WO2014155278A2 - Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'il -17 - Google Patents

Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'il -17 Download PDF

Info

Publication number
WO2014155278A2
WO2014155278A2 PCT/IB2014/060110 IB2014060110W WO2014155278A2 WO 2014155278 A2 WO2014155278 A2 WO 2014155278A2 IB 2014060110 W IB2014060110 W IB 2014060110W WO 2014155278 A2 WO2014155278 A2 WO 2014155278A2
Authority
WO
WIPO (PCT)
Prior art keywords
response
allele
marker
disease
patient
Prior art date
Application number
PCT/IB2014/060110
Other languages
English (en)
Other versions
WO2014155278A3 (fr
Inventor
Arndt Holger BRACHAT
Ying Wang
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Publication of WO2014155278A2 publication Critical patent/WO2014155278A2/fr
Publication of WO2014155278A3 publication Critical patent/WO2014155278A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the disclosure is directed to predictive methods, personalized therapies, kits,
  • autoimmune disease selected from uveitis, rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis (AS), psoriasis, and multiple sclerosis (MS).
  • RA rheumatoid arthritis
  • PsA psoriatic arthritis
  • AS ankylosing spondylitis
  • MS multiple sclerosis
  • AI diseases are pathological disorders caused by a self-reactive immune response.
  • AI diseases include, inter alia, rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis (AS), psoriasis, uveitis and multiple sclerosis (MS).
  • RA rheumatoid arthritis
  • PsA psoriatic arthritis
  • AS ankylosing spondylitis
  • MS multiple sclerosis
  • DMARDs Disease-modifying antirheumatic drugs
  • RA RA-reactive rheumatic drugs
  • Anti-TNF biological agents are the second line of treatment for RA patients, being used in DMARD-failure and DMARD-inadequate responder patients.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • Treatment of NSAIDs-refractory AS is hampered by the lack of efficacy of virtually all standard DMARDs, including MTX.
  • peripheral arthritis associated with AS responds quite well to sulfasalazine.
  • TNF-blocking agents have been successfully used to treat AS (Braun J et al (2002) Lancet 359: 1187-93) and demonstrate prolonged efficacy up to three years of follow-up (Braun et al.
  • MS is a clinically and pathogenic heterogeneous disease
  • therapeutic response varies depending on the disease stage (early vs. late phases), disease course (relapsing remitting vs. secondary progressive vs. primary progressive) and level of disease activity (breakthrough vs. benign).
  • Biologies, such as IFN- ⁇ , constitute the current first-line therapies with only modest efficacy ( ⁇ 30-35% reduction of Annualized Relapse Rate [ARR] vs placebo).
  • ARR Annualized Relapse Rate
  • Natalizumab has superior efficacy (68% reduction of ARR vs placebo), but major safety risks.
  • Mitoxantrone is used in aggressive forms of MS, but its use is progressively declining, mainly due to the cumulative cardiac toxicity and well-known malignancy risk.
  • Fingolimod a first line therapy in the US and second line therapy in the rest of the world, shows superior benefit on relapses over IFN- ⁇ (54 and 52% reduction of ARR vs placebo and IFN- ⁇ ).
  • switches between MS therapies are driven by a lack of efficacy and/or tolerability issues.
  • Secukinumab (AIN457) is a high-affinity fully human monoclonal anti-human antibody that inhibits Interleukin-17A activity.
  • secukinumab has emerged as a desirable treatment for patients with various Al diseases, e.g., uveitis, RA, PsA, AS, psoriasis, and MS.
  • Al diseases e.g., uveitis, RA, PsA, AS, psoriasis, and MS.
  • Al diseases are heterogeneous, patients will not necessarily respond to the same drug in the same way.
  • stratification of Al disease patients using markers predictive of response will help clinicians select and establish an effective treatment strategy with IL-17 antagonists (such as secukinumab).
  • patient stratification will avoid the safety issues and the high financial burden associated with long-term therapy.
  • IL-17 antagonists such as secukinumab
  • SNPs single nucleotide polymorphisms
  • novel predictive methods and personalized therapies for patients having an AI disease (uveitis, RA, PsA, AS, psoriasis, or MS) that maximize the benefit and minimize the risk of IL-17 antagonism in these populations by identifying those patients likely to respond favorably prior to treatment with an IL-17 antagonist.
