiment, the antibody or antigen-binding fragment is characterized as not exhibiting an enhanced binding affinity for PCSK9 at an acidic pH relative to a neutral pH, as measured by surface plasmon resonance. In a specific embodiment, the antibody or fragment thereof exhibits a decreased binding affinity at an acidic pH. id="p-994" id="p-994"
[00994] In another embodiment, the antibody or antigen-binding fragment binds human, human GOF mutation D374Y, cynomolgus monkey, rhesus monkey, mouse, rat and hamster PCSK9. ]00995] In one embodiment, the antibody or antigen-binding fragment binds human and monkey PCSK9, but does not bind mouse, rat or hamster PCSK9. id="p-996" id="p-996"
[00996] In one embodiment, the invention comprises an antibody or antigen-binding fragment of an antibody comprising one or more of a heavy chain variable region (HCVR), light chain variable region (LCVR), HCDRL HCDR2, HCDR3 disclosed in any of paragraphs [023]-[037] of US2011/0065902, the disclosures of which are incorporated herein by reference. id="p-997" id="p-997"
[00997] In a related embodiment, the invention comprises an antibody or antigen-binding fragment of an antibody which specifically binds hPCSK9, wherein the antibody or fragment comprises heavy and light chain CDR domains contained within heavy and light chain sequence pairs selected from the group consisting of SEQ ID NO (using the sequence numbering in US2011/0065902): 2/10, 18/20, 22/24, 26/34, 42/44, 46/48, 50/58, 66/68, 70/72, 74/82, 90/92, 94/96, 98/106, 114/116, 118/120, 122/130, 138/140, 142/144, 146/154, 162/164, 166/168, 170/178, 186/188, 190/192, 194/202, 210/212, 214/216, 218/226, 234/236, 238/240, 242/250, 258/260, 262/264, 266/274, 282/284, 286/288, 290/298, 306/308, 3 10/312, 314/322, 330/332, 334/336, 338/346, 354/356, 358/360, 362/370, 378/380, 382/384, 386/394, 402/404, 406/408, 410/418, 426/428, 430/432, 434/442, 450/452, 454/456, 458/466, 474/476, 478/480, 482/490, 498/500, 502/504, 506/514, 522/524, 526/528, 530/538, 546/548, 550/552, 554/562, 570/572, 574/576, 578/586, 594/596, 598/600, 602/610, 618/620, 622/624, 626/634, 642/644, 646/648, 650/658, 666/668, 670/672, 674/682, 690/692, 694/696, 698/706, 714/716, 718/720, 722/730, 738/740 and 742/744. In one embodiment, the CDR sequences are contained within HCVR and LCVR selected from the amino acid sequence pairs of SEQ ID NO: 50/58, 66/68, 70/72, 74/82, 90/92, 94/96, 122/130, 138/140, 142/144, 218/226, 234/236, 238/240, 242/250, 258/260, 262/264, 314/322, 330/332 and 334/336. In more specific embodiments, the CDR sequences are comprised within HCVR/LCVR sequences selected from SEQ ID NO: 90/92 or 218/226. id="p-998" id="p-998"
[00998] In an example, the invention features a pharmaceutical composition comprising a ligand of the invention, wherein the ligand comprises or consists of a recombinant human antibody or fragment thereof which specifically binds hPCSK9 and a pharmaceutically acceptable carrier. In one embodiment, the invention features a composition which is a combination of a ligand of the invention (eg, an antibody or antigen-binding fragment of an antibody), and a second therapeutic agent. The second therapeutic agent may be any agent that is advantageously combined with the ligand of the invention, for example, an agent capable of inducing a cellular depletion of cholesterol synthesis by 234 inhibiting 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase, such as, for example, cerovastatin, atorvastatin, simvastatin, pitavastin, rosuvastatin, fluvastatin, lovastatin, pravastatin, etc; capable of inhibiting cholesterol uptake and or bile acid re-absorption; capable of increasing lipoprotein catabolism (such as niacin); and/or activators of the LXR transcription factor that plays a role in cholesterol elimination such as 22-hydroxycholesterol. id="p-999" id="p-999"
[00999] In an example, the invention provides a method for inhibiting hPCSK9 activity using the anti-PCSK9 ligand of the invention (eg, an antibody or antigen-binding portion of the antibody of the invention), wherein the therapeutic methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an antibody or antigen-binding fragment of an antibody of the invention. The disorder treated is any disease or condition which is improved, ameliorated, inhibited or prevented by removal, inhibition or reduction of PCSK9 activity. Specific populations treatable by the therapeutic methods of the invention include subjects indicated for LDL apheresis, subjects with PCSK9-activating mutations (gain of function mutations, "GOF"), subjects with heterozygous Familial Hypercholesterolemia (heFH); subjects with primary hypercholesterolemia who are statin intolerant or statin uncontrolled; and subjects at risk for developing hypercholesterolemia who may be preventably treated. Other indications include dyslipidemia associated with secondary causes such as Type 2 diabetes mellitus, cholestatic liver diseases (primary biliary cirrhosis), nephrotic syndrome, hypothyroidism, obesity; and the prevention and treatment of atherosclerosis and cardiovascular diseases. id="p-1000" id="p-1000"
[001000] In specific embodiments of the method of the invention, the ligand of the invention (eg, anti-hPCSK9 antibody or antibody fragment of the invention) is useful to reduce elevated total cholesterol, non-HDL cholesterol, LDL cholesterol, and/or apolipoprotein B (apolipoprotein Bl00). [001001] The ligand (eg, antibody or antigen-binding fragment) of the invention may be used alone or in combination with a second agent, for example, an HMG-CoA reductase inhibitor and/or another lipid lowering drug. id="p-1002" id="p-1002"
[001002] Treatment Population id="p-1003" id="p-1003"
[001003] The invention provides therapeutic methods for treating a human patient in need of a composition or ligand of the invention. While modifications in lifestyle and conventional drug treatment are often successful in reducing cholesterol levels, not all patients are able to achieve the recommended target cholesterol levels with such approaches. Various conditions, such as familial hypercholesterolemia (FH), appear to be resistant to lowering of LDL-C levels in spite of aggressive use of conventional therapy. Homozygous and heterozygous familial hypercholesterolemia (hoFH, heFH) is a condition associated with premature atherosclerotic vascular disease. However, patients diagnosed with hoFH are largely unresponsive to conventional drug therapy and have limited treatment options. Specifically, treatment with statins, which reduce LDL-C by inhibiting cholesterol synthesis and upregulating the hepatic LDL receptor, may have little effect in patients whose LDL receptors are non-existent or defective. A mean LDL-C reduction of only less than about 20% has 235 been recently reported in patients with genotype-confirmed hoFH treated with the maximal dose of statins. The addition of ezetimibe 10 mg/day to this regimen resulted in a total reduction of LDL-C levels of 27%, which is still far from optimal. Likewise, many patients are statin non-responsive, poorly controlled with statin therapy, or cannot tolerate statin therapy; in general, these patients are unable to achieve cholesterol control with alternative treatments. There is a large unmet medical need for new treatments that can address the short-comings of current treatment options. id="p-1004" id="p-1004"
[001004] Specific populations treatable by the therapeutic methods of the invention include patients indicated for LDL apheresis, subjects with PCSK9-activating (GOF) mutations, heterozygous Familial Hypercholesterolemia (heFH); subjects with primary hypercholesterolemia who are statin intolerant or statin uncontrolled; and subjects at risk for developing hypercholesterolemia who may be preventably treated. id="p-1005" id="p-1005"
[001005] Therapeutic Administration and Formulations id="p-1006" id="p-1006"
[001006] The invention provides therapeutic compositions comprising the anti-PCSK9 ligands, 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. 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. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINT™), 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. id="p-1007" id="p-1007"
[001007] 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. When the ligand, eg, antibody, ofthe present invention is used for treating various conditions and diseases associated with PCSK9, including hypercholesterolemia, disorders associated with LDL and apolipoprotein B, and lipid metabolism disorders, and the like, in an adult patient, ii is advantageous to intravenously administer the ligand or antibody ofthe 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. Depending on the severity ofthe condition, the frequency and the duration of the treatment can be adjusted. id="p-1008" id="p-1008"
[001008] Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, thus the composition invention provides the ligand 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, 236 262:4429-4432). Methods of introduction include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The 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. id="p-1009" id="p-1009"
[001009] The 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.). id="p-1010" id="p-1010"
[001010] In certain situations, the pharmaceutical composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201). In another embodiment, polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974). In yet another embodiment, 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. J 15-138, 1984). [0010111 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. As the 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. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared is preferably filled in an appropriate ampoule. A pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe. In addition, with respect to subcutaneous delivery, 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. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, 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. In a disposable pen delivery device, there is no replaceable 237 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. id="p-1012" id="p-1012"
[001012] Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPEN™ (Owen Mumford, Inc., Woodstock, UK), DISETRONIC™ pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25™ pen, HUMALOG™ pen, HUMALIN 70/30™ pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPEN™!, Π and Ill (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIOR™ (Novo Nordisk, Copenhagen, Denmark), BD™ pen (Becton Dickinson, Franklin Lakes, NJ.), OPTIPENT™, OPT1PEN PRO™, OPTIPEN STARLET™, and OPTICLIKT™ (sanofi-aventis, Frankfurt, Germany), to name only a few. Examples of 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 SOLOSTAR™ pen (sanofi-aventis), the FLEXPEN™ (Novo Nordisk), and the KWIKPEN™ (Eli Lilly). id="p-1013" id="p-1013"
[001013] Advantageously, 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 active ingredients. Such 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. id="p-1014" id="p-1014"
[001014] The invention provides therapeutic methods in which the ligand, eg, antibody or antibody fragment, of the invention is useful to treat hypercholesterolemia associated with a variety of conditions involving hPCSK9. The anti-PCSK9 ligands, eg, antibodies or antibody fragments, of the invention are particularly useful for the treatment of hypercholesterolemia and the like. Combination therapies may include the anti-PCSK9 ligand of the invention with, for example, one or more of any agent that (1) induces acellular depletion of cholesterol synthesis by inhibiting 3-hydroxy-3methylglutaryl (HMG)-coenzyme A (CoA) reductase, such as cerivastatin, atorvastatin, simvastatin, pitavastatin, rosuvastatin, fluvastatin, lovastatin, pravastatin; (2) inhibits cholesterol uptake and or bile acid re-absorption; (3) increase lipoprotein catabolism (such as niacin); and activators of the LXR transcription factor that plays a role in cholesterol elimination such as 22-hydroxycholesteroI or fixed combinations such as ezetimibe plus simvastatin; a statin with a bile resin (e.g., cholestyramine, colestipol, colesevelam), a fixed combination of niacin plus a statin (e.g., niacin with lovastatin); or with other lipid lowering agents such as omega-3-fatty acid ethyl esters (for example, omacor). id="p-1015" id="p-1015"
[001015] Tailoring Antibodies to Rare PCSK9 Variant Profile 238 id="p-1016" id="p-1016"
[001016] As outline above, the invention includes the possibility to tailor treatment of humans further by selecting antibody-based ligands with variable domains based on gene segments commonly found in humans of the ethnic populations where the variant PCSK9 forms are found to meet the selection criteria of the invention. An example is provided below for ligands comprising antibody VH domains derived from recombination of human VH3-23. id="p-1017" id="p-1017"
[001017] The inventor analysed the frequencies and distribution of various human VH3-23 alleles and realised the desirability of using ligands based on human VH3-23 alleles comprising SNP rs56069819. This SNP corresponds to a change from leucine at position 24 in the encoded protein sequence to a valine at that position (L24V change) and the SNP is at coordinate 106725482 on human chromosome 14. id="p-1018" id="p-1018"
[001018] Figure 2 shows the cumulative allele frequency distribution across the 1000 Genomes Project database 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. 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. Table 7 shows the VH3-23 variants and the SNPs that they comprise, as well as their cumulative allele frequencies as found in the 1000 Genomes Project database. id="p-1019" id="p-1019"
[001019] Notably, human VH3-23 alleles comprising SNP rs56069819 were found in the CEU population at a frequency that is almost double the frequency of 11 % for all populations. For the ASW and YRI populations the frequency was over a quarter of the population. Thus, the invention advantageously enables one to select a ligand comprising an antibody or antibody fragment, wherein the antibody or fragment comprises a VH domain derived from the recombination of a human VH gene segment, a human D gene segment and a human JH gene segment, the VH gene segment comprising a nucleotide sequence that comprises SNP rs5 6069819 (dbSNP numbering, build number as recited above). (001020] In an example, one can tailor the treatment further by selecting such a ligand that specifically binds to a human PCSK9 selected from forms \f c, m, e, h, p, q and aj, such forms being those appearing in human populations ASW, LWK, YRI, CEU and GBR. id="p-1021" id="p-1021"
[001021] In an example, the VH gene segment is VH3-23*04, which is a commonly found variant that comprises SNP rs56069819 in human populations ASW, LWK, YRI, CEU and GBR. id="p-1022" id="p-1022"
[001022] In an example, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human that expresses a human PCSK9 selected from forms e, ht p, q and aj. [001023] In an example, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human of ASW, LWK, YRJ, CEU or GBR ancestry. 239 id="p-1024" id="p-1024"
[001024] In an embodiment, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human of ASW ancestry, wherein the human expresses a PCSK9 selected from f, c, m, e, h, p and q or the human comprises a corresponding nucleotide or amino acid sequence as set out in Table 6. Optionally this ligand comprises a VH domain derived from recombination of human VH3-23*04. id="p-1025" id="p-1025"
[001025] In an embodiment, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human of LWK ancestry, wherein the human expresses a PCSK9 selected from/ c, m, e and h or the human comprises a corresponding nucleotide or amino acid sequence as set out in Table 6. Optionally this ligand comprises a VH domain derived from recombination of human VH323*04. id="p-1026" id="p-1026"
[001026] In an embodiment, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human of YRI ancestry, wherein the human expresses a PCSK9 selected from/ c, ni e and h or the human comprises a conesponding nucleotide or amino acid sequence as set out in Table 6. Optionally this ligand comprises a VH domain derived from recombination of human VH3-23*04. [001027] In an embodiment, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human of CEU ancestry, wherein the human expresses a PCSK9 selected from f c, p and aj or the human comprises a corresponding nucleotide or amino acid sequence as set out in Table 6. Optionally this ligand comprises a VH domain derived from recombination of human VH3-23*04. [001028] In an embodiment, the ligand is for treating and/or preventing a PCSK9-mediated disease or condition in a human of GBR ancestry, wherein the human expresses a PCSK9 selected from f c and p or the human comprises a corresponding nucleotide or amino acid sequence as set out in Table 6. Optionally this ligand comprises a VH domain derived from recombination of human VH3-23*04. id="p-1029" id="p-1029"
[001029] In an example, the ligand is alirocumab. id="p-1030" id="p-1030"
[001030] In other embodiments, as explained more fully above, the invention provides for ligands which are tailored to the human recipient’s genotype and/or phenotype based on alternative human VH gene segments, or on Vk, VX or constant region gene segments (see further Table 9 for representative variants). id="p-1031" id="p-1031"
[001031] For example, the ligand of the invention comprises or consists of an antibody that 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 IGHV I-18*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV1-I8*O1; or (ii) IGVH 1-46*01 and the genome of the human comprises a human IGHV 1-46*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGHV 1-46*01. id="p-1032" id="p-1032"
[001032] For example, the ligand of the invention comprises or consists of an antibody that comprises a VL domain that is derived from the recombination of a human VL gene segment and a 240 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-l*01; (ii) IGLV2-14*01 and the genome of the human comprises a human IGLV214*01 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGLV2-14*01; or (iii) IGKV 1-13*02 and the genome of the human comprises a human IGKV 1-13*02 nucleotide sequence or the human expresses antibodies comprising variable domains derived from the recombination of human IGKV1-13*O2. id="p-1033" id="p-1033"
[001033] For example, the inventor identified the possibility of addressing the rarer IGHgamma-1 SNPs 204D (observed cumulative frequency of 0.296) and 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. Thus, in this example the ligand of the invention comprises or consists of an antibody that 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. An example of such a ligand is alirocumab. id="p-1034" id="p-1034"
[001034] In another example, the inventor identified the possibility of addressing IGHgamma-2 SNPs. This included consideration of Fc region variation - in this respect, the inventor focused on positions 161 and 257 which are in the Fc region. Thus, in this example the ligand of the invention comprises or consists of an antibody that 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. An example of such a ligand is evolocumab or bococizumab. [001035] In another example, the inventor addressed human kappa constant region variation. Thus, in this example the ligand of the invention comprises or consists of an antibody that 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. An example of such a ligand is alirocumab or bococizumab. 241 id="p-1036" id="p-1036"
[001036] In another example, the inventor addressed human lambda constant region variation. Thus, in this example the ligand of the invention comprises or consists of an antibody that 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. An example of such a ligand is evolocumab. [001037] Further exemplary ligands are in the paragraphs 1-17 as follows.
J. An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a human gamma heavy chain constant region that comprises a first amino acid that is encoded by a human gamma heavy chain constant region gene segment SNP, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation I474V or E670G in SEQ ID NO: I, wherein the human comprises a nucleotide sequence encoding said PCSK9 amino acid sequence and comprises a human gamma heavy chain constant region gene segment comprising said SNP, or the human expresses antibodies comprising human gamma constant regions comprising said first amino acid.
In an alternative, paragraph I provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment 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 the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation I474V or E670G in SEQ ID NO:1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an 1GHG1 *01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-1 constant regions comprising such an Asp and Leu.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G. An example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a 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, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a 242 C-terminal domain comprising a mutation I474V in SEQ ID NO:1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGHG1*O1 human heavy chain constant region gene segment. Optionally, the antibody or fragment comprises an IGHGI *01 human heavy chain constant region.
Another example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a 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, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation E670G in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGHGI *01 human heavy chain constant region gene segment. Optionally, the antibody or fragment comprises an IGHGI *01 human heavy chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a 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 and wherein said human comprises (i) an IGHG I *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation 1474 V or E670G in SEQ ID NO: 1. 2. The antibody or antibody fragment of paragraph 1. wherein the antibody comprises a 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. 3. The antibody or antibody fragment of paragraph 1 or 2, wherein the antibody comprises an IGHGI *01 human heavy chain constant region. 4. The antibody or antibody fragment of any one of paragraphs 1 to 3, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a Cterminal domain comprising said mutation 1474V or E670G in SEQ ID NO: 1 and/or a 243 proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30.
. The antibody or antibody fragment of any one of paragraphs 1 to 4, the method comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 6. The antibody or antibody fragment of paragraph 5, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: E 7. The antibody or antibody fragment of paragraph 6, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and 244 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 8. The antibody or antibody fragment of paragraph 6 or 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 9. The antibody or antibody fragment of any one of paragraphs 1 to 8, wherein said antibody or antibody fragment is for administration to a human that is or has been further determined to be substantially resistant to statin treatment.
. The antibody or antibody fragment of any one of paragraphs 1 to 9, wherein antibody or antibody fragment is for administration to a human that is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 11. The antibody or antibody fragment of paragraph 9 or 10, wherein said antibody or antibody fragment is for administration to the human separately or simultaneously with said statin treatment. 12. The antibody or antibody fragment of any one of paragraphs 6 to 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 13. The antibody or antibody fragment of any one of paragraphs 1 to 12, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474Vor E670G in SEQ ID NO: 1. 14. The antibody or antibody fragment of any one of paragraphs 1 to 13, wherein said human has been diagnosed with at least one condition selected from a lipid disorder. hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and 245 high blood pressure.
. The antibody or antibody fragment of any one of paragraphs 1 to 14, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 16. The antibody or antibody fragment of any one of paragraphs I to 15, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 17. The antibody or antibody fragment of any one of paragraphs I to 16, wherein said antibody or antibody fragment is for administration by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1038" id="p-1038"
[001038] Further exemplary methods are in the paragraphs 1Ί 8 as follows. 1. 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 1474 or E670G in SEQ ID NO: I, wherein the antibody or fragment 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 said human comprises (i) an IGHGl*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-1 heavy chain constant regions comprising such an Asp and Leu and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation 1474V or E670G in SEQ ID NO: 1.
In an embodiment, said mutation is 1474V. In another embodiment, said mutation is E670G.
An example 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 1474 in SEQ ID NO: 1, wherein the antibody or fragment comprises a 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 and wherein said human comprises (i) an IGHGl*01 human heavy chain 246 constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation 1474V in SEQ ID NO: 1. Optionally, the antibody or fragment comprises a IGHG1 *01 human heavy chain constant region.
Another example 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 E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a 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 and wherein said human comprises (i) an IGHG1 *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation E670G in SEQ ID NO: 1. Optionally, the antibody or fragment comprises an IGHG1 *01 human heavy chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a 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 and wherein said human comprises (i) an IGHG1 *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1. 2. The method of paragraph 1, comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of paragraph 1. 3. The method of paragraph 1 or 2, wherein the antibody comprises a 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. 247 4. The method of paragraph 1, 2 or 3, wherein the antibody comprises an 1GHG1 *01 human heavy chain constant region.
. The method of any one of paragraphs 1 to 4, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 6. The method of any one of paragraphs 1 to 5, comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-tenninal domain comprising said mutation 1474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The method of paragraph 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the Cterminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1. 8. The method of paragraph 7, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Cterminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 is present; and 248 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 9. The method of paragraph 7 or 8, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
. The method of any one of paragraphs 1 to 9, wherein said human is or has been further determined to be substantially resistant to statin treatment. 11. The method of any one of paragraphs 1 to 10, wherein said human is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 12. The method of paragraph 10 or 11, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said statin treatment. 13. The method of any one of paragraphs 7 to 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 14. The method of any one of paragraphs 1 to 13, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation I474Vor E670G in SEQ ID NO: 1.
. The method of any one of paragraphs 1 to 14, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 16. The method of any one of paragraphs 1 to 15, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a 249 stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 17. The method of any one of paragraphs 1 to 16, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 18. The method of any one of paragraphs I to 17, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1039" id="p-1039"
[001039] Further exemplary ligands are in the paragraphs 1-17 as follows. 1. An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody 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 the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation I474V or E670G in SEQ ID NO:I, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGHG2*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-2 constant regions comprising such a Pro. Asn, Phe, Val and Ala.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G.
An example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody comprises a human gamma-2 heavy chain constant region that comprises 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 the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation I474V in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGHG2*01 human heavy chain constant region gene segment. Optionally, the 250 antibody or fragment comprises an iGHG2*01 human heavy chain constant region.
Another example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody comprises a human gamma-2 heavy chain constant region that comprises 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 the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation E670G in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGHG2*01 human heavy chain constant region gene segment. Optionally, the antibody or fragment comprises an IGHG2*01 human heavy chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human gamma-2 heavy chain constant region that comprises 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 said human comprises (i) an IGHG2*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1. 2. The antibody or antibody fragment of paragraph 1, wherein the antibody comprises a human gamma-2 heavy chain constant region that comprises 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. 3. The antibody or antibody fragment of paragraph 1 or 2, wherein the antibody comprises an IGHG2*01 human heavy chain constant region. 251 4. The antibody or antibody fragment of any one of paragraphs 1 to 3, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30.
. The antibody or antibody fragment of any one of paragraphs I to 4, the method comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation 1474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation J474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 6. The antibody or antibody fragment of paragraph 5, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1. 7. The antibody or antibody fragment of paragraph 6, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to al least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and 252 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 8. The antibody or antibody fragment of paragraph 6 or 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 9. The antibody or antibody fragment of any one of paragraphs 1 to 8, wherein said antibody or antibody fragment is for administration to a human that is or has been further determined to be substantially resistant to statin treatment.
. The antibody or antibody fragment of any one of paragraphs 1 to 9, wherein antibody or antibody fragment is for administration to a human that is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 11. The antibody or antibody fragment of paragraph 9 or 10, wherein said antibody or antibody fragment is for administration to the human separately or simultaneously with said statin treatment. 12. The antibody or antibody fragment of any one of paragraphs 6 to 8, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 13. The antibody or antibody fragment of any one of paragraphs 1 to 12, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK.9 C-terminal domain comprising a mutation I474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474Vor E670G in SEQ ID NO: I. 14. The antibody or antibody fragment of any one of paragraphs 1 to 13, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 253 . The antibody or antibody fragment of any one of paragraphs 1 to 14, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 16. The antibody or antibody fragment of any one of paragraphs 1 to 15, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 17. The antibody or antibody fragment of any one of paragraphs 1 to 16, wherein said antibody or antibody fragment is for administration by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1040" id="p-1040"
[001040] Further exemplary methods are in the paragraphs 1-18 as follows. 1. 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 1474 or E670G in SEQ ID NO: I, wherein the antibody or fragment 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 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 such a Pro, Asm Phe, Val and Ala and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation 1474V or E670G in SEQ ID NO: 1.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G.
An example 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 1474 in SEQ ID NO: 1, wherein the 254 antibody or fragment comprises a human gamma-2 heavy chain constant region that comprises 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 said human comprises (i) an IGHG2*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V in SEQ ID NO: 1. Optionally, the antibody or fragment comprises an IGHG2*01 human heavy chain constant region.
Another example 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 E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human gamma-2 heavy chain constant region that comprises 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 ofSEQIDNO:44 and an Ala corresponding to position 257 of SEQ ID NO: 44 and wherein said human comprises (i) an IGHG2*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation E670G in SEQ ID NO: 1. Optionally, the antibody or fragment comprises an 1GHG2*O1 human heavy chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human gamma-2 heavy chain constant region that comprises 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 said human comprises (i) an IGHG2*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1. 255 2. The method of paragraph 1, comprising, before said administering, selecting said human comprising said nucleotide sequence of (ii). 3. The method of paragraph 1 or 2, wherein the antibody comprises a human gamma-1 heavy chain constant region that comprises 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. 4. The method of paragraph 1, 2 or 3, wherein the antibody comprises an IGHG2*01 human heavy chain constant region.
. The method of any one of paragraphs 1 to 4, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation 1474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 6. The method of any one of paragraphs 1 to 5, comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The method of paragraph 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 8. The method of paragraph 7, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation J474V or E670G in SEQ ID NO: 1 is present; and/or 256 b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Ctcrminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 9. The method of paragraph 7 or 8, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