  • AI disease uveitis, RA, PsA, AS, psoriasis, or MS
  • This finding is based, in part, on the determinations that:
  • PsA patients carrying at least one PsA response marker selected from an rs28096 response allele and two rs3819024 response alleles display improved response to secukinumab relative to PsA patients that do not carry at least one of these alleles (or, in the case of the rs3819024 response allele, relative to patient to do not carry both rs3819024 response alleles);
  • AS patients carrying at least one AS response marker selected from an rs 10484879 response allele, an rsl937154 response allele, two rs3819024 response alleles, and an rsl 1465770 response allele display improved response to secukinumab relative to AS patients that do not carry at least one of these alleles (or, in the case of the rs3819024 response allele, relative to patient to do not carry both rs3819024 response alleles);
  • psoriasis patients carrying an rs2241046 response allele display improved response to secukinumab relative to psoriasis patients that do not carry at least one of this allele;
  • MS patients carrying at least one MS response marker selected from an rs72773968 response allele, two rsl 7066096 response alleles, two rs2546890 response alleles, an rsl 800693 response allele, and two rs2863212 response alleles display improved response to secukinumab relative to MS patients that do not carry at least one of these alleles (or, in the case of the rsl7066096 response allele, rs2546890 response allele, and rs2863212 response allele, relative to patients to do not carry both alleles);
  • uveitis patients carrying at least one uveitis response marker selected from an rs 72773968 response allele, an rs2241046 response allele, an rsl 1465770 response allele, an rs 17482078 response allele, and an rs800292 response allele display improved response to secukinumab relative to uveitis patients that do not carry
  • an IL-17 antagonist e.g., an IL-17 antibody, such as secukinumab
  • an AI disease selected from uveitis, RA, PsA, AS, psoriasis, and MS who is more likely to respond to treatment of with an IL-17 antagonist, e.g., an IL-17 antibody, such as secukinumab, based on certain aspects of the patient's biochemical profile.
  • an IL-17 antagonist e.g., an IL-17 antibody, such as secukinumab
  • these methods comprise assaying a biological sample from the patient for at least one of the disclosed AI response markers; and thereafter selectively administering a therapeutically effective amount of an IL-17 antagonist, e.g., secukinumab, to the patient if the patient has the at least one AI response marker.
  • an IL-17 antagonist e.g., secukinumab
  • these methods comprise assaying a biological sample from the patient for the level of ERAP1 expression (e.g., mRNA, cDNA, etc.), the level of ERAPl protein, and/or the level of ERAPl activity; and thereafter selectively administering a therapeutically effective amount of an IL-17 antagonist, e.g., secukinumab, to the patient if the patient has a decreased level of ERAPl expression, decreased level of ERAPl protein, and/or decreased level of ERAPl activity relative to a control.
  • ERAP1 expression e.g., mRNA, cDNA, etc.
  • an IL-17 antagonist e.g., secukinumab
  • these methods comprise detecting at least one of the disclosed AI response markers in a biological sample from the patient, wherein the presence of the at least AI response marker is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist.
  • these methods comprise detecting the level of ERAPl expression (e.g., mRNA, cDNA, etc.), the level of ERAPl protein, and/or the level of ERAPl activity in a biological sample from the patient; wherein a decreased level of ERAPl expression, decreased level of ERAPl protein, and/or decreased level of ERAPl relative to a control is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist.
  • ERAPl expression e.g., mRNA, cDNA, etc.
  • a decreased level of ERAPl expression, decreased level of ERAPl protein, and/or decreased level of ERAPl relative to a control is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist.
  • the IL-17 antagonist is an IL-17 binding molecule, preferably a human antibody or antigen-binding portion thereof, most preferably secukinumab.
  • the AI response marker is at least one uveitis response marker, at least one RA response marker, at least one PsA response marker, at least one AS response marker, a psoriasis response marker, and/or a MS response marker as shown in Table 1.
  • the patient has multiple response markers as shown in Table 1.
  • the patient is an RA patient. In some embodiments, the patient is a PsA patient. In some embodiments, the patient is a uveitis patient. In some embodiments, the patient is an AS patient. In some embodiments, the patient is a psoriasis patient. In some embodiments, the patient is an MS patient.
  • Figure 1 shows association of rs 10484879 (located within an intron of IL17A) with secukinumab efficacy in RA trial F2201.
  • Figure 2 shows association of rs4711998 (located near IL17A) with secukinumab efficacy in RA trial F2201.
  • Figure 3 shows association of rs28096 (located within an intron of ERAPl) with secukinumab efficacy in PsA trial A2206.
  • Figure 4 shows association of rs27689 (located within an intron of ERAPl) with secukinumab efficacy in RA trials F2201 (Fig. 4A) and A2101 (Fig. 4B).
  • Figure 5 shows association of rs3214019 (located within an intron of ERAPl) with secukinumab efficacy in both RA trials F2201 (Fig. 5A) and A2101 (Fig. 5B).
  • Figure 6 shows association of rs 72773968 (missense SNP m ERAPl) with secukinumab efficacy in RA trial F2201 (Fig. 6 A) and MS trial B2201 (Fig. 6B).
  • Figure 7 shows association of rs 10484879 (located within an intron of IL17A) with secukinumab efficacy in RA trial F2201 (Fig. 7A) and AS trial A2209 (Fig. 7B).
  • Figure 8 shows association of rsl937154 (located near IL17A) with secukinumab efficacy in RA trial F2201 (Fig. 8A) & AS trial A2209 (Fig. 8B).
  • Figure 9 shows association of rs3819024 (located within the promoter of IL17A) with secukinumab efficacy in PsA trial A2206 (Fig. 9A) & AS trial A2209 (Fig. 9B).
  • Figure 10 shows association of rs2241046 (located within an intron of IL17RA) with secukinumab efficacy in RA trial F2201 (Fig. 10A) & psoriasis trial A2211 (Fig. 10B and IOC).
  • Figure 11 shows association of rsl 1465770 (located within an intron of IL23R) with secukinumab efficacy in RA trial F2201 (Fig. 11 A), AS trial A2209 (Fig. 11B) and uveitis trial A2208 (Fig. 11C).
  • Figure 12 shows association of rsl 7066096 (located interg enic IL20RA-IL22RA2) with secukinumab efficacy in MS trial B2201.
  • Figure 13 shows association of rs2546890 (located in an intron of IL12B) with secukinumab efficacy in MS trial B2201.
  • Figure 14 shows association of rsl 800693 (located in a splicing site of TNFRSF1A) with secukinumab efficacy in MS trial B2201.
  • Figure 15 shows association of rs2863212 (located in an intron of IL23R) with secukinumab efficacy in MS trial B2201.
  • Figure 16 shows association of rs 72773968 (missense oiERAPl) with secukinumab efficacy in Uveitis trial A2208.
  • Figure 17 shows association of rs2241046 (located in an intron of IL17RA) with secukinumab efficacy in Uveitis trial A2208.
  • Figure 18 shows association of rsl7482078 (missense oiERAPl) with secukinumab efficacy in Uveitis trial A2208.
  • Figure 19 shows association of rs800292 (missense of CFH) with secukinumab efficacy in Uveitis trial A2208.
  • composition “comprising” encompasses “including” as well as “consisting,” e.g. a composition “comprising” X may consist exclusively of X or may include something additional, e.g., X + Y.
  • the terms “subject” and “patient” include any human or nonhuman animal.
  • nonhuman animal includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
  • assaying is used to refer to the act of identifying, screening, probing, testing measuring or determining, which act may be performed by any conventional means.
  • a sample may be assayed for the presence of a particular genetic or protein marker by using an ELISA assay, a Northern blot, imaging, serotyping, cellular typing, gene sequencing, phenotyping, haplotyping, immunohistochemistry, western blot, mass spectrometry, etc.
  • detecting means the act of extracting particular information from a given source, which may be direct or indirect.
  • the presence of a given thing e.g., allele, level of protein, etc.
  • determining contemplate a transformation of matter, e.g., a transformation of a biological sample, e.g., a blood sample or other tissue sample, from one state to another by means of subjecting that sample to physical testing.
  • a biological sample e.g., a blood sample or other tissue sample
  • obtaining means to procure, e.g., to acquire possession of in any way, e.g., by physical intervention (e.g., biopsy, blood draw) or non- physical intervention (e.g, transmittal of information via a server), etc.
  • physical intervention e.g., biopsy, blood draw
  • non- physical intervention e.g., transmittal of information via a server
  • a sample may be tested (either directly or indirectly) for either the presence of a given AI response marker.
  • a sample may be tested (either directly or indirectly) for either the presence of a given AI response marker.
  • the presence or the absence of such substance may be used to guide a therapeutic decision. For example, one may determine if a patient has AI response marker by determining the actual existence of particular response allele in the patient or by determining the absence of the particular response allele in the patient. In both such cases, one has determined whether the patient has the presence of the AI response marker.
  • the disclosed methods involve, inter alia, determining whether a particular individual has an AI response marker. This determination is undertaken by identifying whether the patient has one or more of the AI response markers (i.e., uveitis response marker, RA response marker, PsA response marker, MS response marker, AS response marker, or psoriasis response marker) in Table 1. Each of these determinations (i.e., presence or absence), on its own, provides the allelic status of the patient and thus each of these deteriminations equally provide an indication of whether a particular individual would or would not respond more favorably to IL-17 antagonism.
  • the AI response markers i.e., uveitis response marker, RA response marker, PsA response marker, MS response marker, AS response marker, or psoriasis response marker
  • Table 1 shows the various response alleles of the disclosure.
  • Column 1 provides the gene in which the SNP of column 2 resides, and column 3 provides the general location of that SNP in that gene.
  • Column 4 provides the indication for which the SNP has response-predictive value.
  • the p value for a SNP (using the indicated disease measure) is presented in Column 5.
  • Column 6 gives the nucleotide that associates with an improved response for a given SNP and provides the allelic dosage, i.e., one [homozygous or heterozygous] or two [i.e., homozygous], required to show improved response to secukinumab.
  • Column 7 gives the mode of inheritance used in the analysis to get the p values as shown in column 5.