. The method of any one of paragraphs 1 to 9, wherein said human is or has been further determined to be substantially resistant to statin treatment. 11. The method of any one of paragraphs 1 to 10, wherein said human is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 12. The method of paragraph 10 or 11, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said statin treatment. 13. The method of any one of paragraphs 7 to 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 14. The method of any one of paragraphs 1 to 13, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation I474Vor E670G in SEQ ID NO: 1. 257 . The method of any one of paragraphs 1 to 14, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 16. The method of any one of paragraphs 1 to 15, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 17. The method of any one of paragraphs 1 to 16, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 18. The method of any one of paragraphs 1 to 17, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1041" id="p-1041"
[001041] Further exemplary ligands are in the paragraphs 1-17 as follows. 1. An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment 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 the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation 1474V or E670G in SEQ ID NO:1. wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGKC*01 human light chain constant region gene segment, or the human expresses antibodies comprising human kappa light chain constant regions comprising such an Val and Cys.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G.
An example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a human kappa light chain constant region that comprises a Val 258 corresponding to position 84 of SEQ ID NO: 50 and a Cys corresponding to position 87 of SEQ ID NO: 50, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation I474V in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGKC*01 human light chain constant region gene segment. Optionally, the antibody or fragment comprises an JGKC*01 human light chain constant region.
Another example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment 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, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation E670G in SEQ ID NO: I, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises an IGKC*01 human light chain constant region gene segment. Optionally, the antibody or fragment comprises an IGKC*01 human light chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment 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 and wherein said human comprises (i) an IGKC*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1. 2. The antibody or antibody fragment of paragraph 1, wherein the antibody 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. 3. The antibody or antibody fragment of paragraph 1 or 2, wherein the antibody comprises an IGKC*01 human kappa chain constant region. 259 4. The antibody or antibody fragment of any one of paragraphs 1 to 3, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation 1474V or E670G in SEQ ID NO: I and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30.
. The antibody or antibody fragment of any one of paragraphs 1 to 4. the method comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 6. The antibody or antibody fragment of paragraph 5, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1, 7. The antibody or antibody fragment of paragraph 6, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and 260 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 8. The antibody or antibody fragment of paragraph 6 or 7, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 9. The antibody or antibody fragment of any one of paragraphs 1 to 8, wherein said antibody or antibody fragment is for administration to a human that is or has been further determined to be substantially resistant to statin treatment.
. The antibody or antibody fragment of any one of paragraphs 1 to 9, wherein antibody or antibody fragment is for administration to a human that is receiving or has received statin treatment or has reduced responsiveness to statin treatment.
I I. The antibody or antibody fragment of paragraph 9 or 10, wherein said antibody or antibody fragment is for administration to the human separately or simultaneously with said statin treatment. 12. The antibody or antibody fragment of any one of paragraphs 6 to 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 13. The antibody or antibody fragment of any one of paragraphs 1 to 12, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ JD NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474Vor E670G in SEQ ID NO: I. 14. The antibody or antibody fragment of any one of paragraphs 1 to 13, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 261 . The antibody or antibody fragment of any one of paragraphs 1 to 14, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. ] 6. The antibody or antibody fragment of any one of paragraphs 1 to 15, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 17. The antibody or antibody fragment of any one of paragraphs 1 to 16, wherein said antibody or antibody fragment is for administration by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1042" id="p-1042"
[001042] Further exemplary methods are in the paragraphs 1-18 as follows, 1. 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 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment 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 said human comprises (i) an IGKC*01 human light chain constant region gene segment, or the human expresses antibodies comprising human kappa light chain constant regions comprising such an Val and Cys and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1.
In an embodiment, said mutation is 1474V. In another embodiment, said mutation is E670G.
An example 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 1474 in SEQ ID NO: 1, wherein the antibody or fragment 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 and wherein said human comprises (i) an IGKC*01 human light chain constant region 262 gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation 1474V in SEQ ID NO: 1. Optionally, the antibody or fragment comprises an IGKC*01 human light chain constant region.
Another example 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 E670G in SEQ ID NO: 1, wherein the antibody or fragment 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 and wherein said human comprises (i) an 1GKC*O1 human light chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation E670G in SEQ ID NO: I. Optionally, the antibody or fragment comprises an IGKC*01 human light chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1. wherein the antibody or fragment 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 and wherein said human comprises (i) an IGKC*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation J474V or E670G in SEQ ID NO: 1. 2. The method of paragraph 1, comprising, before said administering, selecting said human comprising said nucleotide sequence of (ii). 3. The method of paragraph 1 or 2, wherein the antibody 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. 263 4. The method of paragraph 1,2 or 3, wherein the antibody comprises an IGKC*01 human kappa chain constant region.
. The method of any one of paragraphs 1 to 4, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ JD NO: 29 or 30. 6. The method of any one of paragraphs 1 to 5, comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation 1474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation 1474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 7. The method of paragraph 6, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 8. The method of paragraph 7, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and 264 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 9. The method of paragraph 7 or 8, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
. The method of any one of paragraphs 1 to 9, wherein said human is or has been further determined to be substantially resistant to statin treatment. 11. The method of any one of paragraphs 1 to 10, wherein said human is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 12. The method of paragraph 10 or 11, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said statin treatment. 13. The method of any one of paragraphs 7 to 9, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 14. The method of any one of paragraphs 1 to 13, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation J474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation I474Vor E670G in SEQ ID NO: 1.
. The method of any one of paragraphs 1 to 14, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 16. The method of any one of paragraphs 1 to 15, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a 265 stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 17. The method of any one of paragraphs 1 to 16, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 18. The method of any one of paragraphs 1 to 17, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1043" id="p-1043"
[001043] Further exemplary ligands are in the paragraphs 1-15 as follows. 1. An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a human 1GLC2*O1 lambda light chain constant region, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation I474V or E670G in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises a human 1GLC2*O1 lambda light chain constant region gene segment, or the human expresses antibodies comprising human IGLC2*01 lambda light chain constant regions.
In an embodiment, said mutation is 1474V. In another embodiment, said mutation is E670G.
An example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kezxin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation 1474V in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises a human IGLC2*01 lambda light chain constant region gene segment. Optionally, the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region.
Another example provides:An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region, and the antibody 266 or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation E670G in SEQ ID NO:1 wherein the human comprises a nucleotide sequence encoding said amino acid sequence and comprises a human IGLC2*01 lambda light chain constant region gene segment. Optionally, the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region and wherein said human comprises (i) an IGLC2*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: I. 2. The antibody or antibody fragment of paragraph 1, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 3. The antibody or antibody fragment of paragraphs 1 or 2, the method comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 4. The antibody or antibody fragment of paragraph 3, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1.
. The antibody or antibody fragment of paragraph 4, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a 267 nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ DD NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 6. The antibody or antibody fragment of paragraph 4 or 5, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 7. The antibody or antibody fragment of any one of paragraphs 1 to 6, wherein said antibody or antibody fragment is for administration to a human that is or has been further determined to be substantially resistant to statin treatment. 8. The antibody or antibody fragment of any one of paragraphs 1 to 7, wherein antibody or antibody fragment is for administration to a human that is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 9. The antibody or antibody fragment of paragraph 7 or 8, wherein said antibody or antibody fragment is for administration to the human separately or simultaneously with said statin 268 treatment.
. The antibody or antibody fragment of any one of paragraphs 4 to 8, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The antibody or antibody fragment of any one of paragraphs 1 to 10, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation 1474Vor E670G in SEQ ID NO: 1. 12. The antibody or antibody fragment of any one of paragraphs 1 to 11, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 13. The antibody or antibody fragment of any one of paragraphs 1 to 12, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 14. The antibody or antibody fragment of any one of paragraphs 1 to 13, wherein the nucleotide sequence is SEQ ID NO: 29 or 30.
. The antibody or antibody fragment of any one of paragraphs 1 to 14, wherein said antibody or antibody fragment is for administration by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1044" id="p-1044"
[001044] Further exemplary methods are in the paragraphs 1-16 as follows. 1. 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 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human IGLC2*01 lambda light chain constant 269 region and wherein said human comprises (i) an IGLC2*01 human light chain constant region gene segment, or the human expresses antibodies comprising a human IGLC2*01 lambda light chain constant regions and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation 1474V or E670G in SEQ ID NO: 1.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G.
An example 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 1474 in SEQ ID NO: 1, wherein the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region and wherein said human comprises (i) an 1GLC2*O1 human light chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V in SEQ ID NO: 1. Optionally, the antibody or fragment comprises an IGLC2*01 human light chain constant region.
Another example 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 E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human IGLC2*01 lambda light chain constant region and wherein said human comprises (i) an 1GLC2*O1 human light chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation E670G in SEQ ID NO: 1. Optionally, the antibody or fragment comprises an 1GLC2*O1 human light chain constant region.
In an example, said antibody or antibody fragment has been determined to specifically bind a proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising a mutation 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human 1GLC2*O1 lambda light chain constant region and wherein said human comprises (i) an IGLC2*01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that 270 comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 2. The method of paragraph 1, comprising, before said administering, selecting said human comprising said nucleotide sequence of (ii). 3. The method of paragraph 1 or 2, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: I and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 4. The method of any one of paragraphs 1 to 3, comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human.
. The method of paragraph 4, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 6. The method of paragraph 5, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: I is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and 271 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 7. The method of paragraph 5 or 6, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 8. The method of any one of paragraphs 1 to 7, wherein said human is or has been further determined to be substantially resistant to statin treatment. 9. The method of any one of paragraphs 1 to 8, wherein said human is receiving or has received statin treatment or has reduced responsiveness to statin treatment.
. The method of paragraph 8 or 9, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said statin treatment. 11. The method of any one of paragraphs 5 to 7, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 12. The method of any one of paragraphs 1 to 11, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474Vor E670G in SEQ ID NO: 1. . 13. The method of any one of paragraphs 1 to 12, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 14. The method of any one of paragraphs 1 to 13, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a 272 stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure.
. The method of any one of paragraphs 1 to 14, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 16. The method of any one of paragraphs 1 to 15, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. id="p-1045" id="p-1045"
[001045] Further exemplary ligands are in the paragraphs 1-15 as follows. 1. An antibody or antibody fragment for use in a method of reducing cholesterol level or maintaining previously reduced cholesterol level in a human in need thereof, wherein the antibody or fragment comprises a human variable 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 as defined in any one of clauses 80 to 90, and the antibody or fragment specifically binds a proprotein convertase subtilisin/kexin type 9 (PCSK9) amino acid sequence that comprises a C-terminal domain comprising a mutation 1474V or E670G in SEQ ID NO: 1, wherein the human comprises a nucleotide sequence encoding said amino acid sequence, and comprises said VH gene segment or expresses antibodies comprising VH domains that are derived from the recombination of said human VH gene segment, a human D gene segment and a human JH gene segment.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G. 2. The antibody or antibody fragment of paragraph 1, wherein the human has been determined io comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 3. The antibody or antibody fragment of paragraphs 1 or 2, the method comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration ofthe antibody to the 273 human. 4. The antibody or antibody fragment of paragraph 3, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1.
. The antibody or antibody fragment of paragraph 4, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: I is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present: and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 6. The antibody or antibody fragment of paragraph 4 or 5, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 274 7. The antibody or antibody fragment of any one of paragraphs 1 to 6, wherein said antibody or antibody fragment is for administration to a human that is or has been further determined to be substantially resistant to statin treatment. 8. The antibody or antibody fragment of any one of paragraphs 1 to 7, wherein antibody or antibody fragment is for administration to a human that is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 9. The antibody or antibody fragment of paragraph 7 or 8, wherein said antibody or antibody fragment is for administration to the human separately or simultaneously with said statin treatment. ] 0. The antibody or antibody fragment of any one of paragraphs 4 to 8, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 11. The antibody or antibody fragment of any one of paragraphs 1 to 10, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation I474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising a mutation 1474Vor E670G in SEQ ID NO: 1. 12. The antibody or antibody fragment of any one of paragraphs 1 to 11, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 13. The antibody or antibody fragment of any one of paragraphs 1 to 12, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 14. The antibody or antibody fragment of any one of paragraphs 1 to 13, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 275 . The antibody or antibody fragment of any one of paragraphs 1 to 14, wherein said antibody or antibody fragment is for administration by intravenous or subcutaneous administration and/or is comprised in an injectable preparation.
Further exemplary methods are in the paragraphs 1-16 as follows. 1. 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 1474 or E670G in SEQ ID NO: 1, wherein the antibody or fragment comprises a human variable 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 as defined in any one of clauses 80 to 90 and wherein said human comprises (i) comprises said VH gene segment or expresses antibodies comprising VH domains that are derived from the recombination of said human VH gene segment, a human D gene segment and a human JH gene segment and (ii) a nucleotide sequence encoding said proprotein convertase subtilisin/kexin type 9 (PCSK9) that comprises a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1.
In an embodiment, said mutation is I474V. In another embodiment, said mutation is E670G. 2. The method of paragraph 1, comprising, before said administering, selecting said human comprising said nucleotide sequence of (ii). 3. The method of paragraph 1 or 2, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 4. The method of any one of paragraphs 1 to 3, comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation 1474 V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 276 . The method of paragraph 4, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 6. The method of paragraph 5, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein 10 said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: I is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides 15 of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a C- terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 7. The method of paragraph 5 or 6, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 8. The method of any one of paragraphs 1 to 7, wherein said human is or has been further determined to be substantially resistant to statin treatment. , 9. The method of any one of paragraphs 1 to 8, wherein said human is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 277 . The method of paragraph 8 or 9, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said statin treatment. 11. The method of any one of paragraphs 5 to 7, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 12. The method of any one of paragraphs 1 to 11, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474 V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation I474Vor E670G in SEQ ID NO: 1. 13. The method of any one of paragraphs 1 to 12, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronaiy heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 14. The method of any one of paragraphs 1 to 13, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure.
. The method of any one of paragraphs 1 to 14, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 16. The method of any one of paragraphs 1 to 15. wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 1001046] Regimens id="p-1047" id="p-1047"
[001047] A: The invention further provides the following regimens, ligands and kits. 22. A method for treating a human PCSK9-mediated disease or condition in a human by targeting a rare variant human PCSK9, the method comprising administering to the human a ligand (eg, an 278 antibody or fragment) that has been determined to specifically bind to said PCSK9variant; wherein the human expresses said PCSK9 variant or the genome of the human comprises a nucleotide sequence encoding said PCSK9 variant; wherein said human is treated for said disease or condition.
The variant PCSK9 can, for example, be any rare variant as described herein.
For example, there is provided: la. A method for treating a PCSK9-mediated disease or condition in a human by targeting a PCSK9 that comprises a C-terminal domain amino acid polymorphism (compared to SEQ ID NO: 1), the method comprising administering to the human a ligand (eg, an antibody or fragment) that has been determined to specifically bind to a PCSK9 comprising a C-terminal domain comprising I474V or 670G (numbering according to SEQ ID NO:1); wherein the human expresses said PCSK9 or the genome of the human comprises a nucleotide sequence encoding said PCSK9; wherein said human is treated for said disease or condition.
In an embodiment, determination of said specific binding is by reference to binding assay data, eg, as determined using SPR or ELISA. Determination may, for example, be by reference to information in a printed publication, eg, with knowledge of data presented in the present or another patent application or in a journal article. Once armed with such knowledge (eg, in the absence of further testing of binding), the skilled person is able - by direction of the present invention - to treat a relevant human whose genotype or phenotype matches the binding specificity of the ligand.
The antibody or fragment can be according to any configuration, example, embodiment, aspect, clause or paragraph herein.
In an embodiment, the method comprises, before said administering, selecting a human comprising said nucleotide sequence encoding the PCSK9, wherein the human is said human in clause 1 (eg, la). 23. The method of clause ], comprising before said administering the step of determining that the ligand specifically binds to said PCSK9, eg, using SPR or ELISA. 279 24. The method of clause 1 or 2, wherein the specific binding to said PCSK9 is binding with a dissociation constant (Kd) of InM or less, eg, 100, 10 or IpM or less.
. The method of any of clauses 1 to 3 (eg, clause la), wherein the condition is elevated LDLcholesterol or is caused by elevated LDL-cholesterol. 26. The method of clause 4 (eg, when dependent from clause la), wherein the condition is selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 27. The method of any one of clauses 1 to 5 (eg, when dependent from clause la), wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a Cterminal domain comprising said mutation I474V or E670G in SEQ ID NO: I and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 28. The method of any one of clauses 1 to 6 (eg, when dependent from clause la), comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation 1474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation I474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 29. The method of clause 7, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1.
. The method of clause 8, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex 280 when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Cterminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 31. The method of clause 8 or 9, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 32. The method of any one of clauses 1 to 10, wherein said human is or has been further determined to be substantially resistant to statin treatment. 33. The method of any one of clauses I to 11, wherein said human is receiving or has received statin treatment or has reduced responsiveness to statin treatment. 34. The method of clauses 11 or 12, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with said statin treatment.
. The method of any one of clauses 8 to 10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 36. The method of any one of clauses 1 to 14, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation I474V or E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding 281 the PCSK9 C-terminal domain comprising said mutation I474Vor E670G in SEQ ID NO: 1. 37. The method of any one of clauses 1 to 15, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 38. The method of any one of clauses 1 to 16, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 39. The method of any one of clauses 1 to 17, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 40. The method of any one of clauses 1 to 18, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 41. A ligand (eg, an antibody or fragment) for use in the method of any one of clauses 1 to 19, wherein the ligand specifically binds the PCSK9. 42. A kit comprising the ligand of clause 20 and instructions for carrying out the method of any one of clauses 1 to 19. id="p-1048" id="p-1048"
[001048] B: The invention further provides the following regimens, ligands and kits. 21. A method of reducing cholesterol level or maintaining a previously reduced cholesterol level in a human in need thereof, the method comprising:- a. Carrying out an initial treatment of said human for an initial treatment period by administering an anti-human PCSK9 ligand (eg, an antibody or fragment) to said human, wherein (i) the ligand has been determined to specifically bind to a PCSK9 comprising a C-terminal domain comprising I474V or 670G (numbering according to SEQ ID NO:1); (ii) the human expresses said PCSK9 or the genome of the human comprises a nucleotide 282 sequence encoding said PCSK9 and (iii) the human has received or is receiving statin treatment to lower or maintain cholesterol level; wherein the initial treatment comprises the administration of a single or multiple doses of the ligand to the human; b. Determining to (i) terminate statin treatment (ii) keep the human off statin treatment; or (iii) reduce statin treatment after said initial treatment period; and c. Continuing to administer the ligand to said patient after said time period has expired, thereby reducing cholesterol level or maintaining a previously reduced cholesterol level in said human.
In an embodiment, determination of said specific binding is by reference to binding assay data, eg, as determined using SPR or ELISA. Determination may, for example, be by reference to information in a printed publication, eg, with knowledge of data presented in the present or another patent application or in a journal article. Once armed with such knowledge (eg, in the absence of further testing of binding), the skilled person is able - by direction of the present invention - to treat a relevant human whose genotype or phenotype matches the binding specificity of the ligand.
The antibody or fragment can be according to any configuration, example, embodiment, aspect, clause or paragraph herein.
A pharmaceutically-effective amount of said ligand is administered.
In an embodiment, the method comprises, before said administering, selecting a human comprising said nucleotide sequence encoding the PCSK9, wherein the human is said human recited in clause 1.
In an example, the initial treatment period is 7 days, 14 days, 21 days, 28 days, a month, two months, three months, four months, five months, six months, seven months, eight months, nine months or a year. 22. The method of clause 1, wherein the human has or is suffering from statin-associated memory loss or a statin-associated neurodegenerative condition, or has or is at increased risk of diabetes (eg, statin-associated diabetes). 23. The method of clause 1 or 2, comprising, before said initial treatment, the step of determining that the human has or is suffering from statin-associated memory loss or a statin-associated neurodegenerative condition, or has or is at increased risk of diabetes (eg, statin-associated 283 diabetes). 24. The method of clause 2 or 3, comprising, after step (b) (eg, during step (c)) determining that the memory loss or said neurodegenerative condition has improved.
. The method of any one of clauses 1 to 4, wherein said human is over 40 years of age (eg, 50 or over, 55 or over, 60 or over, 65 or over, or 70 or over). 26. The method of any one of clauses 1 to 5, wherein step (c) comprises determining to increase the doses of said ligand to be administered after said initial treatment period and administering said increased doses to said human. 27. The method of any one of clauses 1 to 6, wherein step (b) comprises determining that the human is intolerant or refactory to treatment by a statin. 28. The method of any one of clauses 1 to 7, wherein the initial treatment comprises the administration of a statin, fenofibrate (eg, Tricor™ or Lofibra™) or ezetimibe to the human in addition to the ligand. 29. The method of any one of clauses I to 8, wherein step (b) comprises terminating or reducing statin, fenofibrate (eg, Tricor™ or Lofibra™) or ezetimibe treatment during step (c).
. The method of any one of clauses 1 to 9, comprising increasing (ie, increasing compared to the initial treatment dose) the ligand dose during step (c). 1. The method of any one of clauses 1 to 10, wherein the human has received high dose statin treatment prior to the initial treatment, and wherein step (c) comprises administering a lower (eg, a medium or low) dose statin treatment in addition to said ligand.
The skilled person is familiar with the meaning of high, medium and low dose treatments (and how to determine according to each patient, eg, the patient’s body mass). For example, the statin is selected from rosuvastatin, atorvastatin and simvastatin.
For example daily statin doses are as follows:- Low 10 to 20mg (eg, 1 Omg); medium >20 and <60mg (eg, 40mg); high 60-100mg (eg, 80mg). 284 32. The method of any one of clauses 1 to 10, wherein the human has received medium dose statin treatment prior to the initial treatment, and wherein step (c) comprises administering a lower (eg, a low) dose statin treatment or no statin in addition to said ligand. 33. The method of any one of clauses 1 to 10, wherein the human has received low dose statin treatment prior to the initial treatment, and wherein step (c) comprises administering no statin in addition to said ligand. 34. The method of any one of clauses 1 to 13, comprising, before the initial treatment, the step of determining that the ligand specifically binds to said PCSK9, eg, using SPR or ELISA.
. The method of any one of clauses 1 to 14, wherein the specific binding to said PCSK9 is binding with a dissociation constant (Kd) of InM or less, eg, 100, 10 or IpM or less. 36. The method of any of clauses 1 to 15, wherein the human is at risk of or suffering from a condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 37. The method of clause 16, wherein step (c) treats or reduces the risk of said condition in the human. 38. The method of any one of clauses 1 to 17, wherein the human has been determined to comprise the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G in SEQ ID NO: 1 and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein encoded by the nucleotide sequence of SEQ ID NO: 29 or 30. 39. The method of any one of clauses 1 to 18, comprising the step of determining that the human comprises the nucleotide sequence that encodes a PCSK9 comprising a C-terminal domain comprising said mutation I474V or E670G and/or a proprotein convertase subtilisin/kexin type 9 (PCSK9) variant protein comprising said mutation 1474V or E670G, optionally, wherein the determining step is performed before administration of the antibody to the human. 40. The method of clause 19, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding the PCSK9 that comprises the C-terminal 285 domain comprising the mutation 1474V or E670G in SEQ ID NO: I. 41. The method of clause 20, wherein the assaying comprises contacting the biological sample with a. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides that can specifically hybridize to and identify in the biological sample a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation 1474V or E670G in SEQ ID NO: 1 or that specifically hybridizes to an antisense of said sequence, wherein said nucleic acid hybridizes to at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 or hybridizes to an antisense sequence thereby forming a complex when at least one nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and/or b. at least one oligonucleotide probe comprising a sequence of at least 10 contiguous nucleotides of a nucleotide sequence encoding the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises at least one nucleotide present in said selected sequence which is not present in SEQ ID NO: 28 thereby forming a complex when the nucleotide sequence encoding the PCSK9 that comprises a Cterminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the PCSK9 that comprises the C-terminal domain comprising the mutation I474V or E670G in SEQ ID NO: 1. 42. fhe method of clause 20 or 21, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 43. The method of any one of clauses 1 to 22, wherein said human is or has been further determined to be substantially resistant to statin treatment. 44. The method of any one of clauses 20 to 22, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 45. The method of any one of clauses 1 to 24, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474V or 286 E670G, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 28, or said human is indicated as homozygous for a nucleotide sequence encoding the PCSK9 C-terminal domain comprising said mutation 1474Vor E670G in SEQ ID NO: 1. 46. The method of any one of clauses 1 to 25, wherein said human has been diagnosed with at least one condition selected from a lipid disorder, hyperlipoproteinemia, hyperlipidemia, dyslipidemia, hypercholesterolemia, a heart attack, a stroke, coronary heart disease, atherosclerosis, peripheral vascular disease, claudication and high blood pressure. 47. The method of any one of clauses 1 to 26, wherein the nucleotide sequence is SEQ ID NO: 29 or 30. 48. The method of any one of clauses 1 to 27, wherein said ligand (eg, antibody or antibody fragment) is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 49. A ligand (eg, an antibody or fragment) for use in the method of any one of clauses 1 to 28, wherein the ligand specifically binds the PCSK9. 50. A kit comprising the ligand of clause 29 and instructions for carrying out the method of any one of clauses 1 to 28. id="p-1049" id="p-1049"