  • two rs3819024 response alleles is also a "PsA response marker," since PsA patients having two copies of this allele (i.e., homozygous for this allele) are more likely to have an improved response to secukinumab relative to PsA patients that do not have two copies of this allele.
  • Most SNPs listed in Table 1 have predictive value for IL-17 antagonism if there is at least one copy of a given response allele (i.e., the patient is either homozygous or heterozygous for a given response allele).
  • the data provided herein shows that, for most of such response alleles, there is an additive mode of effect, i.e., the presence of one copy of the response allele is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist, and the presence of two response alleles is indicative of an even futher increased likelihood that the patient will respond to treatment with the IL-17 antagonist.
  • a biological sample need only be assayed for one RA response marker, but clearly may be assayed for more than one of the RA response markers given in Table 1.
  • a biological sample need only be assayed for one uveitis response marker, but clearly may be assayed for more than one of the uveitis response markers given in Table 1.
  • a biological sample need only be assayed for one PsA response marker, but clearly may be assayed for both of the PsA response markers given in Table 1.
  • a biological sample need only be assayed for one AS response marker, but clearly may be assayed for more than one of the AS response markers given in Table 1.
  • a biological sample need only be assayed for one MS response marker, but clearly may be assayed for more than one of the MS response markers given in Table 1.
  • IL-17 refers to IL-17A, formerly known as CTLA8, and includes wild-type IL- 17A from various species (e.g., human, mouse, and monkey), polymorphic variants of IL-17A, and functional equivalents of IL-17A.
  • Functional equivalents of IL-17A preferably have at least about 85%, 95%, 96%, 97%, 98%, or even 99% overall sequence identity with a wild- type IL-17A (e.g., human IL-17A), and substantially retain the ability to induce IL-6 production by human dermal fibroblasts.
  • a wild- type IL-17A e.g., human IL-17A
  • IL-17 antagonist refers to a molecule capable of antagonizing (e.g., reducing, inhibiting, decreasing, delaying) IL-17 function, expression and/or signalling (e.g., by blocking the binding of IL-17 to the IL-17 receptor).
  • Non-limiting examples of IL-17 antagonists include IL-17 binding molecules and IL-17 receptor binding molecules.
  • an IL- 17 antagonist is employed.
  • IL-17 binding molecule any molecule capable of binding to the human IL- 17 antigen either alone or associated with other molecules.
  • the binding reaction may be shown by standard methods (qualitative assays) including, for example, a binding assay, competition assay or a bioassay for determining the inhibition of IL-17 binding to its receptor or any kind of binding assays, with reference to a negative control test in which an antibody of unrelated specificity, but ideally of the same isotype, e.g., an anti-CD25 antibody, is used.
  • Non-limiting examples of IL-17 binding molecules include small molecules, IL-17 receptor decoys, and antibodies that bind to IL-17 as produced by B-cells or hybridomas and chimeric, CDR-grafted or human antibodies or any fragment thereof, e.g., F(ab') 2 and Fab fragments, as well as single chain or single domain antibodies.
  • the IL-17 binding molecule antagonizes (e.g., reduces, inhibits, decreases, delays) IL-17 function, expression and/or signalling.
  • an IL- 17 binding molecule is employed.
  • IL-17 receptor binding molecule any molecule capable of binding to the human IL-17 receptor either alone or associated with other molecules.
  • the binding reaction may be shown by standard methods (qualitative assays) including, for example, a binding assay, competition assay or a bioassay for determining the inhibition of IL-17 receptor binding to IL-17 or any kind of binding assays, with reference to a negative control test in which an antibody of unrelated specificity, but ideally of the same isotype, e.g., an anti-CD25 antibody, is used.
  • Non- limiting examples of IL-17 receptor binding molecules include small molecules, IL-17 decoys, and antibodies to the IL-17 receptor as produced by B-cells or hybridomas and chimeric, CDR- grafted or human antibodies or any fragment thereof, e.g., F(ab') 2 and Fab fragments, as well as single chain or single domain antibodies.
  • the IL-17 receptor binding molecule antagonizes (e.g., reduces, inhibits, decreases, delays) IL-17 function, expression and/or signalling.
  • an IL-17 receptor binding molecule is employed.
  • antibody as referred to herein includes whole antibodies and any antigen- binding portion or single chains thereof.
  • a naturally occurring “antibody” is a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed hypervariable regions or complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each V H and V L is composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • an antibody to IL-17 or the IL-17 receptor is employed, preferably an antibody to IL-17, e.g., secukinumab.
  • antigen-binding portion of an antibody refers to fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., IL-17). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding portion" of an antibody include a Fab fragment, a monovalent fragment consisting of the V L , V H , CL and CHI domains; a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the V H and CHI domains; a Fv fragment consisting of the V L and V H domains of a single arm of an antibody; a dAb fragment (Ward et al, 1989 Nature 341 :544-546), which consists of a V H domain; and an isolated CDR.