[001049] In an example of any aspect of the invention, the ligand (eg, antibody or fragment, eg, alirocumab, bocovizumab or evolocumab, or an antibody or fragment comprising the variable domains of 316P or 3 11-I4) is administered to the human at a two-weekly dose of from 75 to 150mg (eg, from 75 to 150mg administered once or twice over a two-week period). In an example, the ligand is for such administration to the human.
Determination of Specific Binding of Ligands of the Invention to PCSK9 Variants id="p-1050" id="p-1050"
[001050] Method of SPR Determination of Binding Binding of the antibodies to the PCSK9 variants was carried out by SPR using the ProteOn XPR36™ Array system (BioRad). An anti-human IgG surface (Jackson Labs 109-005-008) was created on a GLC Biosensor chip by primary amine coupling. Test antibodies were captured on this surface as ligands. The PCSK9 variants were used as analytes and passed over the captured antibodies at 256nM, 287 64ηΜ, 16ηΜ, 4ηΜ and InM. Binding curves were double referenced using a buffer injection (i.e. OnM) to remove baseline drift and injection artefacts. Regeneration of the capture surface was with lOOmM phosphoric acid which removed the captured antibody allowing another cycle of capture and binding. The binding sensorgrams generated were analysed using the 1:1 model inherent to the ProteOn XPR36 Array system analysis software. The assay was performed at 25 °C and using IxHBS-EP (Teknova) as running buffer. id="p-1051" id="p-1051"
[001051] Data Three antibodies were tested and the resulting binding data are presented below (Table 3). Antibodies 3 16P and 31H4 are antibodies disclosed in US20110065902 Al (the sequences of these antibodies and their variable domains are incorporated herein by reference for possible use in the present invention and possible inclusion in claims herein). Antibody 316P comprises heavy chain variable domains derived from recombination of human VH3-23*04 and JH2*01 (w ith a D), and light chain variable domains derived from recombination of human Vk4-1*01 and Jk2*01. id="p-1052" id="p-1052"
[001052] Evolocumab comprises a human IGHG2*01 heavy chain and a human IGLC2*01 lambda light chain, a VH derived from recombination of human IGHVl-18*01 and IGHJ6*01 (with a D segment) and a νλ derived from recombination of human IGLV2-14*01 and IGLJ2*01.
Table 3: SPR Determination of Ligand Binding Specificity for PCSK9 Variants Variant/Antibody ka(l/Ms) kd (1/s) KD (nM) PCSK9 a 316P 1.37E+06 2.75E-04 0.201 3IH4 1.I4E+06 6.38E-05 0.056 Evolocumab 1.14E+05 2.62E-05 0.229 PCSK9 a’ 316P 1.5OE+O6 2.72E-04 0.181 31H4 1.23E+06 6.06E-05 0.049 Evolocumab 1.24E+05 2.29E-05 0.185 PCSK9 c 316P 1.49E+06 2.75E-04 0.184 31H4 1.22E+06 5.69E-05 0.047 Evolocumab I.20E+05 2.20E-05 0.183 PCSK9 r 316P 1.40E+06 2.76E-04 0.197 31H4 L15E+06 5.82E-05 0.051 Evolocumab 1.16E+05 2.67E-05 0.230 PCSK9 f 316P 1.39E+06 2.82E-04 0.203 31H4 1.13E+06 5.95E-05 0.053 288 Evolocumab 1.16E+05 2.66E-05 0.229 PCSK9 p 316P 1.39E+06 2.73 E-04 0.196 31H4 1.14E+06 6.12E-05 0.054 Evolocumab 1.14E+05 2.50E-05 0.219 id="p-1053" id="p-1053"
[001053] Results The results showed that all antibodies tested bound to PCSK9 variants equally, with any binding variation seen being within experimental error for such a strong affinity interaction. id="p-1054" id="p-1054"
[001054] Thus, the invention determines that an antibody with the following profile can specifically bind one or more variants of the invention:- 1. An antibody (eg, 316P or alirocumab) that comprises heavy chain variable domains derived from recombination of human 1GHV3-23*O4 and IGHJH2*01 (with a D), and light chain variable domains derived from recombination of human IGKV4-P01 and IGKJ2*01; or 2. An antibody (eg, evolocumab) that comprises human heavy chain variable domains derived from recombination of human IGHV1-18*01 and IGHJ6*01 (with a D segment) and light chain variable domains derived from recombination of human IGLV2-14*01 and IGLJ2*01. id="p-1055" id="p-1055"
[001055] Thus, according to the invention, the skilled person is hereby provided with the required determination of specific binding of the ligand to PCSK9 variants. Applications of this determination are set out above in the context of methods and other aspects of the invention. id="p-1056" id="p-1056"
[001056] REFERENCES: Horton et al, Trends Biochem Sci. 2007 Feb;32(2):71-7. Epub 2007 Jan 9, Molecular biology of PCSK9: its role in LDL metabolism.
Seidah and Prat, J Mol Med (Berl). 2007 Jul;85(7):685-96. Epub 2007 Mar 10, The proprotein convertases are potential targets in the treatment of dyslipidemia.
Benjannet et al, J Biol Chern. 2004 Nov 19;279(47):48865-75. Epub 2004 Sep 9, NARC-1/PCSK9 and its natural mutants: zymogen cleavage and effects on the low density lipoprotein (LDL) receptor and LDL cholesterol.
Lagace et al, J Clin Invest. 2006 Nov; 116(1 1):2995-3005, Secreted PCSK9 decreases the number of LDL receptors in hepatocytes and in livers of parabiotic mice.
Maxwell et al, Proc Natl Acad Sci U S A. 2005 Feb 8;102(6):2069-74. Epub 2005 Jan 27, Overexpression of PCSK9 accelerates the degradation of the LDLR in a post-endoplasmic reticulum compartment. 289 Park et al, J Biol Chem. 2004 Nov 26;279(48):50630-8. Epub 2004 Sep 22, Post-transcriptional regulation of low density lipoprotein receptor protein by proprotein convertase subtilisin/kexin type 9a in mouse liver.
Rashid et al, Proc Natl Acad Sci USA. 2005 Apr 12; 102( 15):5374-9. Epub 2005 Apr 1, Decreased plasma cholesterol and hypersensitivity to statins in mice lacking Pcsk9.
Kotowski et al, Am J Hum Genet. 2006 Mar;78(3):410-22. Epub 2006 Jan 20. A spectrum of PCSK9 alleles contributes to plasma levels of low-density lipoprotein cholesterol.
Chen et al, J Am Coll Cardiol. 2005 May 17;45(10): 1611-9. Epub 2005 Apr 21, 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. Pisciotta et al, Atherosclerosis. 2006 Jun; 186(2):433-40. Epub 2005 Sep 23, Additive effect of mutations in LDLR and PCSK9 genes on the phenotype of familial hypercholesterolemia.
Zhao et al, Am J Hum Genet. 2006 Sep;79(3):514-23. Epub 2006 Jul 18, Molecular characterization of loss-of-function mutations in PCSK9 and identification of a compound heterozygote.
Seidah et al, Proc Natl Acad Sci USA. 2003 Feb 4; 100(3):928-3 3. Epub 2003 Jan 27, The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation.
Table 4: 1000 GENOMES PROJECT HUMAN POPULATIONS Below is a summary of the ethnic populations as per the 1000 Genomes Project sequences.
Population European ancestry Utah residents (CEPH) with Northern and Western European ancestry (CEU) Toscani in Italia (TSI) British from England and Scotland (GBR) Finnish from Finland (FIN) Iberian populations in Spain (IBS) East Asian ancestry Han Chinese in Beijing, China (CHB) Japanese in Toyko, Japan (JPT) Han Chinese South (CHS) 290 Population Chinese Dai in Xishuangbanna (CDX) Kinh in Ho Chi Minh City, Vietnam (KHV) Chinese in Denver, Colorado (CHD) (pilot 3 only) West African ancestry Yoruba in Ibadan, Nigeria (YRI) Luhya in Webuye, Kenya (LWK) Gambian in Western Division, The Gambia (GWD) Malawian in Blantyre, Malawi (MAB) West African Population (TBD) Americas African Ancestry in Southwest US (ASW) African American in Jackson, MS (AJM) African Caribbean in Barbados (ACB) Mexican Ancestry in Los Angeles, CA (MXL) Puerto Rican in Puerto Rico (PUR) Colombian in Medellin, Colombia (CLM) Peruvian in Lima, Peru (PEL) South Asian ancestry Ahom in the State of Assam, India Kayadtha in Calcutta, India Reddy in Hyderabad, India Maratha in Bombay, India Punjabi in Lahore, Pakistan 291 Table 5: TOIs & Related Diseases, Conditions and Example Ligands Human TOI Example Disease or Condition Example Ligand Il-17a Inflammatory Disease Uveitis Rheumatoid Arthritis Psoriasis AIN457 Ixekizumab Angiotensin II Receptor Type 1 (ATi) Hypertension LCZ696 Neprilysin Hypertension LCZ696 Metabotropic Glutamate Receptor 5 (Mglur5) Fragile X Syndrome AFQ056 Mavoglurant A Histone Deacetylase Cancer, Eg, Multiple Myeloma LBH589 Panobinostat Diacylglycerol acyltransferase-1 (DGAT-I) Familial Chylomicronaemia Syndrome LCQ908 Pradigastat Smoothened (Smo) Basal cell carcinoma Solid tumour Myelofibrosis Medulloblastoma Solid tumour LDE225 Smoothened (Smo) receptor Basal cell carcinoma Solid tumors LEQ506 ALK Non small cell lung carcinoma (NSCLC) LDK378 phosphatidylinositol-3-kinase (PI3K) mTOR AKT Cancer, eg, solid tumour, breast cancer, Renal cell carcinoma (RCC), Endometrial cancer, Nonsmall cell lung cancer (NSCLC), prostate cancer, Glioblastoma multiforme (GBM) BKM120 292 CRPC GIST Myelofibrosis mTOR PI3K ART Cancer, eg, solid tumour, breast cancer, Renal cell carcinoma (RCC), Endometrial cancer, Nonsmall cell lung cancer (NSCLC), prostate cancer, Glioblastoma multiforme (GBM) CRPC GIST Myelofibrosis BEZ235 ΡΙ3Κα mTOR PI3K AKT Cancer, eg, solid tumour, breast cancer, Renal cell carcinoma (RCC), Endometrial cancer, Nonsmall cell lung cancer (NSCLC), prostate cancer, Glioblastoma multiforme (GBM) CRPC GIST Myelofibrosis BYL719 Mitogen-activated ERK kinase 1 (MEK1) Mitogen-activated ERK kinase 2 (MEK2) Cancer, eg, solid tumour, melanoma, pancreatic, colon, lung or thyroid cancer Metastasis MEK162 protein kinase protein kinase C FLT-3 c-KIT Acute myeloid leukemia (AML) Myelodysplastic syndrome (MDS) Aggressive systemic mastocytosis (ASM) PKC412 Midostaurin 293 ActRIIB Sporadic inclusion body myositis (sIBM) BYM338 bimagrumab CD 19 • Cancer, eg, Leukaemia, mast cell leukemia, Acute Lymphoblastic Leukemia, Chronic lymphocytic leukemia (CLL) or Hematological tumors CTL019 (formerly CART19) i 11 β-hydroxylase Cushing’s syndrome LCI699 BRAF Cancer, eg, melanoma LGX818 Receptor Tyrosine Kinase FGFR • Cancer, eg, solid tumour • Breast Cancer • Endometrial cancer • Hepatocellular carcinoma • Renal cell carcinoma (RCC) • Bladder cancer • multiple myeloma (MM), hepatocellular carcinoma • endometrial cancer TKI258 (formerly CH1R258) Dovitinib DAC enzyme hematologic malignancy Multiple myeloma Myelodysplastic syndrome (MDS) Myelofibrosis LBH589 panobinostat HSP90 • Cancer, eg, Breast AUY922 294 Cancer, gastric cancer or Nonsmall cell lung cancer (NSCLC) • FGFR • Cancer, eg, solid tumour • Breast Cancer • Endometrial cancer • Hepatocellular carcinoma • Renal cell carcinoma (RCC) • Bladder cancer • multiple myeloma (MM), hepatocellular carcinoma • endometr ial cancer BGJ398 clAPl cIAP2 • Cancer, eg, solid tumour • Breast Cancer • Endometrial cancer • Hepatocellular carcinoma • Renal cell carcinoma (RCC) • Bladder cancer • multiple myeloma (MM), hepatocellular carcinoma • endometrial cancer LCLI6I Akt • Cancer, eg, Solid tumour, gastric cancer (eg, GDC-0068 RG7440 295 castration-resistant prostate cancer), prostate cancer, gastroesophageal junction cancer CD22 Hematologic malignancies Non-Hodgkin's lymphoma (eg, relapsed or refractory follicular non-Hodgkin’s lymphoma) Diffuse large B-cell lymphoma (eg, relapsed or refractory diffuse large Bcell lymphoma) DCDT2980S RG7593 CD79b Hematologic malignancies Non-Hodgkin’s lymphoma (eg, relapsed or refractory follicular non-Hodgkin's lymphoma) Diffuse large B-cell lymphoma (eg, relapsed or refractory diffuse large Bcell lymphoma) DCDS4501A RG7596 endothelin B receptor (ETBR) Melanoma, eg, metastatic or unresectable melanoma DEDN6526A RG7636 HER3 Cancer, eg, metastatic epithelial tumour, metastatic squamous cell carcinoma of the head and neck cancer, metastatic colorectal cancer MEHD7945A RG7597 EGFR Cancer, eg, metastatic epithelial tumour, metastatic squamous cell carcinoma of the head and MEHD7945A RG7597 Necitumumab 296 neck cancer, metastatic colorectal cancer MUC16 Cancer, eg, ovarian cancer (eg, platinum-resistant ovarian cancer) DMUC5754A RG7458 sodium-dependent phosphate transport protein 2b (NaPi2b) Cancer, eg, metastatic nonsquamous non small cell lung cancer, ovarian cancer (eg, platinum-resistant ovarian cancer) DNIB06O0A RG7599 PDL1 (programmed death ligand 1) Cancer, eg, solid tumour metastatic melanoma non-small cell lung cancer MPDL3280A RG7446 STEAP1 (six-transmembrane epithelial antigen of the prostate 1) Cancer, eg, prostate cancer (eg, metastatic castrationresistant prostate cancer) DSTP3086S RG7450 Bcl-2 Cancer, eg, leukaemia (eg, chronic lymphocytic leukaemia), non-Hodgkin's lymphoma GDC-0199 RG7601 Checkpoint kinase 1 (ChK l) Cancer, eg, solid tumour, refractory solid tumour or lymphoma GDC-0425 RG7602 ' G DC-05 75 RG774I mitogen activated protein kinase kinase (MAPKK) MEK Cancer, eg, solid tumour, melanoma (eg, metastatic melanoma) G DC-0973 RG7421 Cobimetinib GDC-0623 RG7420 epidermal growth factor domain-like-7 (EGFL7) Cancer, eg, colorectal cancer (eg, metastatic colorectal cancer), NSCLC Parsatuzumab MEGF0444A RG74I4 phosphatidylinositol-3-kinase (PI3K) Cancer, eg, prostate cancer, renal cell carcinoma, GDC-0032 RG7604 297 endometrial cancer, breast cancer (eg, HER2-negative metastatic breast cancer, metastatic hormone receptor-positive breast cancer), solid tumour GDC-0084 RG7666 Pictilisib GDC-0941 RG7321 mTOR TORC1 TORC2 PI3K Cancer, eg, prostate cancer, renal cell carcinoma, endometrial cancer, breast cancer (eg, HER2-negative metastatic breast cancer, metastatic hormone receptor-positive breast cancer), solid tumour GDC-0980 i RG7422 IL17 Autoimmune disease Ankylosing spondylitis; Asthma; Multiple myeloma; Multiple sclerosis; Polymyalgia rheumatica; Psoriasis; Psoriatic arthritis; Rheumatoid arthritis; Uveitis Secukinumab ixekizumab Ml prime segment of membrane IgE Allergic Asthma Quilizumab MEMP1972A RG7449 IFN alpha Autoimmune disease Systemic lupus erythematosus Rontalizumab PCSK9 (proprotein convertase subtilisin/kexin type 9) coronary heart disease (CHD) or high risk ofCHD hyperlipidaemia hypercholesterolaemia stroke atherosclerosis MPSK3I69A RG7652 298 a cardiovascular disease or condition a condition associated with elevated LDL Vascular Endothelial Growth Factor-A (VEGF-A) Placental Growth Factor (P1GF) Diabetic Macular Edema (DME), Branch Retinal Vein Occlusion (BRVO) Wet age-related macular degeneration (Wet AMD) EYLEA® Aflibercept A VASTIN™ LUCENTIS™ IL-6 receptor Inflammatory disease, eg, rheumatoid arthritis Uveitis (eg, non-infectious uveitis) Ankylosing spondylitis; Cancer Sarilumab REGN88 PCSK9 (proprotein convertase subtilisin/kexin type 9) coronary heart disease (CHD) or high risk ofCHD hyperlipidaemia hypercholesterolaemia stroke atherosclerosis a cardiovascular disease or condition a condition associated with elevated LDL Alirocumab REGN727 LY3015014 NGF Osteoarthritis Pain Fasinumab REGN475 IL-4 receptor alpha IL-13 receptor Allergic asthma eosinophilic asthma Atopic dermatitis Dupilumab REGN668 delta-like ligand-4 (D114) Cancer Enoticumab REGN421 angiopoietin-2 (Ang2) Cancer Nesvacumab REGN9W GDF8 Metabolic disorder REGN1033 299 cancer, obesity, diabetes, arthritis, multiple sclerosis, muscular dystrophy, amyotrophic lateral sclerosis, Parkinson’s disease, osteoporosis, osteoarthritis, osteopenia, metabolic syndromes (including, but not limited to diabetes, obesity, nutritional disorders, organ atrophy, chronic obstructive pulmonary disease and anorexia) Disuse muscle atrophy cancer-related cachexia LY2495655 ERBB3 Cancer REGN1400 angiopoietin -1 (Angl) angiopoietin -1 (Ang2) Cancer, eg, ovarian cancer AMG386 VEGF receptor PDGF receptor Stem cell factor receptor Cancer NSCLC Breast cancer Thyroid cancer Motesanib AMG 706 type 1 insulin-like growth factor receptor (1GF1R) Cancer Breast cancer Pancreatic cancer Ganitumab Linsitinib ASP7487 hepatocyte growth factor (HGF) Cancer Breast cancer Pancreatic cancer Rilotumumab HER3 ErbB3 Cancer Breast cancer Pancreatic cancer AMG 888 U3-1287 IL-17 receptor inflammatory disease Asthma Psoriasis AMG 827 brodalumab 300 sclerostin Bone-related condition postmenopausal osteoporosis fracture healing AMG 785 CDP7851 glucokinase Diabetes AMG 151 ARRY-403 PCSK9 (proprotein convertase subtilisin/kexin type 9) coronary heart disease (CHD) or high risk ofCHD hyperlipidaemia hypercholesterolaemia stroke atherosclerosis a cardiovascular disease or condition a condition associated with elevated LDL AMG 145 Evolocumab VLA2 Integrin α2β1 Inflammatory bowel disease SAR339658 IL-4 IL-13 Idiopathic pulmonary fibrosis SARI 56597 lysophosphatidic acid receptor LPA-1 LPA-3 Systemic sclerosis fibrosis SARI 00842 Androgen receptor Cancer, eg, prostate cancer, breast cancer MDV3100 enzalutamide HER1 EGFR Cancer, eg, NSCLC Erlotinib ERBITUX™ VECTIBIX™ VEGF receptor 1 VEGF receptor 2 VEGF receptor 3 Cancer, eg, colorectal cancer, breast cancer ASP4130 tivozanib JAK JAKI JAK2 Inflammatory disease, eg, rheumatoid arthritis Diabetic nephropathy ASP015K Baricitinib CD40 Prevention of organ transplant rejection ASP 1240 301 GnRH Endometriosis Cancer, eg, prostate cancer ASPI707 degarelix PDE9 Lower unirary tract symptoms associated with benign prostatic hyperplasia ASP4901 TNF alpha Inflammatory disease, eg, rheumatoid arthritis, psoriasis, chrohn’s disease, IBD Certolizumab pegol Programmed cell death protein J Cancer Chronic myelocytic leukemia; Hepatitis C virus infection; Hepatocellular carcinoma; Hodgkins disease; Melanoma; Multiple myeloma; NonHodgkin lymphoma; Nonsmall-cell lung cancer; Renal cell carcinoma; Solid tumor: Stage IV melanoma Nivolumab MK-3475 Hepatocyte growth factor MET Cancer Glioblastoma; Hepatocellular carcinoma; Metastatic colorectal cancer; Metastatic non small cell lung cancer; Metastatic stomach cancer; Non-small-cell lung cancer onartuzumab Angiopoietin ligand-1 Angiopoietin ligand-2 Tek tyrosine kinase receptor Breast tumor; Cancer; Colorectal tumor; Fallopian tube cancer; Gastrointestinal tumor; Glioblastoma; Hepatocellular carcinoma; Metastatic esophageal trebananib 302 cancer; Metastatic gastrointestinal cancer; Metastatic non small cell lung cancer; Metastatic ovary cancer; Metastatic renal cancer; Ovary tumor; Peritoneal tumor; Transitional cell carcinoma CD37 modulator Lymphocyte function antigen-3 receptor SLAM family member 7 Cancer Multiple myeloma elotuzumab IL-2 IL-2 receptor alpha Multiple sclerosis daclizumab EGFR Cancer Metastatic non small cell lung cancer; Solid tumor necitumumab IL-5 Asthma; Eosinophilic esophagitis reslizumab B-lymphocyte cell adhesion molecule CD22 Cancer, eg Acute lymphoblastic leukemia; Follicle center lymphoma; Non-Hodgkin lymphoma; Systemic lupus erythematosus, hairy cell leukaemia Inotuzumab inotuzumab ozogamicin epratuzumab moxetumomab moxetumomab pasudotox IL1 beta Acne vulgaris; Atherosclerosis; Behcets disease; Cardiovascular disease; Dermatomyositis; Insulin dependent diabetes; Multiple myeloma; Osteoarthritis; Paraproteinemia; gevokizumab 303 Polymyositis; Pyoderma gangrenosum; Scleritis; Uveitis CD20 Cancer Multiple sclerosis follicular lymphoma (eg, refractory or relapsed) diffuse large B cell lymphoma (eg, relapsed) chronic lymphocytic leukaemia (eg, first line therapy or relapsed) Oerel izumab ofatumumab IL-23 Crohns disease; Inflammatory disease; Psoriasis tildrakizumab BAFF Neutrokine alpha Autoimmune disease systemic lupus erythematosus Multiple myeloma vasculitis Belimumab Benlysta™ Tabalumab IL5 Asthma mepolizumab IL6 Inflammatory disease rheumatoid arthritis sirukumab Lp-PLA2 Atherosclerosis diabetic macular oedema darapladib CCR9 chemokine receptor Inflammatory disease Vercirnon 304 rheumatoid arthritis Crohn’s disease DOPA decarboxylase Parkinson’s Disease Patrome Her2 EGFR Cancer, eg, gastric cancer, breast cancer, head and neck squamous cell cancer, Tyverb™ Tykerb™ lapatinib A DP receptor Cardiovascular disease or condition Thrombosis, eg, arterial thrombosis Brilinta Brilique VEGFR EGFR Cancer, eg, medullary thyroid cancer Caprelsa LABA LAMA Respiratory disease or condition,eg, COPD PT003 GFF Factor Xa thromboembolism apixaban Oxelumab 0X40 ligand Asthma Graft-versus-host disease Tremelimumab Ticilimumab ipilimumab CTLA-4 CD 152 Cancer, eg, melanoma Autoimmune disease MPDL3280A MEDI4736 PD-L1 Cancer, eg, solid tumour, kidney cancer, lung cancer, melanoma, NSCLC, multiple myeloma Autoimmune disease Nivolumab AMP-514 AMP-224 PD-i Cancer, eg, solid tumour, kidney cancer, lung cancer, melanoma, NSCLC, multiple myeloma 305 Autoimmune disease LIGHT TNFSF14 CD40 ligand JAK Inflammatory disease, eg, rheumatoid arthritis, psoriasis, chrohn’s disease, IBD, ulcerative colitis, Psoriatic Arthritis tofacitinib Nerve Growth Factor Pain Osteoarthritis pain tanezumab Herl receptor Her2 receptor Her4 receptor Cancer, eg, Non-Small Cell Lung Cancer dacomitinib c-MET ALK Cancer, eg, Non-Small Cell Lung Cancer crizotinib Programmed cell death 1 receptor Cancer, eg, renal cell carcinoma Nivolumab SLAMF7 CD319 Cancer, eg, multiple myeloma Elotuzumab CD30 Cancer, eg, Hodgkin lymphoma, systemic anaplastic large cell lymphoma, T-cell lymphoma Brentuximab Brentuximab vedotin GPR40 DPP-4 Diabetes, eg, diabetes mellitus Fasiglifam trelagliptin VEFGR-1 receptor VEFGR-2 receptor VEFGR-3 receptor PDGFR cKit Cancer, eg, non-squamous non-small cell lung cancer Molesanib Motesanib diphosphate amyloid β Alzheimer's disease Solanezumab TNF alpha Inflammatory disease, eg, rheumatoid arthritis, psoriasis, chrohn’s disease, SIMPONI™ HDMIRA™ REMICADE™ 306 IBD, ulcerative colitis, Psoriatic Arthritis ENBREL™ Adalimumab IL-21 Autoimmune disease or condition Inflammatory disease or condition Rheumatoid arthritis Crohn’s disease IBD Ulcerative colitis Systemic lupus erythematosus (SLE) Graft versus host disease Cancer Metastatic melanoma Rena] cell carcinoma Melanoma Solid tumours Acute myeloid leukaemia Non-Hodgkin’s lymphoma Ovarian cancer Colorectal cancer Agonist or antagonist antibody specific for human IL-21 NNC0114-0005 NNC0114-0006 NN8828 ATR-107 Said or an anti-IL-21 antibody in combination with an agent selected from the group consisting of ipilimumab (eg, to treat melanoma), an anti-PDl antibody (eg, to treat solid tumours), sunitinib (eg, to treat renal cell carcinoma), rituximab (eg, to treat NonHodgkin’s lymphoma), sorafenib (eg, to treat renal cell carcinoma), doxorubicin (eg, to treat ovarian cancer) and cetuximab (eg, to treat colorectal cancer). 307 308 Table 6: PCSK9 309 310 in 51’9 520 HKPPVLRPRGQPNQCTGHREASIHASCCHAPGLECKVKEHGiPAPQzQVTVACEEGWTLTGCSALPGTSHVLGAY 670 AVDNTCVVRSRDVSTTGSTSEEAVTAVAICGRSRHLAQASQEL· sipwnleritppry'radeyqppdggsIvevylldtsrqsdhreiegrvmvtdfenvpeedgtrfhrqaskcJs thlagvvsgrdagvakgasmFshvIncqgkgtvsgtHglefirksqlvqpvgplvvIlplaggysrvlnaacqr^^ spasapevftvgatnaqdqpvtlgtlgtnfgrcvdlfapgedifgassdcstcfvsqsgtsqaaahvagiaamrnisaepeltlaelrqrkhfsak 425 445 474 dvineawfpedqrvltpnlvaalppsthGAGWQLFCRTVWSAHSGPTRMATAIARCAPDEELLSCSSFSRSGKRRGERM EAQG G KLVCRAH N AEG GEGVYAIARCCLLPQANCWHTAPPAEASMGTRVHCHQQGHVLTG CSSH WEVEDLGT 519 £20 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPQEQVTVACEEGVvTLTGCSALPGTSHVLGAY 570 AVDNTCVVRSRDVSrrGSTSEEAVTAVAICC.RSRHLAQASQ.ELQ 46 £3 MGΓ/S5R.x?Sl·Vl//PLPLLLtLLULGPAGARAQΞDEDGDYΈEL·VIiAiRSΞEEGLΆEAPEHGTTATEϊiRCAKDP ^RLPGTirrrCLKEETHiSGSERTARRIiC^GAARRGYLTKILH^HGLLPGFLVKMS^ VD YIE E DS S V EAQs ip w nl e r it p p ry ra d ey qp p dggs lv evyfrd t si q s d h reie g rv mvtd fen vpe edgtrf h r q a skcd shg thiagvvsgrdagvakgasmrsIrvincqgkgtvsgWglefirksqlvqpvgpivvIlplaggysrVnaacqrfaragw^ spasapeviWgatnaqdqpvtlgtlgtnfgrcvdlfapgedttgas5dcstcfvsqsgt5qaaah\ragi:aammlsaepettfaelrqrlW 425 4-9 474 dvineawfpedqrvltpntvaalppsthGAGWQLFCRTVWSAHSGPTRMATAVARCAPDEELLSCSSFSRSGKRRGBtM eaqggklvcrahnafggegwaiarccllpqancsvhtappaeasmgtrvhchqqghvltgcsshwevedlgt 619 620 HKPPVLRPRGQPNQCVGHREAStHASCCHAPGLECKVKEHGIPAPQEQ'/TVACEEGWTLTGCSALPGTSHVLGAY ' 672 AVDNTCVVRSRDVSTTGSTSEEAVTAVAICCRSRHLAQASQELQ QEDEDGDYEELVr7UjRSEEDGIjAEAPEBGTTATFH.RCAKDP ΉΑΣΡΟΤ±^~^\ΓΚΕΕΤΗΤ3Ο5ΕΚΤΑΤΙΡ^ΟΑΟΆΆΙΦ..ΟΥΣΤΚΙΣΉνΕΕΟ.Τ^ VDYiEEDSSVFAQsipwnierttppryradeyqppdggsIvevylldtsiqsdhreiegrvrwtdfenvpeedglrf^ thiagwsgrdagvakgasmrslrvlncqgkgtvsgtbgtefiYksqlvqpvgpivvilpiaggysrvlnaacqrlara.gvv^^ spasapevitvgatnaqdqpvtlgtlgtnfgrcvdliapgediigassdcstcfvsqsgtsqaaahvagiaammisaepeltiaelrqrHhfsak 42S ^-45 ^-74 Mature, form Pro-Form with Signal Sequence Pro-Form 311 •n dvineawfpedqrvltpnlvaalppsthGAGWQLFCRTVWSAHSGPTRMATAyARCAPDEEllSCSSFSRSGKRRGERM EAQGGKLVCRAHNAFGGEGWAI ARCCUP QANCSVHTAPPAEASMGTRVHCHQQ.GHVLTGCSSHWEVEDLGT £29 520 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPQEQVr/ACEEGmLTGCSALPGTSHVLGAY 670· AVDNTCVVRSRDVSKGSTSEEAVTAVAICCRSRHLAQASQELQ. s Fp vv η 1 e rit p pry ra dey q pp dggsivevylldts iq s dhre iegrvm vtdf envp eedgtrf hrqaskcds hg thFagvv5grdagvakgasmrslrvlncqgkgtvsgW:gJefirk.sqlvqpvgp!vvllpla^ spasapevftvgatnaqdqpvtlgtlgtnfgrcvdifapgediigassdcsttf^qsgtsqaaahvagiaammlsaepeltlaelrqrlrh'fsak 425 445 474 dvineawfpedqrvlipnlvaalppsthGAGWQLFCRTv'WSAHSGPTRMATAVA^ EAQGGKLVCRAHNAFGGEGVYAiARCCLLPQANCWHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 523520 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPQEQ\mA^ 670 AVDNTCVVRSRDV^TTGSTSEEAWAVAICCRSRHLAQA^^ 46 £3 1MGWSS'.x?RSmVPLPL£lLLLLUGPAGAi?AQEDEDGDYEELVLALRSEED&IAEAPEHGTTATFHRCAKDP W^PGTYtn^lF^ETHLSOSERTARRLC^QAAPRGYLTKILHWHGLiPGE^^ VDYIEEDSSVFAQsipwnleritppr/radeyqppdggsIvevylldtsiqsdhrefegrvm^ldfenvpeedgtrfhrqaskcdshg th]agvvsgrda.gvakgasrΊnrsir^r!ncqgkgtvsgtll·glefίΓksqlvqpvgplvvHplaggysrvl:naacqrlaragvvlvt:aagrΊfrddady spasapevrtvgatnaqdqpvtl.gtlgtnfgrcvdlfapgedrigassdcstcfvsqsgtsqaaahvagiaamnnlsaepeltlaelrqrlihfsak 425 443 474 dvineawfpedqrvltpnivaalppsthGAGWQLFCRTVWSAHSGPTRMATAIARCAPDEELLSCSSFSRSGKRRGERM EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANC5VHTAPPAEASMGTRVHCHQQGHVITGCSSHWEVEDLGT 519 620 HKPPVLRPRGQPNQCVGHREASmASCCHAPGLE'CKVKEHGIPAP^QVT^ACEEGWTLTGCSALPGTSHVLGAY 670 AVDNTCVVRSRDVSTTGSTSEGAVTAVAICCRSRHLAQASQELQ QEDEDGDYEEL‘\LALRSEEDGLAEAPEHGTTATFHRCAKDP . WRLPGTYVH/VLKEETHTSQSERTARRLCSAQAARRGYTTKTLEVFHGLLPGFLVKMSGDLLELALKIPH VDYiEED3SVFAQ5ipwnleritppryradeyqppdggslvevYlldtsiqsdhrefegrvmvtdfenvpeedgtrfhrqaskcdsh.g ^lagwsgrdagvakgasmrslrvlncqgkgt^gtliglefirksqlvqpvgplvvilpiaggysrvlnaacqrlaragvvl^ Mature form Pro-Form with Signal Sequence Pro-Form ‘S’ et u V, 312 mi s spasapevTvgatnaqdqpvtl-gtlgtnfgrcvdlfapgediigassdcstcfvsqsgtsqaaahvagiaammlsaepeltlaefrqrlihfsak 425 443 474 dvineawfpedqrvltpnlvaalppsthGAGWCUTCRTVWSAHSGPTRMATAIARCAPDEELLSCSSFSRSGKRRGERM EAQGGKLVCRAHNAFGOEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 522 52G HKPPVLRPRGClPNQC\/GΉR.EA.SIHASCCHAPGLECKVKEHGIP.APQEQVΓ’/ACEEG\^'TLTGCSAL·PGTSHVLGAY 573 AV D NTCV V R 5 RD VSTTGSTS EGA VTA VAICCRS RH LA QA S QELQ 1*« bid l·— u φ z q < T5 JC UJ O —J T5 ™ LJ 4 X > X 1 2 2 I > (J LG P O QO ra lg H in tip co yi (J p_ £ E Ώ x d Ή σ c uj ΰ $ xz u ra ul U tn ui ra ra Q 0 m ™ & α I w ? £ 5 4 o o fir y T 4 t S ra ,, > p· f 4 J f 3 - g g E §?ϊ » 9- o g ci. ts ¢4 X in IP § #1» Η δ 3 Oj ,½ ;= p U X E X5 r- Z: —J 0C _£z ω φ K < Φ ω 2 3 § δ S’ u ™ g 5 x y s 5 8 8 § S & δ ΰ y < gs s ξ > S’ 7 £ £ 5 f? g. torn a δ cc 2 Ilk'll £ § Hi ig § 1 £ ra * Z 2 > CD C ? J CL. £ Ig s $ -S > > p. C .'ti CL < CL < tnj > /- xz c£ (-. i j s w --1 8 — ζ Q. lZ, ro ° > T ¢7 i/n ω GJ ω CX ί ra a ’4 £1 σ a Q ΐ f S T 4 45 53 M G P/SSRRS W/PLPLLLUUUG PAG ARA QE DE DGDYEE LVXATRSE E DGIAE AP EHGTT ATFHRCAKD ? WPXPGT'’jTAWLKEETHDSQSERTARRDQA^AARRGYDTKITH^5FHGLLPGFrLVKMSGDIiETALKiPH vey i Ξ e ds s v faqs ip w nie rftp p ry ra day qpp dggs ivevyil dts iq s d h r eie g rvmvtdfen vpe ed gtrfh rq asked s hg thiagvvsgrdagvakgasmrslrvlncqgkgtvsgtliglefirksqlvqpvgplwllplaggysrvinaacqrlaragvvlvtaagnfrddacly s p a s a p ev rtvgat n a qd q pvtigt Igtnfgrcvd Ta pge drigas s d cst cfvs q sgts q a aa h va giaa m m Is a epe It 1a e kq rlihf sak 425 443 474 dv'neaudpedqrvltpnlvaalppsihGAGWQLFCRWWSAHSGPTRMATAyARCAPDEELLSCSSF^^ EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSH^^^ 623 520 HKPPVLRPRGQPNQCVGHREASmASCCHAPGLECK.VKEHGPAPQEQWVACEEGWLTGCSALPGTSHVLGAY 570 AVDNTCWRSRDVSTTGSTSEGAVTAVAICCRSRHLAQASQELQ ill A A A Lh CO § A O CU A A ω !i4 & & A Ω H *; 3 H O A Ρί in « to EH 3 Eh d Eh 0 g s M A CU iX § a p; 0 M Q CO Μ O N CO CQ A w A A 'Q L Ο H Ci O EU ill U ,-1 hl # O £ Mature form Pro-Form with Signal Sequence Pro-Form Ct - X- 313 rN r—1 rd "DY I EE DS s FAQs i p w nl e rit p p ry ra d ey qp p dggs Ivevyi Id ts i q sd h reiegrvmvt d f en v pe edgtrf h r q a skcd s hg ihlagwsgrdagvakgasmrslrvlncqgkgtvsgtliglefrrksqlvqpvgplvvllplaggysrvtnaacqrla^^ s pasa p evitvgatn a q dq pvt Igtlgtnfgrcvd If a pge diigass d cstcfvs qsgts q a a a h vagia a m m Isa e pe It la e 1 rq rlihf sak 425 443 474 dvineawfpedqn/ltpnMaalppsthGAGWQLFCRWWSAHSGPTRMATAyAR.^ EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGW^ 525 520 HKPPVLRPRGQPNQWGHREASIHASCCHAPGLECKVKEHGIPAPQEQVTVACEEG-WTLTGCSALPGTSHVLGAY 570 AVDNTCVVRSRDVSTTGSTSE^WAVAICCRSRHLA^ sip-wnteritppryradeyqppdggslvevyMtsiqsdhreiiegrvm'vtdfenvpeedgtrfhrqaskcdshg thlagwsgrdagva.kgasmrslrvi'ncqgkgtvsgtliglefirksqlvqpvgpi:wnplaggysrvlnaacqrtaragvvl.vtaagnfrddacly spasapevitvgatnaqdqpvtl:gtlgtnfjgrcvdlfapgediigassdcstcfvsqsgtsqaaahva.giaamml-saepeltlaelrqrti;hfsak 425 443 474 dvineawfpedqrvltpnlvaalppsthGAGWQLFCRTVWSAHSGPTRMATAyARCAPDEELLSCSSFSRSGKRRGERM EAQ.GGKLVCRAHNAFGGEGVYAfARCCLLPQANCSVHTAPPAEASMGTRVHCHQQ.GHVLTGCSSHWEVEDLGT 61? 522 HKPPVLRPRGQPNQ.CVGHRFA.SIH.ASCCHAPGLECKVKEHGIPAPQEQVTV'ACEEG'.VTLTGCSALPGTSHVLGAY 570 AVDNTCVVRSRDVSTTGSTSEGAXTAVAl^ 45 53 MGΓL·tSSRRSlVL·VrPL^£^ίLHLGPAGARAζ^DΞDGDYEELVLALRSEEEGLVEΛPΈmGTTATFHRCA^^ W RL P GT YWVL KE E T HLSQS E RTARRL QAQAAR^G YL-TKILH VFH GLLP G FL VKMS GDKLELALKL P H VDYiEEDSSVFAQsipwnleritppryradeyqppdggs-lvevylldtsiqsdhrei-egrvmvtdfenvpeedgtrfhrqa.skcdshg' thlagwsgrdagvakgasmrsIrvIncqgkgtogtlfglefirksqlvqpvgpWlplaggysrMnaacqrtaragwta^ 5 p a s a p ev rrvgat n a q d q pvtlgt Igtnfgr cvd If a pgedi iga s s d cstcfvs q sgts q a a a h vagi a a m m Is a e pe !t lae Irq rlihf sak 425 44? 474 dvineawfpedqr^tpnlvaalppsthGAGWQLFCRWWSAHSGPTRMATAyARCAPDEELLSCSSFSRS^^ EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 512 522 HKPPVLRPRGCtPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPQEClVn/ACEEGAJVTLTGCSALPGTSHVLGAY 570 AVDNTCWRSRDVSTTGSTSEEAWAVAICCRSR^ Mature form .......... Pro-Form with Signal Sequence 314 s in tH »D QEDEDGDYEELVO^RSEEDGLVE&PEHGTTATFHRCAKDP WRLPGTlX~v-LKEETHTSQSERTARRXC^LQAARRGYxTKIXHVFEGLLPGrLVWSGDI£LELAXKXPH VDYIEEDSSVFAQsipwmleritppryradeyqppdggsIvwylldtsiqsdhretegrvmvtdfenvpeedgtrfhrqaskcdshg thiagvvsgrdagvakgasmrslrvlncqgkgtvsgtliglefirksqivqpvgplvvllpiag^ spasapevitvgatnaqdqpvtlgtlgtnfgrcvdl’fapgedHgassdcstcfvsqsgtsqaaahvagiaammlsaepeltlaelrqrlihfsak ^25 443 474 dvineav^pedqrvltpnlvaalppsthGAGWCUFCRTVWSAHSGPTRMATAyARCAPDEELLS^ EAQGGKLVCRAHNAFGGEGVYAIARCGLLPC1ANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 529 623 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGWAPQEQ.VWACEEGV/TLTGCSALPGTSHVLGAY 570 AVDNTO/VRSRDVSTTGSTSEEAVTAVAICCRSRHLAQASQELCt 46 53 MGWSSKRSWWPLP£££U£.£aGPAGAR4QEDEDGDYEELVXAxRSEEDGI^EAPEEGTTATFHRCAKDP WRL ? GT YWVX KE E TEX SQS ERT^RL^QAARRG YX TKILHVFHGXLPGFX VWS GDXXE XAXKL P H VDYiEEDSSVFa.Qsfpwnlentppr^radeYqppdggslvevylfdtsiqsdhreiegrvmvtdfenvpeedgtrfhrqaskcdshg thlagwsgrdagvakgasmrsIrvIncqgkgtvsgtliglefTrksqivqpvgpIvvUplaggysrvInaacqrlaragvvMaagnfrddady spasapGVitvgatnaqdqpvtlgtlgtnfgrcvdlfapgediigassdcstcfvsqsgtsqaaahvagiaammlsaepelftaelrqrlrhfsak 425 443 474 dvineawfpedqivltpnlvatlppsthGAGWQLFCRTVWSAHSGPTRMATAIARCAPDEEL^ eaqggklvcrahnafggeo/yajarccllpqancsvhtappaeasmgtrvhchqqghvltgcsshwevedlgt 519 620 HKPPVLRPRGQPNQCVGHREASlHASCCHAPGLEClKVK.EHGtiPAPQECtVTv'ACEE.GVvTLTGCSALPGTSHVLGAY 670 AV D NTCV V RS R DVSTTG STS EEA VTA VAICCRS R H LAQA SQE LQ QEDEDGEYEELTYiALRSEEDGLAEAPEHGTTATFHRCAKDP WRX PGT YWXKE E THL SQSERTARRLQAQAARRGYXTKILH WEGLXP G EX VKMS GDXXEXAXKL PH VD YIE E DS 3 VFAQs ip w nl e r ftp p ry rad ey qp p dggs Vevyii dt s iq sd h reie grvmvt dien v pe edgtrf h rq a skcd s hg thlagwsgrdagvakgasmrslrvlncqgkgtvsgtliglefirksqlvqpvgplwilplaggysrvlnaacqrlaragvvlvtaagnfrddady 5 p a s a p e v it vgat n a q d q pvt Igt Igt nfgrcvd If a pge diiga s s d cstcfvs q sgts q a aa h vag ia a m m 1 s a e pe It 1a e kq rl iht sak 443 474 dvineawfpedqrvltpnlvatppsthGAGWQLFCRWWSAHSGPTRMATAARCAPDEELLSCSS^ EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 529 623 Pro-Form Pro-Form with Signal Sequence Pro-Form Cl £ E 315 ¢0 CFi rd VE^i^EDSSVFAQsipwnlemppryradeyqppdggsIvevTHdtsiqsdhreiBgrvmvtdfe^ thlagvvsgrdagvakgasmrslrvl'ncqgkgtvsgtligleffrksqlvqpvgpivvbplaggysrvlnaacqrlaragvvlvtaagnfrddacly spasapevitvgatnaqdqpvtigtlgtnfgrcvdlfapgedHgassdcstcfvsqsgtsqaaahvagiaammlsaepeftlae^ 425 445 474 dyi^eawfpedq^jtpnlyaalppsthGAGWQLFCRPA^SAHSG^^ E ο 4CD s c v_ O LU 0 k_ Cl E □ Lb g CL 316 cs CNI Fl 22 EAQGGKLVCKAHNAFGGEGVYAIARCCLLPQMCWHW 519 62-3 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAP^ 670 AVDNTCVVRSRDVSnGSTSEEAVTAVAICCRSRHLAQASQELQ sipwnleritppr/radeyqppdggslvevyidtsiqsdhreiegrvmvtdfenvpeedgtrfhrqaskcdshg th lagvvs grd agv a kga s m rs Irv In cqgk gtvsgtliglef irksqlvq p vgp Iv v 11 p 1 aggys rvi na acq ria ragvv 1 vt a agnfr d d ady spasapevitvgatnaqdqpvtl.gtlgtnfgrcvdtfapgedygassdcstcfvsqsgtsqaaahvagiaamml5aepeltl:aelrqrli:hfsak 42.6 —5 474 dvfseawfpedqrvlipnlvaalppsthGAGWQLFCRTW/SAHSGPTRMATAyARCAPDEELLSCSSFSRSGKRRGERM EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 519 620 HKPPVLRPRGQPNQCV'GHREASIHASCCHAPGLECKVKEhGIPAPQEQWACEEGV/FLTGCSALPGTSHVLGAY 670 AVDNTCVVRSRDVSTTGSTSEEAVTAVAICCRSRHLAQASQELQ 4£ 53 MGr/SSffRSUvWPtPiLi.aLaiGPAGAftX^DEDGDYEE.LVYL^JjRSEEDGK^PEHGTTAT^ WRL-PGTYv v '7L.KEETEiSQSERTARRi^QAARRGYLTKILEVFHGLL?GTiTTOGGDLLELAiKLPE "DYIE E D S SVFAQs i p w rd e ritp p ry r a d eyqp p dggs Iv evyl id ts i q s d h r eie g rv rwt dien v pe ed gtrf h r q a sk.cd s hg thlagvvsgrdagvakga5mrsfrvlncqgkgtvsgtliglefirksqlvqpvgplvvllplaggysrvlnaacqrl:aragvvlvta.agnfrddady spasapevitvgatnaqdqpvtlgtlgtnfgrcvdlfapgedhgassdcstcfvsqsgtsqaaahvagiaammlsaepeltlaelrqrlihfsak 425 443 474 dvl·neawfpedqr/kpnlvatlppsthGAGWQITCIRrΓVWSAHSGPTRM^^ EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEA.SMGTRVH€HC>Q.GHVLTGCSSHWEVEDLGT 619 520 Η KP P V LR P RG QP NQCVGH REAS I HAS CCH AP G IE CKVK EH G1P AP PIQVTVACEE G WTLTG CS ALP GTSH V LGAY 670 AVDNTCVVRSRDVSTTGSTSEEAXTAVAICCRSRHLA^ s·’ § -S ? σd ra m ΕΓ 3 « Μ & Ο > QJ crj J bo m g 8. c k. > <τ G S' ω tu 5 TO ra I Έ 1 ? Π no w gg s 1« £ f °· ru ό Lo S ΰ ij Jr H § -S: cr th krl l-M μί in CQ S TS T5 ii Iff y it ι u w cl ET £ A W Ή C fe £1 S fe CQ A G in V ^1 I d A E4 E CT w A S ϋ A A M A A [w W ί -2' ’ - m i CO UI > 03 no > KI Ό ID El N S φ A O A > Q A A M £.03 » H ft .TO S A A A (— rS CX £ :> £ Mature form Pro-Form with Signal Sequence Pro-Form cu 41 317 cn ΓΝ Ol 425 443 474 dvineawfpedqrudtpnlvatlppsthGAGWQLFCRTVWSAHSGPTRMATAiARCAPDEELLSCSSFSRSGKRRGERM EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTP^CHQQGHVLTGCSSHW 119523 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGWAPPEQWVACEEGWTLTGCSALPGTSHVLGAY 570 AV 0 N TC V V RS R DVSTTGSTS EEA VTA VAICCRS RH LAQA SQELQ. sipwnleritppryradeyqppdggsIvevyUdtsiqsdhreiegrvmvtdfenvpeedgtrfhrqaskcdshg thlagvvsgrdagvakgasmrstrvIncqgkgtvsgtligleflrksqlvqpvgp^vliplaggysiMnaacqrlarag^ spasapevitvgatnaqdqpvtl.gtlgtnfgrcvdlfapgedHgas5dcstcfv5qsgtsqaaahvagiaannmlsaepeltlaelrqrlj:hfsak 425 443 474 dvineawfpedqrvltpnlvatippsthGAG.WQLFCRWWSAHSGPTRMATAIARCAPDEEL^ EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 519 520 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPPEQ^^ 670 AVDNTCVVRSRDVSTTGSTSEEAVTAVAICCRSRHLAQA SQELQ 45 53 MGTV5SRRSL·V¼·'PLPLLLLLίLLLGPAGA QE DE DG DY E Ei^AL LS EE DGEAE APEHGT T ATFH RCAKD P WRLPGTlΎ4Γ:/-LΓEET?ΣLSQSERTΆRRLQ^QΆΆRRGYLTKILEWEGLLPGFLVKI4SGDIίL-ELJAL·KLPH v DYIE E DS s vfaqs ί p wnleritp pryradey qp pdggs h/'evyfldtsiq sd h reiegrvnwtdfen vpeedgtrf h rq ask cds hg Mature form Pro-Form with Signal Sequence Pro-Form 'o’ ‘o’ 318 26 27 ThlagvvsgrdagvakgasmrsliMncqgkgtvsgtltglefirksqlvqpvgpIvvilpiaggysrvInaacqrlarag^ spasapev ftvga tn a qd q pvt! gt 1 gtn fgrcvd l=f a pge diiga ss d cs t cf vsq sgts q a a a hvagi a a m m I sa e pe It la e 1 r q rl ihi sak 425 4-i3 474 dvineawfpedqwltpnlvaalppsthGAGWQLFCRTVWSAHSGPTRM EAQGGKLVCRAHNAFGGEGVYAJARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT 525 520 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPQEQVWACEEGV/TLTGCSALPGTSHVLGAY 570 AV DNTCVV RS RDVSTTGSTSEEA VTAVAtCCRSRHLAQASQELQ 45 55 Μ£π'55ΚΚ.5^/Ρ1Ρ£Είάωα6ΡΑ6ΑΚΑζ^ΒΕΒ6ΒΥΕΕΐνΒΑΕΕ·Ρ5ΕΕϋ3Βν^ WRLPGTY^rs^KEETHLSQSERTARRB^QAARRGYlTKIBHVFHGLLFGFLVmSGDIiELABKLPH VDYiEEcssvFAQslpwnieritppryradeyqpp-dggsIvevylldtsiqsdhreiegrvmvldfenvpeedgtrfhrqaskcdshg thiagvvsgrdagvakgasmrsIrvIncqgkgtvsgWgiefirksqfvqpvgpiwi^ spasapevitvgatnaqdqpvtigtlgtnfgrcvdlfapgediigassdcstcfvsqsgtsqaaahvagiaammisaepeltlaelrqrlihfsak 425 443 474 dvineavvtpedqrv'itpnlvaalppsthGAGWQLFCRWWSAHSGPTRMATAJARCAPDEELLSCSSFSRSGKRRGERM EAQGGKLVCRAHNAFGGEGVYAIARCCLLPQANCSVHTAPPAEASMGTRVHCHQQ^ 6as 620 HKPPVLRPRGQPNQ.WGHREASIHASCCHAPGLECKVKEHGIPAPQEQVrtX/ACEEGV/TLTGCSALPGTSHVLGAY 570 ' AVDNTCVVRSRDVSTTGSTSEGAVTAVAICCRSRH^ QEEEDGEYEELVLAIiRSEEDGIVEAPEHGTTATFHRCAKDP VDYlEEDSSVFAQsipVMmlerftppryradeyqppdggsIvevylidtsiqsdhreiegrvmvtdfenvpeedgtrforqaskcdshg thlagvvsgrdagvakgasmrslrvlncqgkgtvsgtliglefirk.sqlvqpvgpivvllpl'aggysrs4riaacqrfaragw^ spasapevitvgatnaqdqpvttgtigtnfgrcvd IfapgediigassdcstcfvsqsgtsqaaahvagraammlsaepeltlaelrqrliMsak 425 443 474- dvineawfpedqrvltpnlvaalppsthGAGWClLFCRTVWSAHSGPTRMATAIARCAPDEELLSCSSFSRSGKRRGERM EAQGGK.LVCRAHNAFGGEGVYAiARCCLLPQANCSVHTAPPAEASMGTRVHCHQQGHVLTGCSSHWEVEDLGT £23 520 HKPPVLRPRGQPNQCVGHREASIHASCCHAPGLECKVKEHGIPAPQFQVWACEFGWLTGCSALPGTSHVLGAY 570 AVDNTCVVRSRDVSTTGSTSEGAWAVAICGRSRHLAQASQEIQ Pro-Form with Signa!. Sequence _____ Pro-Form Cr cr 319 EH EH υ o 0 EH 0 0 Eh 0 0 Eh U 0 EH d EH CJ O EH u B o O υ EH O O EH o o 0 D 0 U 0 EH U 0 0 NUCLEOTIDE SEQUENCES 0 0 8 cd CD CD kJ CD CD CD kJ CD cp CJ 0 CJ 0 0 U o 0 U f< O 320 0 0 Eh ?l 01 < ID 0 3 o ID δ o c b o o 1= φ o ω < o rj 0 § 0 0 < ID 0 0 b o ID 0 b u u < l·0 CD ID < 0 0 t u 0 0 ID 0 ID b w < o w r*r UJ UJ cd (J o' I0 i— O b CD b g ? s 0 E? 0 3 95 P CD P P 0 0 0 δ ρ P P p lC Ε· 0 0 Ei U 0 0 fP! 0 Φ co; 0 0 3 8 8 b o P u P P P u P 0 0 0 CD Eh O 0 0 0 O 0 0 ώ 0 O O U 0 υ o 0 U CD CD 0 Ei 0 U 0 0 U u δ CJ u o CD ID δ P Π 0 Q KC Eh O 0 Fi CD δ CD O O CD o Ό EH Eh U EH 0 EH U O H δ 0 δ ο u 0 0 0 FI δ O U ω 0 Eh U 0 Q CD Eh υ O u 0 o o c 0 fl', EH y Ej 0 0 CD 0 &H 0 0 EH 0 Eh O o o 0 Eh lC ft ft bp ft b£i bp bp u 0 g Eh 0 A 0 Eh U O Eh U < 0 0 Eh < 0 d 0 U U E4 0 Eh O bp fto CD bo co a co cc bp do bp CP ft U E ftb ti bij bp bo PP ft PP & «3 Ln ftp Ct! cn bo CD bp 03 ω 03 8 S’ bo on bp re bo OP ft CD bp □3 S’ s 03 bp bp bo bo bp CD S’ ra ct bP ft DC ftj ep be ft op bt (J bO bp ft bp bp bp bo op β bp bp tip bp bti bp ftp bp to ft op CO ft gg ft CO ft ftp gg t; co bp GO & bo bp bp bn Ct ft bO ft ft co bp ft MJ bp bp S’ PP £ hp bp ra bo ft? GJ bp bp bp CP Ct bp ft CW 8 ft bp ft cv ft bp tuo bp ft 58 "& bp to p ^8 0 0 0 U 0 0 < co bp bp y 8 3 co CD bp 2 8 p &8" 8 3 bp 0 •k 0 3 >5 8 « En C 8 c ftb 03 ιΎΊ tw vf ^3 m £ δ < bp Ct' bo uo cn bo ft cu bp ft S 8 +H B cm bP g <0 w> ft ft ft 83 o bp bo too < CO ft bp bo CD bp SP to £P 8 ft H b& w s« co ♦O F bp < bp < ft 3 be bp 5 bn ft oo ft bo bl) bo ID b < u CD P < < ro CD bO cu §8 aj QC ft & be ft oj □0 bp ft CD 0 0 Lb cc CD CD CD b < P u ID Q < G u 0 CD E— CD O 0 < 0 b 0 Iδ CD P CD 321 ο cn CD < CD g r>J LD <0 Ch ci b e? 19 b p O < 8 0 o < 0 0 H CD Ω CD CO *.D 0 δ 0 u 0 0 b u δ 0 t9 g Eh Ei U U EH s 0 EH u O Ο P I— P Ό i5 Φ p P 8 p| < u U h0 o o o 0 0 δ t to δ o o Eh 0 0 o f4 0 u o 0 0 0 0 0 0 u 0 0 0 O U 0 0 o Ei 0 0 U U 0 CJ 0 0 EH 0 0 0 0 0 u 0 0 0 0 0 O o 0 0 ra ft ft Γη O Eh Ό 0 bo CD u u u o o CJ 0 0 CD Ο Eh 0 0 CD 0 0 0 fi! ra $ cu ϋ En EH 0 0 EH 0 0 EH 0 0 0 EH CJ A EH 0 0 Eh 0 ra ra 33 Π? s tj CD s ra bO ra bO ra ft ba to to ra 8 bo 3 O 0 u Ei 0 o Eh Ei 0 0 Eh 0 0 *ζ· 0 u 0 0 ft bO ra P bfl ra ft ft bO ra o 0 ΓΗ ft) 110 bO ra g§ ra 8) ft CD CD CJ ft bi) § 3> 85 o ω 8 bn an ft tj bO bo ra ra hD ft ba bp ft tW ra ft bp s ft ra ra ra QC ft ng ra E ra 3 QO ft bo ft. f ft ho ft ft ra □ 5P ω ftp ft bo CJ bO ra bo ra b) co bO CD CJ bO ft ru 85 S3 CD ra In no ft 0 < 0 u ft CJ bo bo bp ft ft) ft ra ft bp ra bp bp bQ ft bo ra up tnj P P P b P P § £ P o 0 (j U O ϋ Ο EH U 0 0 6 0 ϋ Ο 0 U 0 EH 0 & 0 0 0 U 0 0 0 Ei 0 d! o Eh Eh CJ U Eh Eh U d 0 EH o U U 0 a EH Eh 0 Eh U Eh 0 0 0 EH EH 0 O 0 EH 0 EH ft ω bn ra ra bp ft S b€ bo bO be ftj bo co bO £ cu CD bO tap ft u 3» no ft o R to u ra tio SB CD I? C < s 0 0 < C < 0 0 ba ra bp b) bo bp bO y. ra bO 4-» S £2 ft §2 s & M bi) ra bO CD bu ra ra CHJ bi) ra bO ft cj 3> CD ft uP bp ra bo CJ ft ba ra UO tui ft co ft ω bQ ft ω UP CD bo ft) ft ft bp ra uo bO 8 bi ft ft < U 0 P 0 ra uo ft bn ft be be CD CO bO to bi) ra ra ra οΰ ft 6 ra < 0 o o o 1= < P 0 P 0 < P b 0 ίο AGGGAGGCCAGCATCCACGCTrCCTGCTGCCATGCCCCAGGTCTGGAATGCAAAGTCAAGGAGCATGGAA Q52?PCAGto LEG E520G GAG to GGG ’ TCCCG G CCCCTCAGGAG GAG GTGACCGTGGCCTG CG AGG AG G GCTG GA CCCTG ACTG G CTG GAG TGCCCF CC 322 CJ CJ EH Bl I CTGGGACCTCCCACGTCCrGGGGGCCTACGCCGTAGACAACACGTGTGTAGTCAGGAGCCGGGACGTCAGCA 323 ΓΝ CT) ID IL1 CD b 0 CD 0 CJ CD ID CD CD Q CD CD 0 CD CD £ Eh EH 0 o 0 < U O U ΰ 0 H U W Ei B U 0 Eh CJ 0 Eh 0 0 CJ fit ft 0 0 3 0 o CJ Eh U U CJ Eh 0 Si '* 0 U EH CJ U o o ft 0 CD u CD CD < CD CD CD CD CD CD CD Q.
H 0 0 pt ft 0 ft o o 0 Eh 0 CJ 0 0 CD O 0 Eh Ei U o & 8 Ei O 0 ft 0 CJ Eh ft Eh Ei 0 O A Eh U 0 0 Ό 0 0 Eh ft :ccgtggaacctggagcggattacccctcca a ca ccagcatacagagtgaccaccgggaa a caga caggccagcaa gtgtgacagt cat 1 TO TO 8 tip tj TO TO cj t> tic $ no tlC tj CJ oe 8 8 S § 1 no S TO it tJ ro ί£ CJ bp Q (J no u no re ηδ S| ft L Λ B no < TO 3 IIP TA TO ΗΊ u ho no i Hl* O s° no » 8 Jk cj (D kJ 3 CJ < no H no «>8 B o TO 03 8 ο o CJ ci 1 I ' I I I I I 0 0 O 1 0 ; § S: 0 < 0 0 3 0 1 L £ b 0 0 g U u 0 < u CJ _ u = b b S 1 2 § 3 3 ° Η !t s 0 0 0 0 < 0 0 0 0 0 3 0 0 0 3 0 < 0 0 0 0 0 H 0 0 0 H0 < U 0 < 3 0 0 0 3 < 0 0 3 0 3 0 < 4 0 0 8 0 to & to 0 3 0 0 0 Eh Eh 8? " °P £ 6 8 £ po S 8 s to ω u £ to be iii TO TO <33 bO tun TO Εύ S ω u no s (J no 8 CJ no TO no ft TO ru TO TO 8 TO (J i zattggtgcctccagcg? no TO nil TO S’ ft 0 3 TO Jr 0 0) is no i— g) id no 0 8 TO. 3 o . H CJ 0 H ' CJ CD 3 3 u 0 0 0 O CJ 0 U 0 0 0 3 o b u 0 0 3 b u & (_> 8 8 ID 0 0 0 U 3 0 0 < ti < 0 H < 0 CJ □ EH 0 B Eh 0 U EH U 0 0 3 0 ft 0 g§ b? ro S tn o W S’g & 8 oii 1 sv §8 pnffi U-s|s8a· 8 TO η Η ω DS § pt cat caatgaggcctggtt ccct gagg _ _ _ _ _______ __ _ _ -___ u 3 ID 3 Ξ £ ω st U 12 □ CD b CD 3 0 < < § CJ CD 0 CD CD 0 b 0 < < 0 0 0 0 0 0 < < 8 0 0 < 0 0 4 0 0 0 0 s b 0 0 b 0 0 0 0 ci ID § 3 8 u £ u 0 1— 0 H 0 O 0 b 0 0 H U 0 0 ID J <; 0 U 0 < 0 3 8 P b 0 b 0 B 0 0 3 O EH m ft & no no TO CJ TO S’ no TO & 8 s t b 0 1— 3 8 0 0 0 ft U ft Eh TO TO bp TO tap to no no no no £ ft t5 bp ho tJ no no 1 TO 1 TO , 8 1 0 < u fe 0 0 o U 0 < U 0 0 0 b < 0 0 0 0 B 3 0 o ft 0 0 EH 0 Ei ft 0 bi) g> tiO bo S’ o TO f is a Sy S gg qij ra TO bo TO £ L no Lj s nn o to no tw cj to Q £/ o C nij to co to ηΰ no op tw atccacttctctgc u 0 u £ TO TO GJ '-r r- '1 s 3 3 < 0 < 0 u 0 0 0 0 ΰ 0 0 < u H 0 H 0 H < CJ 0 < H 0 o 4 0 0 < 0 0 1— ID 0 0 0 0 < 0 0 0 < TCCCG G CCCCTCAGGAGCAG GTGACCGTGGCCTG CG AGG AGG GCTGGACCCTGACTGG CTGCAGTGCCCTCC CTG G G AC CTC CCACGTC CTGG GGGCCT A CG CCG TAGACAACAC GTG TGTA GTCAG GAG CCG GGACG TCAGCA E6?3GGAGtaGGG _______ __________________ CTACAGGCAGCACCAGCGAAGAGGCCGTGACAGCCGTTGC CATC CC GCCGGAGCCGG CACCTGGCGCAGG 324 m ΠΊ I bO , i, J £ Q ra bo bO ra < ra 9P O $ ra H § ra fc & bp t G ft ί t? ra bo u u 3 bo bO bp G S Q bp CD Eh bO fc bp t; ύ b£> co s bo ft < bo UQ bp ra ra ra 888g ft Cl ra bD ω ρηΐ ^83 ΰ Si ij ra do E-i ra be uo 7? K 0 o ο υ ο ο bp ra & υ Π< bo co CO bo bp tJ CD no s υ bp 0 s bp ω o t bO bn U bp t GO ra s € bn Ed u 0 ft bo ra *7 S> 3 ω E> bp be bp to ra ft 85 no o bn s as 2P ft ί8 * bo co υ O Ei 0 0 0 ω fl 0 ώ ra 0 U nl 0 0 ra «κ §>& bp ω H ID 0 c bb ft bo ft £ CD ra bj G ra ω υ o 0 < 0 0 0 O U 0 0 Η EH 0 O ra ω bp ra 0 CD ri 0 0 U Eh U 0 bo ft G bp 0 fJ 0 0 ft bo L o ra O fX 0 O 0 $ bo s u bi) ra Eh H ri ra L -g 0 L· ri C < 03 s 0 o Η ¢.) 0 E^ Eh 0 bO bO Eh *ί g u a 0 bp ft w bti bO ra bp U Li) 0 Π) Eh 0 0 < 0 0 U < be 3 te < 13 δ b l·K b bo ’G G 0 c £ CJ 0 0 0 δ CD g δ 0 0 0 EH H 0 O 0 EH 0 0 EH 0 0 Eh 0 0 rtl 0 0 0 0 0 Ei U 0 0 hp be Ϊ5 8 u 0 0 0 l·0 0 c o ω ri o Ο ώ ri CD 0 U ri U 0 C 0 0 £ φ b £ ri S 8 0 0 CD O ri 0 o 0 0 0 Eh E< A 0 Eh 0 b e b to < IJ t b c < 0 0 1= U 0 0 g u o 0 U 0 EH U £ o cn CD 8? Ό u bo ft s ft bO S bp ra ftg 4^4 eZi CD Ο C 0 b ω b t £ g t S CD < o Eg g w Q o 0 ri U ri Ei CD ri 0 0 f= < 0 0 H 0 fl ft co b(5 t? bp G bo Ό bo ts bp bp <, 0 b 0 [2 £ 0 0 0 0 £ CD δ 0 0 s 325 c ra δ ϋ tLO tJ bQ CD w 0 bo 1j bp do do ba _ to e δΡ ™ u w nn QQ TO 0 0 H co Q Φ 0 b w Pi u P 0 P P 0 b u u £ CJ 3 Φ CJ P ri 0 TO bO bfj no no tj TO TO Q υ no OJO ? bO Q bp O P δ CD CJ 0 CJ 0 P υ CD u CJ i u CJ CJ 0 n< O ri CJ ri u ri u u 0 ri U ri O 0 O H CJ P CJ CJ ri o CJ ό fi Ci O fi (j P u u »i H ii P P u P O £ TO do bO d) TO dO dO U TO TO no u TO on tb TO no 3 bxi 8 S bO tb t? no tj Cj Cj 0 3 0 0 CJ 0 TO O nn TO £ ss o e P no TO bO bn bn O u ho ou do TO U.0 du o 15 u o CJ CD O CJ < CJ b o H CJ 5 CD ni P U 0 p O O B CJ 0 ii 0 £ Is p CJ o CJ o 3 3 b P π 8 o < CJ 0 u bO TO bo CD P U U P u u u 0 0 P cm b k9 b ™ % o JH no TO CD fi u u u 0 P P P u P 0 0 < 0 U u 0 £ 0 b 0 bp TO UP no CJ B U i 0 U u < 0 CJ CJ CJ < CJ Φ < 8 01 £ no TO to bp OO TO I bp TO dp TO O no ro ... bo? S 8^ u &p t ώ bn to ω ηδ ω co b 3 u ο bO fi TO no *i C5 ω 8P TO $ bJO 8§ & bo ϋ bC bp DO CD bn bn u| 0 u cd oo TO TO bo bq bg co no Pl ii p u bn co bo 0 B u o ri 0 CJ ri u ft TO & gg Έ ο W u Η H S3 I 3 S Eh U u Eh U U ri u 0 Eh U bo ΰ hr t! dp TO np b P b s o ri a cd do on bp S & 45 8 U u o o --0 CD Cj CJ CD & t> do g TO bO on P U U P P U CD TO bp TO 31 TO to do 61 to u by TO dO I TO a to b u b to tj β 0 < o o 0 »ΐ ci o CD o u $ P 0 0 a δ B O ΕΓλ UJ ’‘W Eg $ M & bp to 8 TO no 8 TO O do < t 3 ΰ C9 b cj o CJ t) u < U o 15 0 B U CJ 0 0 Q r\ 0 0 ii 0 0 υ r) O P P CD 0 Ai u o p 0 U U 0 u u u 0 P CJ i 0 u P fi CJ i P u < 0 o P 0 bO L & & TO u 0 t= TO OO bo bO U CJ a C9 CJ CJ 3 u CJ t O U CJ o o Ci Cj Cj σ) (9 O <5 < P 01 0 326 u ID LA [ ι ΰ υ U ra ra £0 α bo 4 Ο ϋ U 8 8>8 ® ϋ & £Ρ flJfijAFH m £C dp Hn =H i?« <-> S3? Bgsi alt? apsfs >h ri f h H Oho S R to Y' C M HO H RWRR ® M Sr ?) H / 1 8 W 3 W Η 1 SKS^iaaSfffsi hU ϋ ΰ F S & ra rt & 1 "ti OfClJ^lD^y £ 2P §J dr- S o h H 'J □ Ko nj ra S +- cu Ed o rj Ed p J u £, £p pp Λ $ ω ύ rtj cd 8 £,8 £. % ra ra tj O Ki υ Eh ra U £? M O Fi U 8 tt F 8 ra W 8 Ho OrjOlDboB^^R^o ra gg U 3 CJ H ra +j £a ™ 5 ¾ W u Θ 0 U m L y ω +g 0 π U o 3 " σ’ ™ sp£ go S ggu|8®^a£8s|a» ϋ Η ώ A Sa^Ol^Sl; O 0 0 A El S?S M 8) β ω O ppi? bO H s 8 3 g aS «8 sig Bt «S ra C ra q 8 b Ο <ζ m U ra jr 00 S ^-j LJ L> rn ra 25 IV i& bp O ra H u CSJ Ψ 0 £ y < 0 g 0 s £ s 5 ω 3 | S E y § l9 < u U φ Η o O P £ 8 § F a y S -° □ £ £ b J S § CD fe g O 0 < < < CJ ΰ 8 5 8 S g δ b § 3 ω O g 5 < u X U CJ rn CD CD H CD P b 8 53§g O (J 01 g U O ID fl ο o a 8 U Φ 0 O 3 3 b g υ < □ H 8 3 5 δ < U H CJ b 3 8 £ 2 8 w b h w W ω u a o u b S L> □ S5 4 F 8 ϋ < 3 g g 8 p gs < c5 CD < U H ω cd y ? 8 £ H CD CJ H 0) g id y a 3 3 s CD □ < < φ 3 8 3 b 8 y o ψ 0 p o ECD P CJ CJ to o XGTGACAGCC.GrTGCCATCTGCTGCCGGAGCCGGCACCTGGCGCAGG jj CD CD £ £ u s b € s s F CJ F u § g 8 CJ ϋ S2 P i? C CD r u E o 0 -T Ei o 0 Ed U CD EH CD ID Eh O CD ri; CD CD CD o B 3 E^ u a ω s u 8 οι ο cdI ID Ei U O 0 o ϋ ID CD CD ID 3 0 Ed U o o O HD 0 u CD CD EH Eh U O H CJ EH U CD CD § 8 CD U CD CD Ed HD O O cd 0 H O O Ed Ei o CD Ci C.i O EH Η CJ H Ed O Q U O LJ CD CD p? O U Ei O Ei O tfl CD O H CD Ei o ra 85 8 o co ra H ra ra ra op ra s 1 ra pos· ϊ pn to q ra u 0 £ & § E £ ΰ ο a? bp bb ra α Ή 3^85½^ & & B P iip tin ra θ no Ek " H ^3 B P 5? Il hn m bn m a cca actttggccgctgtgtggacctctttgccc 8 ί sn •c $ $ ra o ji & T5 bp bo ro v 8 +-» R co 88 ra bO I Rd । 2 1 ra , o 1 1 3 ! & 1 Sj I ra i ra . S ' £> 1 n 1 8; E ra o J u □ < u d o t □ ID s □ □ $ 8 o Ά P LJ ω CD u CD H o l·- CJ « CJ o CJ ω CD ί 31 21 CJ CD 3 S CJ 8 t CJ r n CD Li O 5 < o 8 δ §8 o t u < u cJ < CJ U CD CJ u CD < c ϋ HC CD b 3 8 b CJ 8 < CD CD CD P < o CD < CJ ΰ 5 u 3 u CJ 3 a CD 8 CJ CJ 3 CD < CD CD b b s U is t= u L? cd a < 8 H 2 CJ L.J 3 S C J CD ό < ω S CJ 0 CD oCD ™ is CD « CD CD < 8 CD CD < P < CD CD CJ E— CJ CJ CD CD U CJ CJ CJ CD CD CD cp CD b CJ 1— CD O < CJ CJ b CJ < CD CD P H w ό w < ID (J tp LU Φ 1 cdI 5 8 CD < U CJ 3 CD 3 (D CD < U < Ε- CTCCCAGGAGCTCCAGTGAC P P CJ P P δ Is P 2 P y 3 P P y 8 S y P y P u P P P P P CJ o o o o ra tib ra tub bo tuo bO is 8 < CJ E- CD CD < H CJ υ u P ή ο ο ο ο Ε-ι Η ο ο ο ο cd CD ΐΰ ω ο ο ο CD a ο Ο Ο ID Ed Ο Ο Φ ώ CD ω ω 327 O 0 U 0 0 Eh as Eh ra £ ra ra o DQ DC G) CO DO ra Si? Do DO ra ao G Ct DO tiO tip do E ra ra EH 0 EH 0 0 Eh no ω 0 $ O Eh DO DO G G DC to ra co DO ae ip □0 ra EH ίί O 0 EH 0 0 to tip □o ra G ra bo ra E. 0 Φ u 0 Eh O 0 0 Eh υ u ω u O 0 A 0 0 H A 0 tn 0 0 EH o DO G DO CO o 83 be DO G DO ra Do ft DO ro ra ω S i S fj <0 ho bo bu DO ra bO ra DO to CD W) ra ra tu) ra S5 G bo DO Eh A A 0 A 0 A 0 O EH up G ra ra DO G bO ra do ra ρη ao §8 88 G CO to CD ap S δ 1^8 ra do ap L DO .¾ & Dp S’ S’ S’ ra PP αύ do DO DO G to G bp ra DO ρ ο P G b ID P u δ < o b C b S p P o P P p P P P p < P P p P P P P P P < 0 σ P ra DO G ra (5 b < 8 p b c 0 fc J ra to & 83 og UO ac ra DO 0 p G do to G bo E. fi fe U O Eh δ r bOg E o 4-1 U„ ΜΊ ft DO ra (j ra $ ra t? bn < o !« P 0 P P 0 ly ra S’ °? Μ ω E M Of) DO Do Ο ΣΤ ra bo ra ra tw bij t? 00 fe ra u & ra QU ra ra 8 t? ra ra u Do at) ra Pl < O 0 0 P P P P ra U < 8 8 £ P P P P P P Pl 3 < c P < υ b l·s s b £ P £ 0 P £ 19 £2 ΰ 8 o Φ δ ts § 0 0 u P P £ *3 Cr ο 0 c; Ρ a u 0 0 0 O o A A ω o o 0 0 d (J 0 0 0 Fi 0 0 Q 0 i?1 A 0 O 0 o < 0 O O o o O ή EH O u u 0 H O 0 O A O o 0 A O 328 329 330 Table 7: Human VH3-23 Variant Alleles SNPs rs61750837 1 . rs61752504 rsl 055799 rs61752504 rs61750837 rsl064090 rsl055799 rsl064091 rsl064091 rsl 064090___ rs56069819 rs56069819 rs56069819 rs56069819 O\ 5 05 Ό o (Λ s— rs56069819 rs560698I9 Cumulative allele frequency oo O' o o 0.0087 0.0046 6000Ό 0.0005 0.0005 SOOO’O O VH3-23 haplotype o * rn cA > II cd Ό 2 (Z> u. TOTAL: 331 332 SEQ ID NOs comprising an anti-PCSK9 Patent or patent Pblication monoclonal antibody or fragment thereof wc MD rcm CM CM m CM MD ΓΟΟ ND oo -^F •^r CM CM o cm' Λ CM o MD CD Γ- Cd' 'e* NO CD Γ- CD r- tT r ’ o MD .. c r o MD rv r« r> v«— DF r—« ,-1 1 1 — ’xt . * r·· ’ cd CM Γ- On cd' CD CM o md MN MD o o Γ o o ON ON O o On ON cd Df NO OO cd' CD >—' ND OO '—1 - ex MD 9\ <-> «s MD r\ oo MD Γ- 00 MD >x r- r- CM f——< o CM ND' T}· o' MD MD oo ·> MD CD MD CM CM MD , o MD NO r. MD oo rMD . r CD NO o CM XT ON ND 00 o CM ON MD OO Tt cm' . tn DF 4N Tf cd' nd' CM . r MD -r r CD ND' o o rM df ND oo' VF ND r 00 ON CM MD ’Tt oo' NO OO — xt Γ— cd' o' CM ’— ^4? CD o' CM r—' > CM xF r- (< N© 00 CM Γ- r- ND OO ^f Λ MD r> — tT rx ΣΣ CM On Φ o CM ON o o 6 CM m δ ND oo CM δ CM CD kD δ NO oo cm' z n CM oo' rD z Q 00 md c< r- e- ^r z Q 1 < CM oo' CD ’M’ z q oo' MD r- ON r- Γ- ♦—* r·' o' Γ- cd' c- OO o' o' C-' cn r- ΟΟ o σ CM CD o CD r- Q CM CD o MD r- w o< MD' UJ no' r- ω On' NO rx U-J no' <- cd & MD r- (X? CD 1" MD QO u oo md' X on MD s J oo' md' X Dh ID md' o cd MD Γ— od P CD * MD Γ- 0 cd D< o 0 'dF ND o Q cd' ON CD 0 ^r' U -· cd -rF O MD Γ- o CD O MD c- 333 Patent or patent Pblication US20120020976A1 US20130085265A1 SEQ ID NOs comprising an anti-PCSK9 monoclonal antibody or fragment thereof CDRL SEQ ID NO: 5, 7, 9, 10, 12, 13, 15, 16, 17, 18, 19,20,21,22,23,24,26,28,30,31,32, 33, 35, 36, 37, 38, 39, 40, 42, 44, 46, 222, 229, 238, 405, 407, 409, 411, 413, 415, 417; CDRH SEQ ID NO: 47,48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 60, 62, 64, 65, 67, 69, 71, 72, 74, 76, 77, 78, 79, 80, 81, 83, 85, 87, 89, 91, 247, 256, 265, 404, 406, 408, 410,412, 414, 416; j CDRL SEQ ID NO: 5, 7,9, 10, 12, 13, 15, 16, 17, 18, 19,20,21,22,23,24, 26, 28,30,31,32, 33, 35, 36, 37, 38, 39, 40, 42, 44, 46, 405, 407, 409,411,413,415,417,461,465,485; CDRH SEQ ID NO: 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 60, 62,64, 65, 67, 69, 71,72, 74, 76, 77, 78, 79, 80, 81,83, 85, 87, 89, 91,404, 406, 408,410, 412,414, 416, 459, 463, 483: 334 SEQ ID NOs comprising an anti-PCSK9 Patent or patent Pblication monoclonal antibody or fragment thereof tn OX ci ω 00 CN ix o > * co 4Λ tn o' O . e. m VT tn 00 OO*' XO vr rd vt «X in xo Vt xo rd vf- vf- rx fs vf- Cd Cd vr XO Λ <*, vT cn o r> cd vr r- Cd o\ cd cn * oo cd cn vr o' r- cri rd cn vt <σ xo rv m »—I 00 tri vt f— cn Λ c\ cn o cn cn tn in rGO rxo tn Ο «X vr xo Γ» cd oo tn tn xo m tn tn tn οχ ox oo ΓΟΟ tn oo oo OO O' oo XO cd o Cd o cd rvr cn oo tr» in Cd Oi' O\ tn e X© ’-—I τ! rx vlr> CO ox rin ω GO oo r* o m A tn rtn cd vt cd cn cd rd o rd oo r* vr vt ed vt <σ Cd oo in rvT vT tn vfOx cd in in in OO vfOO Ox xo rxo tn vir' O' OO rs CO r. »n oo oo XO oo tn in tn tn in Vt tn oo r o© r> k e> tn rvt 335 SEQ ID NOs comprising an anti-PCSK9 Patent or patent Pblication monoclonal antibody or fragment thereof cm o CM cn cm c/l nr ri «X CM o CM rx CM o rc cn Γ- nr χό' m Γ'' m MT r> o in n rx o i/’) c r . r nf- ’—1 F *— »—· cn in m ci r- Ch m m CX CM Ch oo m m fs m in r\ rx o' o d' O\ o o . .X Ch σχ rx cn nt r * xo co m m xt in f 1 \D oo rn tn .X rx rx oo in rV rx xD QC d γ- 0© \D r^- ci CO o' m χο rx co ' ci CM rx in O in Ό r- 00 r. Ch in in ^- xo ΙΤϊ in m o CO d xO co o CM O\ \D 00o -nr r rx • c r\ r ο r c CM nr m xo ^r rq F* M m d' CM nt- co' nr \D r 00 ’—* nr oC CM xO oo o ίχ oo /- ►— ^r cn θ' • <\ CM »—< cn o' Λ-Χ CM CM nr xo 0© d CM 1 xo oo in ci d I— nr nT o' γο ’—1 nt uT ci Cb M- o d' δ CM cn in δ xo oo ci d CM m tn δ xo 00 cf z Q ,—· cm 0© nr z 0 οά un d z Q p~* CM 00 m Q oo tn Ch C' d r^ cn d oo o )—* o' cn 00 o O' CM m nr o in Γ- xt σ CM m O tn Γ- ω d d ω M2 d w Ch \D cd M2 Γ- GO cn f—« UO tn 0© GO r·^ cn F-^00 tn ΟΟ U ί» in nr X in \D O mJ 00 in zx tn fx xo o P 1 cn 2 in rx rn cd in rx 0 d o Q nf Ό O 0 f-^ cn Ch o 0 xo o O cn nr ο m r- 3- U —· cn ω m 336 SEQ ID NOs comprising an anti-PCSK9 Patent or patent Pblication monoclonal antibody or fragment thereof v· v O' oo in CO GO CO o m O o o m O CO G9 3 so CO r ro Cn CM r * o ‘91 ‘ CM .- in Γ »^· * V* o V· r V* ·—» I o co - δ cn cm r- O' oo in r\ CM O' tz^ V i-V in m r o o ..- o\ Os o o O' O' m ν’ in T—1 so OO cn ch rn V- τ—< SO oo OO in r Γ' so «X. ··. in - r, oo so V C- Γ- V 00 Ό so OO CH CO V- o' SO ΟΟ fX cm CM V- o -X Λ. in in r- O'. t' m r> « 50 V S© in in in SO V m so in o' CO V V· Q\ oo V o' CM V O' 00 V* ri r> > r v· r> so V* r CM V- o V· r' m . SO CM V \o oo 50 00 *—· O' CM V oo SO oo V V* o V ί' V- r. r< V m o CM . ~ 1 (—- cn o • CM >-----' «X CM Γ— r- 50 oo V CM V- Γ- SO oo in CM os V v- o o V in CM O' V V- o o V- o CM m O SO oo cm o CV m in O 5© oo cm ·—« oo z oo oC z F—< oo F < Z oo O' Q CM CO V 0. in r- r. r· 0 CM m V· 0 in r- r> V ί- O £**-* cn >—< r- oo o o t-x co ) < δ 00 Ο O' CM cn V O m Γ- T—< o CM m X σ r- V- ω O' so 0 so r< •X ω O' so 0 so δ Γ» GO 1— m OD in r- oo m V— CO m oo 0 00 in V □5 in so o V 0 oo in V- 0 m so o c£ cn 2 in Γ- I—« cn in r- r Q cn O' o 0 V v^ so o 0 m O' o 0 V v· SO o O ·— V- . o in V O cn V U in r- V 337 Patent or patent Pblication US20130245235A1 SEQ ID NOs comprising an anti-PCSK9 monoclonal antibody or fragment thereof CDRLSEQIDNO: 5, 7,9, 10, 12, 13, 15, 16, 17, 18, 19, 20,21,22, 23,24, 26,28,30,31,32, 33, 35,36, 37, 38, 39, 40, 42, 44, 46, 405, 407, 409,411,413,415,417; CDRH SEQ ID NO: 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 60, 62, 64, 65, 67, 69, 71, 72, 74, 76, 77, 78, 79, 80, 81,83, 85, 87, 89, 91,404, 406, 408,410,412,414,416; 338 Table 9: Human variable & constant variants distributed over several human ethnic populations - useful for ligand tailoring Cum 0.296 0.704 0.283 0.717 Hom Freq7 (Het + Hom freq8) 9600 (0.496) 0.504 (0.904) 0.104 (0.462) 0.538 (0.896) Het Freq6 0.400 0.400 0.358 \ 0.358 No. Individ s5 153 366 Human Populations4 < (European ancestry) < (European ancestry < (European ancestry) < (European Variant Nucleotide Position 14:106208086 | (forward strand) 14:106208086 (forward strand) 14:106208082 (forward strand) 14:106208082 (forward strand) Nucleotide Variation2 (NCB1 dbSNP reference number)3 GAT GAG (rsl045853) CTG 1— <1 Amino Acid Position as Variation 204D (CH3 variation) 204 E (CH3 variation) 206L (CH3 variation) 206M Example Human Allele1 IGHG1*O1 IGHGl*03 IGHGl*01 IGHGl*03 Human Gene Segment Type IGHG1 339 00 o ό 0.292 0.997 0.004 0.540 (0.876) 0.124 0.993 0.336 0.336 0.007 1 . 0.007 ancestry) CO DO < < 14:106110914 (forward strand) 14:106110914 (forward strand) 14:106110904 (forward strand) 14:106110904 (forward strand) cn LTJ rd t-H Cxi to rH tH Lh k_ — CCC ACC (rsll627594) AAC AGC (r$201590297) oi (CH3 variation) 72P (CHI variation) cz H x ·Β - 5 .2 CZ 2 X 5 £ ϋ x CO > 75S (CHI variation) 76F (CHI variation) IGHG2*01 IGHG2*02 IGHG2*01 IGHG2*04 IGHG2*01 IGHG2 340 0.711 0.289 CM _ CD m m στ - 2 oo m o o V O b b 0.539 (0.881) 0.118 (0.46) m nF cm σ? cd cn cd στ v io CD CD ό ό ό ό *—- oo r? CM CD o o o o mm ο o b b b g 0.342 0.342 CD Γ-> CD Ο νΗ Ο ό ό στ στ o rH O b ό CO CO Ο Q 14:10611013 (forward strand) 14:10611013 (forward strand) 14:106109752 (forward strand) 14:106109752 (forward strand) ΰΙ t= GTG ATG (rs8009156) GCC TCC (rs4983499) 76L (CHI variation) 161V (CH2 variation) 161M (CH2 variation) 257Α (CH3 variation) 257S (CH3 variation) IGHG2*04 IGHG2*01 IGHG2*O2 IGHG2*01 IGHG2*06 341 Table Footnotes: 1. IMGT notation (wwJmgt.org); refer to figures for other alleles comprising this variation. 2. SNP Underlined in Codon. z (Z) MD CD co ULI MD ND oo σϊ 342 Table 10: FURTHER SEQUENCES SE Q ID NO Human Allele Nucleotide/Amino Acid Sequence HEAVY CHAIN ALLELES 41 IGHGl*01 (CH1+Hinge+ CH2+CH3+CHS) gcctccaccaagggcccatcggtottccccctggcaccctcctccaaga gcacctctggg ggcacagcggccctgggctgcctggtcaaggactacttccccgaaccgg tgacggtgtcg tggaactcaggcgccctgaccagcggcgtgcacaccttcccggctgtcc tacagtcctca ggactctactccctcagcagcgtggtgaccgtgccctccagcagcttgg gcacccagacc tacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaaga aagttgagccc aaatcttgtgacaaaactcacacatgcccaccgtgcccagcacctgaac tcctgggggga ccgtcagtcttcctcttccccccaaaacccaaggacaccctcatgatct cccggacccct gaggtcacatgcgtggtggtggacgtgagccacgaagaccctgaggtca agttcaactgg tacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggagg agcagtacaac agcacgtaccgggtggtcagcgtcctcaccgtcctgcaccaggactggc tgaatggcaag gagtacaagtgcaaggtctccaacaaagccctcccagcccccatcgaga aaaccatctcc aaagccaaagggcagccccgagaaccacaggtgtacaccctgcccccat cccgggatgag ctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatc ccagcgacatc gccgtggagtgggagagcaatgggcagccggagaacaactacaagacca cgcctcccgtg ctggactccgacggctccttcttcctctacagcaagctcaccgtggaca agagcaggtgg cagcaggggaacgtcttctcatgctccgtgatgcatgaggctctgcaca accactacacg cagaagagcctctccctgtctccgggtaaa 42 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLQSS GLYS LS SWTVPS S S LGTQTYICNVNHKPSNTKVDKKVEPKS CDKTHTC PPCPAPELLGG PSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHN AKTKPREEQYN STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP QVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFL YSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLS PGK 343 Exposition 204 Exposition 206 43 IGHG2*01 (CH1+Hinge+ CH2+CH3+CHS) gcctccaccaagggcccatcggtcttccccctggcgccctgctccagga gcacctccgag agcacagccgccctgggctgcctggtcaaggactacttccccgaaccgg tgacggtgtcg tggaactcaggcgctctgaccagcggcgtgcacaccttcccagctgtcc tacagtcctca ggactctactccctcagcagcgtggtgaccgtgccctccagcaacttcg gcacccagacc tacacctgcaacgtagatcacaagcccagcaacaccaaggtggacaaga cagttgagogc aaatgttgtgtcgagtgcccaccgtgcccagcaccacctgtggcaggac cgtcagtcttc ctcttccccccaaaacccaaggacaccctcatgatctcccggacccctg aggtcacgtgc gtggtggtggacgtgagccacgaagaccccgaggtccagttcaactggt acgtggacggc gtggaggtgcataatgccaagacaaagccacgggaggagcagttcaaca gcacgttccgt gtggtcagcgtcctcaccgttgtgcaccaggactggctgaacggcaagg agtacaagtgc aaggtctccaacaaaggcctcccagcccccatcgagaaaaccatctcca aaaccaaaggg cagccccgagaaccacaggtgtacaccctgcccccatcccgggaggaga tgaccaagaac caggtcagcctgacctgcctggtcaaaggcttotaccccagcgacatcg ccgtggagtgg gagagcaatgggcagccggagaacaactaoaagaccacacctcccatgc tggactccgac ggctccttcttcctctacagcaagctcaccgtggacaagagcaggtggc agcaggggaac gtcttctcatgctccgtgatgcatgaggctctgcacaaccactacacgc agaagagcctc tccctgtctccgggtaaa 44 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLQSS GLYSLSSWTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPC PAPPVAGPSVF LFPPKPKDTLMISRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKTK PREEQFNSTFR WSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYT LPPSREEMTKN QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKL TVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK Exposition 72 imposition 75 F=position 76 V=position 161 A=position 257 344 45 IGHVl-18*01 caggttcagctggtgcagtctggagctgaggtgaagaagcctggggcct cagtgaaggtc tcctgcaaggcttctggttacacctttaccagctatggtatcagctggg tgcgacaggcc cctggacaagggcttgagtggatgggatggatcagcgcttacaatggta acacaaactat gcacagaagctccagggcagagtcaccatgaccacagacacatccacga gcacagcctac atggagctgaggagcctgagatctgacgacacggccgtgtattactgtg cgagaga 46 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAP GQGLEWMGWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDD TAVYYCAR 47 IGHV1-46*01 caggtgcagctggtgcagtctggggctgaggtgaagaagcctggggcct cagtgaaggtt tcctgcaaggcatctggatacaccttcaccagctactatatgcactggg tgcgacaggcc cctggacaagggcttgagtggatgggaataatcaaccctagtggtggta gcacaagctac gcacagaagttccagggcagagtcaccatgaccagggacacgtccacga gcacagtctac atggagctgagcagcctgagatctgaggacacggccgtgtattactgtg cgagaga 48 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAP GQGLEWMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSED TAVYYCAR LIGHT CHAIN ALLELES 49 IGKC*01 cgaactgtggctgcaccatctgtcttcatcttcccgccatctgatgage agttgaaatct ggaactgcctctgttgtgtgcctgctgaataacttctatcccagagagg ccaaagtacag tggaaggtggataacgccctccaatcgggtaactcccaggagagtgtca cagagcaggac agcaaggacagcacctacagcctcagcagcaccctgacgctgagcaaag cagactacgag aaacacaaagtctacgcctgcgaagtcacccatcagggcctgagctcgc ccgtcacaaag agcttcaacaggggagagtgt 50 RTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQD SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC V=position 84 C=position 87 345 51 IGLC2*01 ggtcagcccaaggctgccccctcggtcactctgttcccgccctcctctg aggagcttcaa gccaacaaggccacactggtgtgtctcataagtgacttctacccgggag ccgtgacagtg gcttggaaagcagatagcagccccgtcaaggcgggagtggagaccacca caccctccaaa caaagcaacaacaagtacgcggccagcagctatctgagcctgacgcctg agcagtggaag tcccacagaagctacagctgccaggtcacgcatgaagggagcaccgtgg agaagacagtg gcccctacagaatgttca 52 GQPKAAPSVTLFPPSSEELQANKATLVCLTSDFYPGAVTVAWKADSSPV KAGVETTTPSK QSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS 53 IGKV4~l*01 atggtgttgcagacccaggtcttcatttctctgttgctctggatctctg gtgcctacggg gacatcgtgatgacccagtctccagactccctggctgtgtctctgggcg aga-gggccacc atcaactgcaagtccagccagagtgttttatacagctccaacaataaga actacttagct tggtaccagcagaaaccaggacagcctcctaagctgetcatttactggg catctacccgg gaatccggggtccctgaccgattcagtggcagcgggtctgggacagatt tcactctcacc atcagcagcctgcaggctgaagatgtggcagtttattactgtcagcaat attatagtact cctcc 54 DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLA WYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDV AVYYCQQYYST P 346 55 IGKV1-13*O2 atggacatgagggtccccgctcagctcctggggcttctgctgctctggc tcccagcaggt gccagatgtgccatccagttgacccagtctccatcctccctgtctgcat ctgtaggagac agagtcaccatcacttgccgggcaagtcagggcattagcagtgctttag cctggtatcag cagaaaccagggaaagctcctaagctcctgatctatgatgcctccagtt tggaaagtggg gtcccatcaaggttcagcggcagtggatctgggacagatttcactctca ccatcagcagc ctgcagcctgaagattttgcaacttattactgtcaacagtttaatagtt accctcagtgc cagatgtgccatccagttgacccagtctcoatcotccctgtctgcatct gtaggagacag agtcaccatcacttgccgggcaagtcagggcattagcagtgctttagcc tggtatcagca gaaaccagggaaagctcctaagctcctgatctatgatgcctccagtttg gaaagtggggt cccatcaaggttcagcggcagtggatctgggacagatttcactctcacc atcagcagcct gcagcctgaagattttgcaacttattactgtcaacagtttaatagttac cctca 57 IGKJ2*01 tgtacacttttggccaggggaccaagctggagatcaaac 58 YTFGQGTKLEIK 59 IGLJ2*01 tgtggtattcggcggagggaccaagctgaccgtcctag 60 VVFGGGTKLTVL 61 An IGHGl*01 Heavy Chain Constant Region ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKV EPKSCDKTHTCPPCPAPELLGGPS VFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAK TKPREEQYNST YRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV YTLPPSRDELT KNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRWQQGNVFS CSVMHEALHNHYTQKS LS LS PG 62 An IGKC*01 Kappa Light Chain Constant Region RTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV TKSFNRGEC 63 An IGHG2*01 Heavy Chain Constant Region ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTV ERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRWSVLTWHQDWLNGK EYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKS RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 64 An IGLC2*01 Lambda QPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVK AGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKT 347 Light Chain Constant Region VAPTECS 65 An IGHG2*01 Heavy Chain Constant Region ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLQSSGLYSLSSWTVPSSNFGTQTYTCNVDHKPSNTKVDKTV ERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCWVDVSH EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRWSVLTWHQDWLNGK EYKCKVSNKGLPSSIEKTISKTKGQPREPQVYTL>PPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKS RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 66 An IGKC*01 Kappa Light Chain Constant Region RTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV TKSFNRGEC 348 Example 2: Rarer IL6R Variants id="p-1057" id="p-1057"
[001057] The present invention provides anti-IL6R ligands; and IL6R-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 IL6R, in particular human IL6R or its ligands and in screening assays to identify other antagonists of IL6R activity. Some of the ligands ofthe invention are useful for inhibiting binding of IL6R to JL6 and/or gp 130, or inhibiting IL6R-mediated activities. id="p-1058" id="p-1058"
[001058] Anti-IL6R ligands (eg, antibodies and anti-sense RNA) have been developed based on targeting and neutralising so-called "wild-type" human IL6R, which is a commonlyoccurring form (see, eg, SEQ ID NO: 78). While such therapies are useful for human patients harbouring this form of human IL6R, the inventor considered it useful to investigate the possibility of targeting rarer - but still naturally-occurring - forms of IL6R amongst human populations. In this way, the inventor arrived at insight into the natural occurrences and distributions of rarer human IL6R forms that can serve as useful targets (at the protein or nucleic acid level) for human treatment, prophylaxis and diagnosis pertinent to diseases and conditions mediated or associated with IL6R activity. This particularly provides for tailored therapies, prophylaxis and diagnosis in humans that are devoid of the common IL6R gene or protein. id="p-1059" id="p-1059"
[001059] The skilled person will know that SNPs or other changes that translate into amino acid variation can cause variability in activity and/or conformation 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 more effectively tailor medicines and diagnosis of patients. The invention, therefore, provides for tailored pharmaceuticals and testing that specifically addresses rarer IL6R polymorphic variant forms. Such forms or "alleles" (at the nucleotide level), comprise one or more changes at the nucleotide and amino acid levels from the corresponding common form nucleotide and amino acids sequences, ie, there are one or more non-synonymous changes at the nucleotide level that translate into one or more corresponding changes in the protein target in humans. [001060] Furthermore, the inventor surprisingly realised that the rarer natural forms, although present in humans at much lower frequencies than the common form, nevertheless are represented in multiple and ethnically-diverse human populations and usually with many human examples per represented ethnic population. Thus, the inventor realised that targeting such rarer forms would provide for effective treatment, prophylaxis or diagnosis across many human ethnic populations, thereby extending the utility of the present invention. id="p-1061" id="p-1061"
[001061] With this realisation, the inventor realised that there is significant industrial and medical application for the invention in terms of guiding the choice of anti-IL6R ligand for administration to human parents for*therapy and/or prophylaxis of IL6R-mediated or associated 349 diseases or conditions. Tn this way, the patient receives drugs and ligands that are tailored to their needs - as determined by the patient’s genetic or phenotypic makeup. Hand-in-hand with this, 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. [001062] In developing this thinking, in this non-limiting example the present inventor decided to determine a set of human IL6R 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:- • Naturally-occurring human IL6R variation having a cumulative human allele frequency of 35% or less; • Naturally-occurring human 1L6R variation having a total human genotype frequency of about 50% or less; • Naturally-occurring human IL6R variation found in many different human ethnic populations (using the standard categorisation of the 1000 Genomes Project; see Table 12 below); and • Naturally-occurring human IL6R variation found in many individuals distributed across such many different ethnic populations. {001063] On the basis of these criteria, the inventor identified the variants listed in Table 11 below. The inventor's selection included, as a consideration, selection for nucleotide variation that produced amino acid variation in corresponding 1L6R forms (ie, non-synonymous variations), as opposed to silent variations that do not alter amino acid residues in the target protein. 350 Table 11: Human IL6R variants distributed over several human ethnic populations & genotype frequency Cum Freq6 089Ό o ΓΝ ό 0.947 0.053 Hom Freq4 (Het + Hom freq5) 0.489 (0.872) 0.128 (0.511) 0.904 (0.99) 0.010 (0.096) Het Freq3 0.383 0.383 9800 ID OO o ό having desired total human ; (a) Amino acid variability, population distributions and frequencies: Exon 9 No. Unique Pops2 3 No. Individs1 557 1 SOT Human Populations YRI,ASW,GBR,TSI, CLM,CHB,LWK,CHS, MXL,PUR,J PT,IBS,FIN, CEU (See note 7) LWK,ASW,YRI,PUR (see note 8) 358D 1 1 385V 3851 Most common Variant Most common , Variant 351 352 fb) Nucleotide Sequence Variations of Selected Alleles o c 4—* o o u.