  • Exemplary antigen binding sites include the CDRs of secukinumab as set forth in SEQ ID NOs: 1-6 and 11-13 (Table 2), preferably the heavy chain CDR3.
  • the two domains of the Fv fragment, V L and V H are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al, 1988 Science 242:423-426; and Huston et al, 1988 Proc. Natl. Acad. Sci. 85:5879-5883).
  • scFv single chain Fv
  • Single chain antibodies are also intended to be encompassed within the term "antibody”.
  • Single chain antibodies and antigen-binding portions are obtained using conventional techniques known to those of skill in the art. In some embodiments of the disclosed methods, regimens, kits, processes, uses and compositions, a single chain antibody or an antigen-binding portion of an antibody against IL-17 (e.g., secukinumab) or the IL-17 receptor is employed.
  • an “isolated antibody”, as used herein, refers to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds IL-17 is substantially free of antibodies that specifically bind antigens other than IL-17).
  • the term "monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • the term "human antibody”, as used herein, is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from sequences of human origin. A "human antibody” need not be produced by a human, human tissue or human cell.
  • the human antibodies of the disclosure may include amino acid residues not encoded by human sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro, by N- nucleotide addition at junctions in vivo during recombination of antibody genes, or by somatic mutation in vivo).
  • the IL-17 antagonist is a human antibody, an isolated antibody, and/or a monoclonal antibody.
  • K D is intended to refer to the dissociation rate of a particular antibody-antigen interaction.
  • K D is intended to refer to the dissociation constant, which is obtained from the ratio of K d to K a (i.e. 3 ⁇ 4/ ⁇ 3 ) and is expressed as a molar concentration (M).
  • KD values for antibodies can be determined using methods well established in the art. A method for determining the KD of an antibody is by using surface plasmon resonance, or using a biosensor system such as a Biacore® system.
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g.,
  • IL-17 receptor binding molecule e.g., IL-17 receptor antibody or antigen- binding portion thereof binds human IL-17 with a KD of about 100-250 pM.
  • affinity refers to the strength of interaction between antibody and antigen at single antigenic sites. Within each antigenic site, the variable region of the antibody “arm” interacts through weak non-covalent forces with antigen at numerous sites; the more interactions, the stronger the affinity.
  • Standard assays to evaluate the binding affinity of the antibodies toward IL-17 of various species are known in the art, including for example, ELISAs, western blots and RIAs.
  • the binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore analysis.
  • an antibody that "inhibits" one or more of these IL-17 functional properties will be understood to relate to a statistically significant decrease in the particular activity relative to that seen in the absence of the antibody (or when a control antibody of irrelevant specificity is present).
  • An antibody that inhibits IL-17 activity affects a statistically significant decrease, e.g., by at least about 10% of the measured parameter, by at least 50%, 80% or 90%, and in certain embodiments of the disclosed methods, uses, processes, kits and compositions, the IL-17 antibody used may inhibit greater than 95%, 98% or 99% of IL-17 functional activity.
  • “Inhibit IL-6” as used herein refers to the ability of an IL-17 antagonist (e.g., secukinumab) to decrease IL-6 production from primary human dermal fibroblasts.
  • an IL-17 antagonist e.g., secukinumab
  • the production of IL-6 in primary human (dermal) fibroblasts is dependent on IL-17 (Hwang et al, (2004) Arthritis Res Ther; 6:R120-128).
  • human dermal fibroblasts are stimulated with recombinant IL-17 in the presence of various concentrations of an IL-17 binding molecule or human IL-17 receptor with Fc part.
  • the chimeric anti-CD25 antibody Simulect ® (basiliximab) may be convienently used as a negative control.
  • An IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen- binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) as disclosed herein typically has an IC 50 for inhibition of IL-6 production (in the presence 1 nM human IL-17) of about 50 nM or less (e.g., from about 0.01 to about 50 nM) when tested as above, i.e., said inhibitory activity being measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
  • IL-17 antagonists e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof) and functional derivatives thereof have an IC 50 for inhibition of IL-6 production as defined above of about 20 nM or less, more preferably of about 10 nM or less, more preferably of about 5 nM or less, more preferably of about 2 nM or less, more preferably of about 1 nM or less.
  • derivative unless otherwise indicated, is used to define amino acid sequence variants, and covalent modifications (e.g., pegylation, deamidation, hydroxylation,
  • an IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL- 17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 receptor antibody or antigen-binding portion thereof) according to the present disclosure, e.g., of a specified sequence (e.g., a variable domain).
  • IL-17 binding molecule e.g., IL- 17 antibody or antigen-binding portion thereof, e.g., secukinumab
  • IL-17 receptor binding molecule e.g., IL-17 receptor antibody or antigen-binding portion thereof
  • a "functional derivative” includes a molecule having a qualitative biological activity in common with the disclosed IL-17 antagonists, e.g., IL-17 binding molecules.