CD cn CA TO Nucleotide change (compared to most common allele) giving rise to an amino acid change in the variant form (compared to most common allele); and NCBI dbSNP reference number (NCBI dbSNP Build 138 released on Apr 25, 2013).
CJ 353 id="p-1064" id="p-1064"
[001064] Example 3: Tailoring Antibodies to Rarer IL6R Variant Profile id="p-1065" id="p-1065"
[001065] As outlined above, the invention includes the possibility to tailor treatment of humans further by selecting antibody-based ligands with variable and/or constant domains based on gene segments found in many humans of the ethnic populations where the variant IL6R forms are found to meet the selection criteria of the invention. An example is provided for ligands comprising antibody VH domains derived from recombination of human IGHV gene segments comprising selected nucleotides at positions in the HCDR1 or FW3 where there is variability in humans (ie, where SNPs occur in humans). id="p-1066" id="p-1066"
[001066] The inventor analysed human IGHV variation and used this to choose ligands based on human IGHV alleles comprising said selected nucleotides and for matching to human recipient genotypes and/or phenotypes. The inventor identified utility in using gene VH gene segments encoding (i) a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109 and wherein said human comprises a VH gene segment encoding a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109, or the human expresses VH domains that comprise a CDR1 comprising a Phe at position 4 shown in SEQ ID NO: 109; or (ii) a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111 and wherein said human comprises a VH gene segment encoding a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111 or the human expresses VH domains that comprise a FW3 comprising a Thr at position 33 shown in SEQ ID NO: 111. Further information is provided in Table 14, which shows variation at these positions, as well as the variant distributions across the 1000 Genomes Project database relating to many human populations. id="p-1067" id="p-1067"
[001067] In other embodiments, as explained more fully above, the invention provides for ligands which are tailored to the human recipient’s genotype and/or phenotype based on alternative human VH gene segments, or on Vk, VZ or constant region gene segments (see further Table 14 for representative variants). id="p-1068" id="p-1068"
[001068] In an example, following this guidance, the chosen ligand can be sarilumab. [0010691 Further examples, therefore are:- (i) wherein 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*O1, a human D gene segment and a human JH segment. (ii) wherein the ligand comprises a Vk domain derived from the recombination of human Vk segment IGKV1-12*O1 and a human Jk segment, and wherein said human comprises a IGKVI-12*0I Vk gene segment or the human expresses Vk domains derived from the recombination of human Vk segment IGKV 1-12*01 and a human Jk segment. 354 (iii) wherein 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: 113 and wherein said human comprises a Vk gene segment encoding a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113, or the human expresses Vk domains that comprise a CDR3 comprising a Pro at position 7 shown in SEQ ID NO: 113; or (ii) a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 and wherein said human comprises a Vk gene segment encoding a FW3 comprising a Ser at position 15 shown in SEQ ID NO: 115 or the human expresses Vk domains that comprise a FW3 comprising a Ser at position 1 5 shown in SEQ ID NO: 115. (iv) wherein the ligand comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHGl*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: 81 or a Leu at position 206 shown in SEQ ID NO: 81. (v) wherein 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: 83, an Asn at position 75 shown in SEQ ID NO: 83, a Phe at position 76 shown in SEQ ID NO: 83, a Val at position 161 shown in SEQ ]D NO: 83 and an Ala at position 257 shown in SEQ ID NO: 83 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: 83, Asn at position 75 shown in SEQ ID NO: 83, Phe at position 76 shown in SEQ ID NO: 83, Val at position 161 shown in SEQ ID NO: 83 or Ala at position 257 shown in SEQ ID NO: 83. (vi) wherein the ligand comprises a human kappa chain constant region that comprises a Val at position 84 shown in SEQ ID NO: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and wherein said human comprises (i) an IGKCI*01 human heavy 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: 93 or a Cys at position 87 shown in SEQ ID NO: 93 and (ii) a nucleotide sequence encoding said 1L6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78. (vii) wherein the ligand comprises a human IGLCl*01 lambda chain constant region and wherein said human comprises (i) a human 1GLC1 *01 lambda chain constant region gene segment, or the 355 human expresses antibodies comprising human JGLC1 *01 lambda chain constant regions. id="p-1070" id="p-1070"
[001070] For example, as per example (iv), the inventor identified the possibility of addressing the rarer IGH-gamma-1 SNPs 204D (observed cumulative frequency of 0.296) and 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. Thus, in this example the ligand of the invention comprises or consists of an antibody that comprises a human gamma-1 heavy chain constant region that comprises an Asp corresponding to position 204 of SEQ ID NO: 81 or a Leu corresponding to position 206 of SEQ ID NO: 81 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. An example of such a ligand is sarilumab. id="p-1071" id="p-1071"
[001071] In another example, as per example (v), the inventor identified the possibility of addressing IGH-gamma-2 SNPs. This included consideration of Fc region variation - in this respect, the inventor focused on positions 161 and 257 which are in the Fc region. Thus, in this example the ligand of the invention comprises or consists of an antibody that 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: 83, an Asn corresponding to position 75 of SEQ ID NO: 83, a Phe corresponding to position 76 of SEQ ID NO: 83, a Val corresponding to position 161 of SEQ ID NO: 83 and an Ala corresponding to position 257 of SEQ ID NO: 83; 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. id="p-1072" id="p-1072"
[001072] In another example, as per example (vi), the inventor addressed human kappa constant region variation. Thus, in this example the ligand of the invention comprises or consists of an antibody that comprises a human kappa light chain constant region that comprises a Val corresponding to position 84 of SEQ ID NO: 93 or a Cys corresponding to position 87 of SEQ ID NO: 93; 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. An example of such a ligand is sarilumab. [001073] In another example, as per example (vii), the inventor addressed human lambda constant region variation. Thus, in this example the ligand of the invention comprises or consists of an antibody that comprises a human 1GLC2*O1 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. 356 Example 4: Determination of Specific Binding of Ligands of the Invention to IL6R Variants The specific binding of ligands of the invention to IL6R variants can be performed using the following method. [001074| Method of SPR Determination of Binding Binding of the antibodies to the IL6R variants is carried out by SPR using the ProteOn XPR36™ Array system (BioRad). An anti-human IgG surface (Jackson Labs 109-005-008) was created on a GLC Biosensor chip by primary amine coupling, Test antibodies are captured on this surface as ligands. The IL6R variants are used as analytes and passed over the captured antibodies at 256nM, 64nM, 16nM, 4nM and InM. Binding curves are double referenced using a buffer injection (i.e. OnM) to remove baseline drift and injection artefacts. Regeneration ofthe capture surface is with 1 OOmM phosphoric acid which removes the captured antibody allowing another cycle of capture and binding. The binding sensorgrams generated are analysed using the 1:1 model inherent to the ProteOn XPR36 Array system analysis software. The assay is performed at 25 °C and using IxHBS-EP (Teknova) as running buffer. id="p-1075" id="p-1075"
[001075] References: The references cited herein are incorporated by reference in their entirety 1. Ferreira et al (PLoS Genet. 2013 Apr; 9(4):el003444. doi: 10.1371/journal.pgen. 1003444. Epub 2013 Apr 4, "Functional 1L6R 358Ala allele impairs classical IL-6 receptor signaling [sic] and influences risk of diverse in flammatory diseases"; 2. Rantala A et al, Hum Immunol. 2011 Jan;72(l):63-8. doi: 10.1016/j.humimm.2010.10.010. Epub 2010 Oct 15. "Association of IL-6 and IL-6R gene polymorphisms with susceptibility to respiratory tract infections in young Finnish men"; 3. Zhang HY etal, Oral Dis. 2014 Jan;20(l):69-75. doi: 10.111 l/odi.12075. Epub 2013 Feb 24, "The association of IL-6 and IL-6R gene polymorphisms with chronic periodontitis in a Chinese population"; 4. J C Galicia et al, Genes and Immunity (2004) 5, 513-516. doi: 10.1038/sj.gene.6364120 Published online 12 August 2004, "Polymorphisms in the IL-6 receptor (IL-6R) gene: strong evidence that serum levels of soluble IL-6R are genetically influenced"; 357 . Esparza-Gordillo J et al. J Allergy Clin Immunol. 2013 Aug;132(2):371-7. doi: .1016/j jaci.2013.01.057. Epub 2013 Apr 9, "A functional IL-6 receptor (IL6R) variant is a risk factor for persistent atopic dermatitis". 358 1) Table 12: 1000 GENOMES PROJECT HUMAN POPULATIONS Below is a summary of the ethnic populations as per the 1000 Genomes Project sequences. (a) 100 Genome Populations Population Code Population Description Super Population Code CHB Han Chinese in Bejing, China ASN J PT Japanese in Tokyo, Japan ASN CHS Southern Han Chinese ASN CDX Chinese Dai in Xishuangbanna, China ASN KHV Kinh in Ho Chi Minh City, Vietnam ASN CEO Utah Residents (CEPH) with Northern and Western European ancestry EUR TSI Toscani in Italia EUR FIN Finnish in Finland EUR GBR British in England and Scotland EUR IBS Iberian population in Spain EUR YRI Yoruba in Ibadan, Nigera AFR LWK Luhya in Webuye, Kenya AFR GWD Gambian in Western Divisons in The Gambia AFR MSL Mende in Sierra Leone AFR ESN Esan in Nigera AFR ASW Americans of African Ancestry in SW USA AFR ACB African Carribbeans in Barbados AFR MXL Mexican Ancestry from Los Angeles USA AMR PUR Puerto Ricans from Puerto Rico AMR CLM Colombians from Medellin, Colombia AMR PEL Peruvians from Lima, Peru AMR GIH Gujarati Indian from Houston, Texas SAN PJL Punjabi from Lahore, Pakistan SAN BEB Bengali from Bangladesh SAN STU Sri Lankan Tamil from the UK SAN ITU Indian Telugu from the UK SAN (b) Super Populations AFR, African AMR, Ad Mixed American 359 ASN, East Asian EUR, European SAN, South Asian (c) Population Ancestries European ancestry Utah residents (CEPH) with Northern and Western European ancestry (CEU) Toscani in Italia (TSI) British from England and Scotland (GBR) Finnish from Finland (FIN) Iberian populations in Spain (IBS) East Asian ancestry Han Chinese in Beijing, China (CHB) Japanese in Toyko, Japan (JPT) Han Chinese South (CFIS) Chinese Dai in Xishuangbanna (CDX) Kinh in Ho Chi Minh City, Vietnam (KHV) Chinese in Denver, Colorado (CHD) (pilot 3 only) West African ancestry Yoruba in Ibadan, Nigeria (YRI) 360 Luhya in Webuye, Kenya (LWK) Gambian in Western Division, The Gambia (GWD) Malawian in Blantyre, Malawi (MAB) West African Population (TBD) Americas African Ancestry in Southwest US (ASW) African American in Jackson, MS (AJM) African Caribbean in Barbados (ACB) Mexican Ancestry in Los Angeles, CA (MXL) Puerto Rican in Puerto Rico (PUR) Colombian in Medellin, Colombia (CLM) Peruvian in Lima, Peru (PEL) South Asian ancestry Ahom in the State of Assam. India Kayadtha in Calcutta, India Reddy in Hyderabad. India Maratha in Bombay, India Punjabi in Lahore, Pakistan 361 Table 13: Exemplary anti-IL6R disclosures, eg of antibodies and/or antibody fragments, assays, treatments, formulations, kits, methods and indications, useful in any and all aspects of the invention Patent or patent application which is incorporated by reference in its entirety, and specifically, eg, with respect to the SEQ ID Nos. comprising an anti-IL6R monoclonal antibody or fragment thereof US8568721, US20130157313A1, US20130149310A1, US20130122003A1, US8192741, US8183014, US20120003697A1, US8080248.
US8043617, US20110171241 Al, US201003 16636A1.
US20100316627A1, US7582298 Table 14: Human antibody gene segment variants distributed over several human ethnic populations - useful for ligand tailoring Cum o Tr 2 u. 00 00 00 6 Hom Freq8 (Het + Hom freq9) 0.847 Het Freq7 0.082 No. Individs6 Human Populations5 co o Variant Nucleotide Position 14:10655231 Nucleotide Variation3 (NCBI dbSNP reference number)4 E1 TCT ACG Amino Acid Coordinate2 Variation 30F (IMGT numbering) (Phe at position 4 in SEQ ID NO: 32) (CDR1 variation) 1 SOS 1 (IMGT numbering) (CDR1 variation) HOT Example Human Allele1 IGHV3-9*01 r\J o * CT) rh > <5 o * στ ch > X 52 Example Human Gene Segment Type IGHV3-9 _ 364 0.112 996Ό 0.034 0.961 0.039 (0.929) 0.071 (0.153) 0.934 (0.998) 0.002 (0.066) 1- . 0.924 (0.998) 0.002 0.082 0.064 0.064 0.074 £ o ό 167 1090 72 1090 83 CQ CO CD CO co (forward strand) 14:106552310 (forward strand) 2:89326669 (forward strand) ... - 2:89326669 (forward strand) 2:89326754 (forward strand) 2:89326754 ATG (rs8020204) CCT CAT (rsl82958807) 131 CCT (Thr at position 33 in SEQ ID NO: 34) (FW3 variation) 110M (FW3 variation) 115P (Pro at position 7 in SEQ ID NO: 36) (CDR3 variation) 115H (CDR3 variation) 87S (Ser at position 15 in SEQ ID NO: 38) (FW3 variation) 87P o 1 m > <0 o rn IGKV3-ll*01 IGKV3-11 365 0.243 in o 0.296 0.704 0.283 (0.076) - 0.071 (0.416) 0.584 (0.929) 0.096 (0.496) 0.504 (0.904) 0.104 (0.462) 0.345 LD o 0.400 0.400 1 I 0.358 454 1015 153 366 co OQ A | (European ancestry)A (European ancestry A (European (forward strand) 2:89156948 (forward strand) 2:89156948 (forward strand) 2:89156939 (forward strand) 2:89156939 (forward strand) 14:106208086 (forward strand) 14:106208086 (forward strand) [___________________________________________________________________________________________________ 14:106208082 (forward strand) (rsl91612627) GTC CTC (rs232230) TGC GGC (rs200765148) GAT ! GAG (rsl045853) CTG (FW3 variation) 84V (Val at position 84 in SEQ ID NO: 16) ________1 3 87C (Cys at position 87 in SEQ ID NO: 16) 87G 204D (CH3 variation) 204 E (CH3 variation) 206L o u 3 * o <0 IGKC*01 )GKC*02 IGHGl*01 IGHGl*03 IGHGl*01 IGKC ' "Ί IGHG1 366 0.717 OO o 6 0.292 0.997 o o ό 0.538 (0.896) 0.540 (0.876) 0.124 0.993 0.358 0.336 0.336 0.007 0.007 ! ancestry) A (European ancestry) CQ co < < 14:106208082 (forward strand) 14:106110914 (forward strand) 14:106110914 (forward strand) 14:106110904 (forward strand) 1__ 14:106110904 (forward strand) ATG (rsll621259) CCC ACC (rs!1627594) i .. AAC AGC (rs201590297) TTC (CH3 variation) 206M (CH3 variation) 72P (CHI variation) 72T (CHI variation) 75N (CHI variation) 75S (CHI variation) 76F (CHI variation) 76L (CHI variation) IGHG1*O3 IGHG2*01 IGHG2*02 IGHG2*O1 IGHG2*04 rd o * CM X o IGHG2*04 IGHG2 367 0.711 0.289 0.592 0.995 00 o ό 0.005 0.539 (0.881) 0.118 (0.46) 0.493 (0-692) 0.992 (6660) 0.308 (0.507) 0.002 (0.009) 0.342 0.342 661'0 0.007 0.199 1 0.007 o .5 D GO Έ δ CD co o Q o Q d .2 c3 > LS -i—» Οβ .s 'tn & s o o GO GO q Έ 'on o 14:10611013 (forward strand) 14:10611013 (forward strand) 14:106109752 (forward strand) 14:106109752 (forward strand) o ω GTG ATG (rs8009156) o o | tcc (rs4983499) Q go O (D 13 Q -C +-» o <4-4 GO ω g O£ IE o CD q on 8 m o CM 161V (CH2 variation) 161M (CH2 variation) 257A (CH3 variation) 257S (CH3 variation) 1 g GO E Ή .£ c o o CM K < E O -o υ co aj CD OO cn IGHG2*01 IGHG2*02 IGHG2*01 IGHG2*06 1 otes: c o o c .2 a GO aj on .£ δ U c -o .S a> □ 2 '3 CQ a. Z (Z) X) Ό Me Footn 5 s >—4 o E E z f—-4 CU Z □ο CM . < CQ U z en f--4 Tai 368 14. Human population used for representative variant frequency analysis. c u o CL o 369 Table 15: SEQUENCES SEQ ID NO: Human Allele Nucleotide/Amino Acid Sequence 78 IL-6R MLAVGCALLAALLAAPGAALAPRRCPAQEVARGVLTSLPGDSVTLTCPGVEPED NATVHWVLRKPAAGSHPSRWAGMGRRLLLRSVQLHDSGNYSCYRAGRPAGTVHL LVDVPPEEPQLSCFRKSPLSNWCEWGPRSTPSLTTKAVLLVRKFQNSPAEDFQ EPCQYSQESQKFSCQLAVPEGDSSFYIVSMCVASSVGSKFSKTQTFQGCGILQP DPPANITVTAVARNPRWLSVTWQDPHSWNSSFYRLRFELRYRAERSKTFTTWMV KDLQHHCVIHDAWSGLRHWQLRAQEEFGQGEWSEWSPEAMGTPWTESRSPPAE NEVSTPMQALTTNKDDDNILFRDSANATSLPVQDSSSVPLPTFLVAGGSLAFGT LLCIAIVLRFKKTWKLRALKEGKTSMHPPYSLGQLVPERPRPTPVLVPLISPPV SPSSLGSDNTSSHNRPDARDPRSPYDISNTDYFFPR Deposition 358 V=position 385 79 ATGCTGGCCGTCGGCTGCGCGCTGCTGGCTGCCCTGCTGGCCGCGCCGGGAGCG GCGCTGGCCCCAAGGC GCTGCCCTGCGCAGGAGGTGGCGAGAGGCGTGCTGACCAGTCTGCCAGGAGACA GCGTGACTCTGACCTG CCCGGGGGTAGAGCCGGAAGACAATGCCACTGTTCACTGGGTGCTCAGGAAGCC GGCTGCAGGCTCCCAC CCCAGCAGATGGGCTGGCATGGGAAGGAGGCTGCTGCTGAGGTCGGTGCAGCTC CACGAC TCTGGAAACT ATTCATGCTACCGGGCCGGCCGCCCAGCTGGGACTGTGCACTTGCTGGTGGATG TTCCCCCCGAGGAGCC CCAGCTCTCCTGCTTCCGGAAGAGCCCCCTCAGCAATGTTGTTTGTGAGTGGGG TCCTCGGAGCACCCCA TCCCTGACGACAAAGGCTGTGCTCTTGGTGAGGAAGTTTCAGAACAGTCCGGCC GAAGACTTCCAGGAGC CGTGCCAGTATTCCCAGGAGTCCCAGAAGTTCTCCTGCCAGTTAGCAGTCCCGG AGGGAGACAGCTCTTT CTACATAGTGTCCATGTGCGTCGCCAGTAGTGTCGGGAGCAAGTTCAGCAAAAC TCAAACCTTTCAGGGT TGTGGAATCTTGCAGCCTGATCCGCCTGCCAACATCACAGTCACTGCCGTGGCC AGAAACCCCCGCTGGC TCAGTGTCACCTGGCAAGACCCCCACTCCTGGAACTCATCTTTCTACAGACTAC GGTTTGAGCTCAGATA TCGGGCTGAACGGTCAAAGACATTCACAACATGGATGGTCAAGGACCTCCAGCA TCACTGTGTCATCCAC GACGCCTGGAGCGGCCTGAGGCACGTGGTGCAGCTTCGTGCCCAGGAGGAGTTC GGGCAAGGCGAGTGGA GCGAGTGGAGCCCGGAGGCCATGGGCACGCCTTGGACAGAATCCAGGAGTCCTC CAGCTGAGAACGAGGT GTCCACCCCCATGCAGGCACTTACTACTAATAAAGACGATGATAATATTCTCTT CAGAGATTCTGCAAAT GCGACAAGCCTCCCAGTGCAAGATTCTTCTTCAGTACCACTGCCCACATTCCTG GTTGCTGGAGGGAGCC TGGCCTTCGGAACGCTCCTCTGCATTGCCATTGTTCTGAGGTTCAAGAAGACGT GGAAGCTGCGGGCTCT GAAGGAAGGCAAGACAAGCATGCATCCGCCGTACTCTTTGGGGCAGCTGGTCCC GGAGAGGCCTCGACCC ACCCCAGTGCTTGTTCCTCTCATCTCCCCACCGGTGTCCCCCAGCAGCCTGGGG TCTGACAATACCTCGA GCCACAACCGACCAGATGCCAGGGACCCACGGAGCCCTTATGACATCAGCAATA CAGACTACTTCTTCCC CAGATAG 370 HEAVY CHAIN ALLELES ___ 80 IGHGl*01 (CHlTHinge+CH 2+CH3+CH-S) gcctccaccaagggcccatcggtcttccccctggcaccctcctccaagagcacctctggg ggcacagcggccctgggctgcctggtcaaggactacttccccgaaccggtgacggtgtcg tggaactcaggcgccctgaccagcggcgtgcacaccttcccggctgtcctacagtcctca ggactctactccctcagcagcgtggtgaccgtgccctccagcagcttgggcacccagacc tacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaagaaagttgagccc aaatcttgtgacaaaactcacacatgcccaccgtgcccagcacctgaactcctgggggga ccgtcagtcttcctcttccccccaaaacccaaggacaccctcatgatctcccggacccct 9a99tcacatgcgtggtggtggacgtgagccacgaagaccctgaggtcaagttcaactgg tacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtacaac agcacgtaccgggtggtcagcgtcctcaccgtcctgcaccaggactggctgaatggcaag gagtacaagtgcaaggtctccaacaaagccctcccagcccccatcgagaaaaccatctcc aaagccaaagggcagcoccgagaaccacaggtgtacaccctgcccccatcccgggatgag ctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagcgacatc gccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctcccgtg ctggactccgacggctccttcttcctctacagcaagctcaccgtggacaagagcaggtgg cagcaggggaacgtcttctcatgctccgtgatgcatgaggctctgcacaaccactacacg cagaagagcctctccctgtctccgggtaaa 81 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSWTVPSSSLGTQTYTCWNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN STYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGK D=position 204 Imposition 206 82 IGHG2*01 (CHl+Hinge+CH 2+CH3+CH-S) gcctccaccaagggcccatcggtcttccccctggcgccctgctccaggagcacctccgag agcacagccgccctgggctgcctggtcaaggactacttccccgaaccggtgacggtgtcg tggaactcaggcgctctgaccagcggcgtgcacaccttcccagctgtcctacagtcctca ggactctactccctcagcagcgtggtgaccgtgccctccagcaacttcggcacccagacc tacacctgcaacgtagatcacaagcccagcaacaccaaggtggacaagacagttgagcgc aaatgttgtgtcgagtgccoaccgtgcccagcaccacctgtggcaggaccgtcagtcttc ctcttccccccaaaacccaaggacaccctcatgatctcccggacccctgaggtcacgtgc Htggtggtggacgtgagccacgaagaccccgaggtccagttcaactggtacgtggacggc gtggaggtgcataatgccaagacaaagccacgggaggagcagttcaacagcacgttccgt gtggtcagcgtcctcaccgttgtgcaccaggactggctgaacggcaaggagtacaagtgc aaggtctccaacaaaggcctcccagcccccatcgagaaaaccatctccaaaaccaaaggg cagccccgagaaccacaggtgtacaccctgcccccatcccgggaggagatgaccaagaac caggtcagcctgacctgcctggtcaaaggcttctaccccagegacatcgccgtggagtgg gagagcaatgggcagccggagaacaactacaagaccacacctcccatgctggactccgac ggctccttcttcctctacagcaagctcaccgtggacaagagcaggtggcagcaggggaac gtcttctcatgctccgtgatgcatgaggctctgcacaaccactacacgcagaagagcctc tccctgtctccgggtaaa 83 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLiQSS GLYSLSSWTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVF leppkpkdtlmisrtpevtcvwdvshedpevqfnwyvdgvevhnaktkpreeqfnstfr WSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKN QVSLTCLVKGFYPSDIAVEWESNGQPΞNNYKTTPPMLDSDGSFFLYSKL·TVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK P-pCSition 72 Exposition 75 Exposition 76 V=position 161 Apposition 257 84 IGHV3-9*01 gaagtgcagctggtggagtctgggggaggcttggtacagcctggcaggtccctg agactctcctgtgcagcctctggattcacctttgatgattatgccatgcactgg gtccggcaagctccagggaagggcctggagtgggtctcaggtattagttggaat agtggtagcataggctatgcggactctgtgaagggccgattcaccatctccaga gacaacgccaagaactccctgtatctgcaaatgaacagtctgagagctgaggac 371 acggccttgtattactgtgcaaaagata t-nucleotide number 86 c=nucleotide number 272 85 EVQLVESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVSGISWN SGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKD F- position 30 (IMGT nomenclature) T- position 110 (dbSNP nomenclature) 86 IGHV3-7*01 gaggtgcagctggtggagtctgggggaggcttggtccagcctggggggtccctg agactc tcctgtgcagcctctggattcacctttagtagctattggatgagctgggtccgc caggct ccagggaaggggctggagtgggtggccaacataaagcaagatggaagtgagaaa tactat gtggactctgtgaagggccgattcaccatctccagagacaacgccaagaactca ctgtat ctgcaaatgaacagcctgagagccgaggacacggctgtgtattactgtgcgaga ga 87 EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVANIKQD GSEKYYVDSVKGRFTISRDNAKNSLiYLQMNSLRAEDTAVYYCAR 88 IGHJ3*02 T GAT GCT TTT GAT ATC TGG GGC CAA GGG ACA ATG GTC ACC GTC TCT TCA G 89 D A F D IWGQGTMVTVS S 90 IGHJ6*01 AT TAC TAC TAC TAC TAC GGT ATG GAC GTC TGG GGG CAA GGG ACC ACG GTC ACC GTC TCC TCA G 91 YYYYYGMDVWGQGT T V T V S S ___ ____________________ LIGHT CHAIN ALLELES _______________ 92 IGKC*O1 cgaactgtggctgcaccatctgtcttcatcttcccgccatctgatgagcagttgaaatct ggaactgcctctgttgtgtgcctgctgaataacttctatcccagagaggccaaagtacag tggaaggtggataacgccctccaatcgggtaactcccaggagagtgtcacagagcaggac agcaaggacagcacctacagcctcagcagcaccctgacgctgagcaaagcagactacgag aaacacaaagtctacgcctgcgaagtcacccatcagggcctgagctcgcccgtcacaaag agcttcaacaggggagagtgt 93 RTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQD SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC ^position 84 exposition 87 94 IGLC1*O1 cccaaggccaaccccacggtcactctgttcccgccctcctctgaggagctccaagccaac aaggccacactagtgtgtctgatcagtgacttctaccegggagetgtgacagtggcttgg aaggcagatggcagccccgtcaaggcgggagtggagacgaccaaaccctccaaacagagc aacaacaagtacgcggccagcagctacctgagcctgacgccegageagtggaagtcccac agaagctacagctgccaggtcacgcatgaagggagcaccgtggagaagacagtggcccct acagaatgttca 95 PKANPTVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADGSPVKAGVETT KPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS 96 IGKV1-12*O1 gacatccagatgacccagtctccatcttccgtgtctgcatctgtaggagacagagtcacc atcacttgtcgggcgagtcagggtattagcagctggttagcctggtatcagcagaaacca gggaaagcccctaagctcctgatctatgctgcatccagtttgcaaagtggggtcccatca aggttcagcggcagtggatctgggacagatttcactctcaccatcagcagcctgcagcct gaagattttgcaacttactattgtcaacaggctaacagtttccctcc 97 DIQMTQSPSSVSASVGDRVTITCRASQGISSWLAWYQQKPGKAPKLLIYAASSLQSGVPS RFSGSGSGTDFTLTISSLQPEDFATYYCQQMSFP 98 IGKV3-ll*01 gaaattgtgttgacacagtctccagccaccctgtctttgtctccaggggaaagagccacc ctctcctgcagggccagtcagagtgttagcagctacttagcctggtaccaacagaaacct 372 ggccaggctcccaggctcctcatctatgatgcatccaacagggccactggcatcccagcc aggttcagtggcagtgggtctgggacagacttcactctcaccatcagcagcctagagcct gaagattttgcagtttattactgtcagcagcgtagcaactggcctcc t-nucleotide number 199 c=nucleotide number 284 99 EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYEASNRATGIPA RFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNWP exposition 87 Exposition 115 ICO IGKJ2*01 tgtacacttttggccaggggaccaagctggagatcaaac 101 YTFGQGTKLEIK 102 IGKJ4*01 G CTC ACT TTC GGC GGA GGG ACC /LAG GTG GAG ATC AAA C 103 LTFGGGTKVEIK 104 An IGHGl*01 Heavy Chain EVQLVESGGG LVQPGRSLRL SCAASRFTFD DYAMHWVRQA PGKGLEWVSG ISWNSGRIGY ADSVKGRFTI SRDNAENSLF LQMNGLRAED TALYYCAKGR DSFDIWGQGT MVTVSSASTK GPSVFPLAPS SKSTSGGTAA LGCLVKBYFP EPVTVSWNSG ALTSGVHTFP AVLQSSGLYS LSSWTVPSS SLGTQTYICN VNHKPSNTKV DKKVEPKSCD KTHTCPPCPA PELLGGPSVF LFPPKPKDTL MISRTPEVTC WVDVSHEDP EVKFNWYVDG VEVHNAKTKP REEQYNSTYR WSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG QPREPQVTYL PPSRDELTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGK 105 An IGKC*01 Kappa Light Chain DIQMTQSPSS VSASVGDRVT ITCRASQGIS SWLAWYQQKP GKAPKLLIYG ASSLESGVPS RPSGSGSGTD FTLTISSLQP EDFASYYCQQ ANSFPYTFGQ GTKLEIKRTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC 106 An IGHGl*01 Heavy Chain EVQLVESGGGLVQPGGSLRLSCAASGFTFSPFAMSWVRQAPGKGLEWVAKISPG GSWTYY 3ΟΤνΤΟΡΓΤΙ8ΚΟΝΑΚΝ3ΕΥΕΰΜΝ3ΕΡΆΕϋΤΑνΥΥΟΑΡ0ΕΝαΥΥΑΕϋΙΝα0σΤΤ VTVSSA STKGPSVFPLAPSSKSTSGGTAALGCL·VKDYFPEPVTVSWNSGAL·TSGVHTFPA VLQSSG ΕΥ3Ε33νντνΡ333ΕαταΤΥΙ0ΝνΝΗΚΡ8ΝΤΚλ7ηΚΚνΕΡΚ300ΚΤΗΤ0ΡΡ0ΡΑΡ ELLGGP SVFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPR EEQYNS TYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQWTLP PSRDEL TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV DKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK 107 An IGKC*01 Kappa Light Chain EIVL·TQSPΆTLSL·SPGERATL·SCSASISVSYMYWYQQKPGQAPRL·I.IYDMSNLA SGIPAR FSGSGSGTDFTLTISSLEPEDFAVYYCMQWSGYPYTFGGGTKVEIKRTVAAPSV FIFPPS ΟΕ0ΕΚδΟΤΑεννσΕΕΝΝΡΥΡΡΕΑΚνςΜΚνΌΝΑΕ030Ν3(2Ε3νΤΕ(2Ό3Κ03ΤΥ3Ε SSTLTL SKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC 108 IGHV3-9*O1 CDR1 (IMGT ggattcacctttgatgattatgcc 373 nomenclature) imposition 11 109 GFTFDDYA Exposition 4 110 IGHV3-9*01 FW3 (IMGT nomenclature} ggc tat gcg gac tct gtg aag ggc cga ttc acc ate tcc aga gac aac gee aag aac tcc etg tat etg caa atg aac agt etg aga get gag gac acg gee ttg tat tac tgt c=position 98 111 G Y A D S V K GRFTISR DNAKNSLYLQMNSL RAEDTALYYC imposition 33 112 IGKV3 -11*01 CDR3 (IMGT Nomencalture) CAG CAG CGT AGC AAC TGG CCT CC C=nucleotide 20 113 QQRSNWP P=position 7 114 IGKV3-ll*01 FW3 (IMGT Nomencalture) aacagggccactggcatcccagccaggttcagtggcagtgggtctgggacagacttcact ctcaccatcagcagcctagagcctgaagattttgcagtttattaetgt T=nucleotide 43 115 NRATGIPAR FSGS GSGTD FTLTIS S LE PED FAVY YC S=position 15 Reference to sequence positions throughout this specification are to the sequence positions as identified in the description and tables. For example, positions 204 and 206 of SEQ JD NO:81 are the positions identified in SEQ JDNO:81 in Table 15. 374 STATEMENTS OF INVENTION: A. A method of treating or reducing the risk of an IL6R-mediated disease or condition in a human in need thereof, the method comprising administering to said human an antibody or antibody fragment that specifically binds an 1L6R protein that comprises a mutation Asp358Ala or Val385lle in SEQ ID NO: 78, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 or a Leu at position 206 shown in SEQ ID NO: 81 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: 81 or a Leu at position 206 shown in SEQ ID NO: 81 and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ue in SEQ ID NO: 78.
B. The method of statement A, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81.
C. The method of statement A, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHG1 *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78.
D. The method of any preceding statement, wherein the antibody or fragment comprises an IGHG1 *01 human heavy chain constant region.
E. The method of any preceding statement comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of statement 1.
F. The method of any preceding statement, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 and/or an 1L6R protein comprising said mutation Asp358Ala and/or 375 Val385Ile in SEQ ID NO: 78.
G. The method of any one of statements A-E, comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Va!385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile, optionally, wherein the determining step is performed before administration of the antibody to the human.
H. The method of statement G, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 1. The method of statement H, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the IL6R protein comprising said mutation Asp358AIa and/or Val3 85 He in SEQ ID NO: 78 is present; and detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78.
J. The method of statement H or I, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format.
K. The method of any one of statements HO, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human.
L. The method of any preceding statement, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385He in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide 376 sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78.
M. The method of any preceding statement, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment.
N. The method of any preceding statement, wherein said human is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment.
O. The method of any preceding statement, wherein said antibody or antibody fragment is administered to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment.
P. The method of any preceding statement, wherein said disease or condition is an inflammatory disease or condition.
Q. The method of any preceding statement, wherein said disease or condition is selected from the group consisting of an inflammatory bowel disease (IBD), Crohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen’s syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
R. The method of any preceding statement, wherein said human has been diagnosed with at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
S. The method of any preceding statement, wherein said antibody or antibody fragment treats or reduces the risk in said human of at least one condition selected from the group consisting of an inflammatory bowel disease (IBD), Chrohn’s disease, rheumatoid arthritis, psoriasis, bronchiolitis, gingivitis, transplant rejection, allogenic transplant rejection, graft-versus-host disease (GvHD), 377 asthma, adult respiratory distress syndrome (ARDS), septic shock, ulcerative colitis, Sjorgen's syndrome, airway inflammation, Castleman’s disease, periodontitis, atopic dermatitis, systemic lupus erythematosus and coronary heart disease.
T. The method of any preceding statement, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736.
U. The method of any preceding statement, wherein said antibody or antibody fragment is administered by intravenous or subcutaneous administration and/or is comprised in an injectable preparation.
Further Statements of Invention are as follows: 1. An antibody or antibody fragment for use in a method of treating or reducing the risk of rheumatoid arthritis in a human in need thereof, wherein the antibody or antibody fragment specifically binds an 1L6R protein that comprises a mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an amino acid selected from the group consisting of: an Asp at position 204 shown in SEQ ID NO:81 and a Leu at position 206 shown in SEQ ID NO: 81; wherein said human comprises (i) an IGHGl*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-1 heavy chain constant regions comprising said amino acid and (ii) a nucleotide sequence encoding said IL6R protein comprising said Asp358Ala or Val385Ile in SEQ ID NO: 78. 2. The antibody or antibody fragment of statement 1, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81. 378 3. The antibody or antibody fragment of statement 1, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81 and wherein said human comprises (i) an IGHG 1 *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding said IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 4. The antibody or antibody fragment of any preceding statement, wherein the antibody or fragment comprises an IGHG 1*01 human heavy chain constant region.
. The antibody or antibody fragment of any preceding statement, the method comprising, before said administering, selecting a human comprising said nucleotide sequence of (ii), wherein the human is the human of statement 1. 6. The antibody or antibody fragment of any preceding statement, wherein the human has been determined to comprise the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 7. The antibody or antibody fragment of any one of statements 1 -5, the method comprising the step of determining that the human comprises the nucleotide sequence that encodes an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile and/or an IL6R protein comprising said mutation Asp358Ala and/or Val385Ile. optionally, wherein the determining step is performed before administration of the antibody to the human. 8. The antibody or antibody fragment of statement 7, wherein the step of determining comprises assaying a biological sample from the human for a nucleotide sequence encoding a IL6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78. 9. The antibody or antibody fragment of statement 8, wherein 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 an IL6R protein comprising said mutation Asp358Ala and/or Val385 He in SEQ ID NO: 78 or comprising an antisense sequence of said contiguous nucleotides, wherein said sequence of contiguous nucleotides comprises a nucleotide sequence encoding said mutation Asp358Ala and/or Val385IIe in SEQ ID NO: 78, thereby forming a complex when at least one nucleotide sequence encoding the 1L6R protein comprising said mutation Asp358Ala and/or Val385Ile in SEQ ID NO: 78 is present; and 379 detecting the presence or absence of the complex, wherein detecting the presence of the complex determines that the human comprises the IL6R protein comprising said mutation Asp358Ala and/or Val385He in SEQ ID NO: 78.
. The antibody or antibody fragment of statement 8 or statement 9, wherein the assaying comprises nucleic acid amplification and optionally one or more methods selected from sequencing, next generation sequencing, nucleic acid hybridization, and allele-specific amplification and/or wherein the assaying is performed in a multiplex format. 11. The antibody or antibody fragment of any one of statements 8-10, wherein said biological sample comprises serum, blood, faeces, tissue, a cell, urine and/or saliva of said human. 12. The antibody or antibody fragment of any preceding statement, wherein said human is indicated as heterozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78, optionally, wherein said human is further indicated as comprising the nucleotide sequence of SEQ ID NO: 79, or said human is indicated as homozygous for a nucleotide sequence encoding the IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78. 13. The antibody or antibody fragment of any preceding statement, wherein said human is or has been further determined to be substantially resistant to an anti-TNF alpha treatment. 14. The antibody or antibody fragment of any preceding statement, wherein said human is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment.
. The antibody or antibody fragment of any preceding statement, wherein said antibody or antibody fragment is for administration to the human separately or simultaneously with methotrexate or an anti-TNF alpha treatment. 16. The antibody or antibody fragment of any preceding statement, wherein said human has been diagnosed with rheumatoid arthritis. 17. The antibody or antibody fragment of any preceding statement, wherein the nucleotide sequence comprises SNP rs2228145 and/or SNP rs28730736. 380 18. The antibody or antibody fragment of any preceding statement, wherein said antibody or antibody fragment is for administration by intravenous or subcutaneous administration and/or is comprised in an injectable preparation. 19. The antibody or antibody fragment of any preceding statement, wherein the antibody is a human antibody.