  • a functional derivative includes fragments and peptide analogs of an IL-17 antagonist as disclosed herein.
  • Fragments comprise regions within the sequence of a polypeptide according to the present disclosure, e.g., of a specified sequence.
  • Functional derivatives of the IL-17 antagonists disclosed herein preferably comprise V H and/or V L domains that have at least about 65%, 75%, 85%, 95%, 96%, 97%, 98%, or even 99% overall sequence identity with the V H and/or V L sequences of the IL-17 binding molecules disclosed herein (e.g., the V H and/or V L sequences of Table 2), and substantially retain the ability to bind human IL-17 or, e.g., inhibit IL-6 production of IL-17 induced human dermal fibroblasts.
  • substantially identical means that the relevant amino acid or nucleotide sequence (e.g., V H or V L domain) will be identical to or have insubstantial differences (e.g., through conserved amino acid substitutions) in comparison to a particular reference sequence.
  • Insubstantial differences include minor amino acid changes, such as 1 or 2 substitutions (e.g., conservative substitutions, such as swapping a serine for a threonine, or substitutions at positions not involved in antibody activity, structural integrity, complement fixation, etc.) in a 5 amino acid sequence of a specified region (e.g., V H or V L domain).
  • the second antibody has the same specificity and has at least 50% of the affinity of the same.
  • sequences substantially identical e.g., at least about 85% sequence identity
  • sequence identity of a derivative IL-17 antibody can be about 90% or greater, e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher relative to the disclosed sequences.
  • Identity with respect to a native polypeptide and its functional derivative is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity, and not considering any
  • the percent identity can be determined by standard alignment algorithms, for example, the Basic Local Alignment Search Tool (BLAST) described by Altshul et al. ((1990) J. Mol. Biol, 215: 403 410); the algorithm of Needleman et al. ((1970) J. Mol. Biol., 48: 444 453); or the algorithm of Meyers et al. ((1988) Comput. Appl. Biosci., 4: 11 17).
  • BLAST Basic Local Alignment Search Tool
  • a set of parameters may be the Blosum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percent identity between two amino acid or nucleotide sequences can also be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4: 11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • amino acid(s) refer to all naturally occurring L-a-amino acids, e.g., and include D- amino acids.
  • amino acid sequence variant refers to molecules with some differences in their amino acid sequences as compared to the sequences according to the present disclosure. Amino acid sequence variants of a polypeptide according to the present disclosure, e.g., of a specified sequence, still have the ability to bind the human IL-17 or, e.g., inhibit IL-6 production of IL-17 induced human dermal fibroblasts.
  • Amino acid sequence variants include substitutional variants (those that have at least one amino acid residue removed and a different amino acid inserted in its place at the same position in a polypeptide according to the present disclosure), insertional variants (those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a polypeptide according to the present disclosure) and deletional variants (those with one or more amino acids removed in a polypeptide according to the present disclosure).
  • pharmaceutically acceptable means a nontoxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s).
  • administering in relation to a compound, e.g., an IL-17 binding molecule or another agent, is used to refer to delivery of that compound to a patient by any route.
  • a "therapeutically effective amount” refers to an amount of an IL-17 antagonist, e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 receptor antibody or antigen- binding portion thereof) that is effective, upon single or multiple dose administration to a patient (such as a human) for treating, preventing, preventing the onset of, curing, delaying, reducing the severity of, ameliorating at least one symptom of a disorder or recurring disorder, or prolonging the survival of the patient beyond that expected in the absence of such treatment.
  • IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 receptor antibody or antigen- binding portion thereof) that is effective, upon single
  • an active ingredient e.g., an IL-17 antagonist, e.g., secukinumab
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • treatment refers to both prophylactic or preventative treatment (as the case may be) as well as curative or disease modifying treatment, including treatment of a patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a patient having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a patient beyond that expected in the absence of such treatment.
  • a patient upon being delivered a particular treatment, e.g., an IL-17 binding molecule (e.g., secukinumab) shows a clinically meaningful benefit from said treatment.
  • a particular treatment e.g., an IL-17 binding molecule (e.g., secukinumab) shows a clinically meaningful benefit from said treatment.
  • such criteria include, e.g., ACR20, ACR50, ACR70, DAS28, etc.
  • AS such criteria include, e.g., ASAS40, ASAS56, etc.
  • psoriasis such criteria include, e.g., PASI, PASI75, etc.
  • Other criteria include MRI scans, vitreous haze analysis, neural scans, etc. All such criteria are acceptable measures of whether a patient is responding to a given treatment.
  • the phrase “respond to treatment” is used to mean that a patient, upon being delivered a particular treatment, e.g., an IL-17 binding
  • RA patient having an RA response marker is predicted to have more benefit from treatment with an IL-17 antagonist than an RA patient who does not have the RA response marker.