Claims (5)

Claims:
1. An injectable preparation comprising an antibody or antibody fragment for use in a method of treating or reducing the risk of rheumatoid arthritis in a human in need thereof, wherein the antibody or antibody fragment specifically binds an IL6R protein that comprises a mutation Asp358Ala or Val38511e in SEQ ID NO: 78, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an amino acid selected from the group consisting of: an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81; wherein said human comprises (i) an IGHGl*01 human heavy chain constant region gene segment, or the human expresses antibodies comprising human gamma-1 heavy chain constant regions comprising said amino acid and (ii) a nucleotide sequence encoding an IL6R protein comprising said Asp358Ala or Val385Jle in SEQ ID NO: 78.
2. The preparation of claim 1, wherein the antibody or antibody fragment comprises a human gamma-1 heavy chain constant region that comprises an Asp at position 204 shown in SEQ ID NO: 81 and a Leu at position 206 shown in SEQ ID NO: 81, optionally wherein said human comprises (i) an IGHG1 *01 human heavy chain constant region gene segment and (ii) a nucleotide sequence encoding an IL6R protein comprising said mutation Asp358Ala or Val385Ile in SEQ ID NO: 78.
3. The preparation of claim 1 or claim 2, wherein the antibody or fragment comprises an IGHG1 *01 human heavy chain constant region.
4. The preparation of any preceding claim, wherein the antibody or fragment is for administration to a human that is receiving or has received an anti-TNF alpha treatment or has reduced responsiveness to an anti-TNF alpha treatment. 382
5. The preparation of any preceding claim, wherein said antibody or antibody fragment is a human antibody or fragment.
IES20140315A 2014-08-12 2014-12-16 An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis IES86644B2 (en)

Applications Claiming Priority (22)

Application Number Priority Date Filing Date Title
US14/457,536 US9017678B1 (en) 2014-07-15 2014-08-12 Method of treating rheumatoid arthritis using antibody to IL6R
US14/457,566 US8945560B1 (en) 2014-07-15 2014-08-12 Method of treating rheumatoid arthritis using antibody to IL6R
US14/472,828 US8986691B1 (en) 2014-07-15 2014-08-29 Method of treating atopic dermatitis or asthma using antibody to IL4RA
US14/472,698 US8986694B1 (en) 2014-07-15 2014-08-29 Targeting human nav1.7 variants for treatment of pain
US14/472,818 US8980273B1 (en) 2014-07-15 2014-08-29 Method of treating atopic dermatitis or asthma using antibody to IL4RA
US14/472,685 US8992927B1 (en) 2014-07-15 2014-08-29 Targeting human NAV1.7 variants for treatment of pain
US14/490,160 US8999341B1 (en) 2014-07-15 2014-09-18 Targeting rare human PCSK9 variants for cholesterol treatment
US14/490,175 US9040052B1 (en) 2013-12-17 2014-09-18 Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
EP14185297.0A EP2975058A1 (en) 2014-07-15 2014-09-18 Antibodies for use in treating conditions related to specific PCSK9 variants in specific patient populations
US14/490,112 US9034331B1 (en) 2014-07-15 2014-09-18 Targeting rare human PCSK9 variants for cholesterol treatment
US14/490,091 US9068012B1 (en) 2014-07-15 2014-09-18 Targeting rare human PCSK9 variants for cholesterol treatment
US14/500,233 US9045548B1 (en) 2014-07-15 2014-09-29 Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US14/500,397 US10618971B2 (en) 2013-12-17 2014-09-29 Targeting rare human PCSK9 variants for cholesterol treatment
US14/507,368 US9034332B1 (en) 2014-07-15 2014-10-06 Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
EP14190945 2014-10-29
US14/536,129 US9062105B1 (en) 2014-07-15 2014-11-07 Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
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 US9067998B1 (en) 2014-07-15 2014-11-10 Targeting PD-1 variants for treatment of cancer
US14/552,816 US10611849B2 (en) 2013-12-17 2014-11-25 Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
EP14196638 2014-12-05
EP14196641 2014-12-05
EP14196645 2014-12-05