  • These carriers of RA response markers respond more favorably to treatment with the IL-17 antagonist, and may simply be said to "respond to treatment” with an IL-17 antagonist.
  • receiving data is used to mean obtaining possession of information by any available means, e.g., orally, electronically (e.g., by electronic mail, encoded on diskette or other media), written, etc.
  • selecting and “selected” in reference to a patient is used to mean that a particular patient is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient has an AI response marker (i.e., an RA response marker, a uveitis response marker, an AS response marker, a PsA response marker, a psoriasis response marker, or a MS response marker).
  • an AI response marker i.e., an RA response marker, a uveitis response marker, an AS response marker, a PsA response marker, a psoriasis response marker, or a MS response marker.
  • selectively treating refers to providing treatment to a patient having a particular disease, where that patient is specifically chosen from a larger group of patients on the basis of the particular patient having a predetermined criteria, e.g., an RA patient specifically chosen for treatment due to the patient having a RA response marker.
  • selectively administering refers to administering a drug to a patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., a particular genetic or other biological marker.
  • selectively treating and selectively administering it is meant that a patient is delivered a personalized therapy based on the patient's particular biology, rather than being delivered a standard treatment regimen based solely on the patient having a particular disease.
  • Selecting in reference to a method of treatment as used herein, does not refer to fortuitous treatment of a patient that has an AI response marker, but rather refers to the deliberate choice to administer an IL-17 antagonist to a patient based on the patient having an AI response marker.
  • selective treatment differs from standard treatment, which delivers a particular drug to all patients, regardless of their allelic status.
  • predicting indicates that the methods described herein provide information to enable a health care provider to determine the likelihood that an individual having an AI disease selected from uveitis, RA, PsA, AS, psoriasis and MS will respond to or will respond more favorably to treatment with an IL-17 binding molecule. It does not refer to the ability to predict response with 100% accuracy. Instead, the skilled artisan will understand that it refers to an increased probability.
  • Likelihood and “likely” is a measurement of how probable an event is to occur. It may be used interchangably with “probability”. Likelihood refers to a probability that is more than speculation, but less than certainty. Thus, an event is likely if a reasonable person using common sense, training or experience concludes that, given the circumstances, an event is probable. In some embodiments, once likelihood has been ascertained, the patient may be treated (or treatment continued, or treatment proceed with a dosage increase) with the IL-17 binding molecule or the patient may not be treated (or treatment discontinued, or treatment proceed with a lowered dose) with the IL-17 binding molecule.
  • the phrase "increased likelihood” refers to an increase in the probability that an event will occur.
  • some methods herein allow prediction of whether a patient will display an increased likelihood of responding to treatment with an IL-17 binding molecule or an increased likelihood of responding better to treatment with an IL-17 binding molecule in comparison to a patient having an AI disease selected from uveitis, RA, PsA, AS, psoriasis and MS who does not have an AS response marker (i.e., a uveitis response marker, an RA response marker, an AS response marker, a PsA response marker, a psoriasis response marker, or a MS response marker).
  • an AI disease selected from uveitis, RA, PsA, AS, psoriasis and MS who does not have an AS response marker (i.e., a uveitis response marker, an RA response marker, an AS response marker, a PsA response marker, a psoriasis response marker, or
  • SNP single nucleotide polymorphism
  • a SNP may be present in an exon or an intron of a gene, an upstream or downstream untranslated region of a gene, or in a purely genomic location (i.e., non- transcribed).
  • the SNP may be silent (i.e., a synonymous polymorphism) due to the redundancy of the genetic code, or the SNP may result in a change in the sequence of the encoded polypeptide (i.e., a non-synonymous polymorphism).
  • SNPs are identified by their Single Nucleotide Polymorphism Database (dbSNP) rs number, e.g., "rs4263839".
  • dbSNP Single Nucleotide Polymorphism Database
  • NCBI Biotechnology Information
  • NHGRI National Human Genome Research Institute
  • a polymorphic site such as a SNP
  • SNP is usually preceded by and followed by conserved sequences in the genome of the population of interest and thus the location of a polymorphic site can often be made in reference to a consensus nucleic acid sequence (e.g., of thirty to sixty nucleotides) that bracket the polymorphic site, which in the case of a SNP is commonly referred to as the "SNP context sequence".
  • Context sequences for the SNPs disclosed herein may be found in the NCBI SNP database available at: www.ncbi.nlm.nih.gov/snp.
  • the location of the polymorphic site may be identified by its location in a reference sequence (e.g., GeneBank deposit) relative to the start of the gene, mRNA transcript, BAC clone or even relative to the initiation codon (ATG) for protein translation.
  • a reference sequence e.g., GeneBank deposit
  • ATG initiation codon
  • any polymorphic site described herein by reference to a particular position in a reference or context sequence is merely for convenience and that any specifically enumerated nucleotide position literally includes whatever nucleotide position the same polymorphic site is actually located at in the same locus in any individual being tested for the genetic marker of the invention using any of the genotyping methods described herein or other genotyping methods known in the art.