Publications (2)

Publication Number Publication Date
IES20140315A2 IES20140315A2 (en) 2016-02-24
IES86644B2 true IES86644B2 (en) 2016-05-04

Family

ID=55443019

Family Applications (2)

Application Number Title Priority Date Filing Date
IES20140315A IES86644B2 (en) 2014-08-12 2014-12-16 An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis
IES20140314A IES86646B2 (en) 2014-08-12 2014-12-16 An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis

Family Applications After (1)

Application Number Title Priority Date Filing Date
IES20140314A IES86646B2 (en) 2014-08-12 2014-12-16 An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis

Country Status (1)

Country Link
IE (2) IES86644B2 (en)

Also Published As

Publication number Publication date
IES86646B2 (en) 2016-05-04
IES20140315A2 (en) 2016-02-24
IES20140314A2 (en) 2016-02-24

Similar Documents

Publication Publication Date Title
US20240052025A1 (en) Precision medicine for cholesterol treatment
US20230024543A1 (en) Methods of treating anaemia
US9109034B1 (en) Precision medicine by targeting PD-L1 variants for treatment of cancer
US8945560B1 (en) Method of treating rheumatoid arthritis using antibody to IL6R
US9067998B1 (en) Targeting PD-1 variants for treatment of cancer
US8980273B1 (en) Method of treating atopic dermatitis or asthma using antibody to IL4RA
JP7202431B2 (en) Ligands that specifically bind to human targets of interest
US9017678B1 (en) Method of treating rheumatoid arthritis using antibody to IL6R
WO2015092393A2 (en) Human targets
WO2016071701A1 (en) Treatment of disease using ligand binding to targets of interest
US8986694B1 (en) Targeting human nav1.7 variants for treatment of pain
US8986691B1 (en) Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8992927B1 (en) Targeting human NAV1.7 variants for treatment of pain
WO2016023916A1 (en) Treatment of disease using ligand binding to targets of interest
US9150660B1 (en) Precision Medicine by targeting human NAV1.8 variants for treatment of pain
IES86644B2 (en) An injectable antibody preparation for use in treating or reducing the risk of rheumatoid arthritis
TWI713444B (en) Human targets
GB2521355A (en) Human targets I
IES86598B2 (en) An injectable antibody preparation for use in treating or reducing the risk of atopic dermatitis or asthma