  • genetic polymorphisms include translocations, insertions, substitutions, deletions, etc., that occur in gene enhancers, exons, introns, promoters, 5' UTR, 3'UTR, etc.
  • rs 10484879 refers to an A/C (rev) SNP located within an intron of the human IL-17A gene (GeneBank Accession No. NM_002190.2).
  • the IL-17A gene encodes the IL-17A cytokine, which is involved in the Thl7 pathway, and various autoimmine and inflammatory responses.
  • the rs 10484879 polymorphic site is located at chromosomal postion 52051957 (build 37.3; assembly GRCh37.p5), which is position 51991957 of Contig
  • rsl 0484879 response allele refers to the the "C” allele ("G" allele, in the case of the complementary strand) at the rsl 0484879 polymorphic site.
  • the patient has at least one rsl 0484879 response allele.
  • rs4711998 refers to an A/G (fwd) SNP located near the human IL-17A gene (GeneBank Accession No. NM_002190.2).
  • the rs4711998 polymorphic site is located at chromosomal postion 52050353 (build 37.3; assembly GRCh37.p5), which is position 51990353 of Contig NT_007592.15.
  • the phrase “rs4711998 response allele” as used herein refers to the “G” allele ("C” allele, in the case of the complementary strand) at the rs4711998
  • the patient has at least one rs4711998 response allele.
  • the ERAPl gene encodes the ERAPl protein, a multifunctional enzyme involved in cytokine receptor cleavage and trimming of peptides for presentation by major histocompatibility complex (MHC) class I molecules.
  • MHC major histocompatibility complex
  • rs30187 refers to a T/C SNP in the human ERAPl gene located on chromosome 5, which is associated with both AS and MS (Wellcome Trust et al (2007) Nat Genet. 39(11): 1329-37; M. Brown (2008) Rheumatology 47(2): 132-7; Guenni et al.
  • the rs30187 polymorphic site is located at chromosomal position 96,150,086 (NCBI genome build 36.3), position 30,519 of the human ERAPl gene set forth as GeneBank Accession No. NG_027839.1, position 1,688 of the human ERAPl transcript variant 3 mRNA set forth as GeneBank Accession No. NM_001198541, position 1,930 of the human ERAPl transcipt variant 2 mRNA set forth as GeneBank Accession No. NM_001040458; codon encoding amino acid 528 of the human ERAPl protein set forth as GeneBank Accession No. NP_057526.3).
  • the rs30187 "C” allele encodes a K528R variant of ERAPl that has impaired catalytic properties, and is associated with reduced incidence of AS. (Kochan et al. (2011) Proc Natl Acad Sci U S A. 108(19):7745-50). Each "T” allele of rs30187 increases the odds of having AS by about 1.4x. We have previously determined that AS patients having a rs30187 "T” allele display decreased secukinumab response (US Provisional Patent Application 61/636,062, which is incorporated by reference herein in its entirety).
  • rs27434 refers to an A/G SNP in the human ERAPl gene. (Lin et al. (2011) J Rheumatol. 38(2):317-21).
  • the rs27434 polymorphic site is located at position 25,337 of the human ERAPl gene set forth as GeneBank Accession No. NG_027839.1 (position 1,415 of the human ERAPl mRNA set forth as GeneBank Accession No. NM_016442.3; codon encoding amino acid 356 of the human ERAPl protein set forth as GeneBank Accession No. NP_057526.3).
  • the rs27434 SNP is a synonymous polymorphism occuring in the codon for Ala356 of the ERAPl protein. (Harvey et al. (2009) Hum. Mol. Genet. 18 (21): 4204-4212). Each "A" disease allele of rs27434 increases the odds of having AS by about 1.19x. We have previously determined that AS patients having a rs27434 "A” allele display decreased response to secukinumab (US Provisional Patent Application 61/636,062, which is incorporated by reference herein in its entirety).
  • rs27689 refers to a G/T SNP located in an intron of the human ERAPl gene (position 47647 of GeneBank Accession No. NG_027839.1). The ERAPl mRNA sequence is found in GeneBank Accession No. NM_016442.3. The rs27689 polymorphic site is located at chromosomal postion 96107202 (build 37.3; assembly GRCh37.p5), which is position 4421074 of Contig NT_034772.6.
  • the phrase “rs27689 response allele” as used herein refers to the the "T” allele ("A" allele, in the case of the complementary strand) at the rs27689 polymorphic site. In some embodiments of the disclosed methods, uses, and kits, the patient has two rs27689 response alleles.
  • rs3214019 refers to an A/G SNP located in an intron of the human ERAPl gene (position 110370 of GeneBank Accession No. NG_029490.1).
  • the ERAPl mRNA sequence is found in GeneBank Accession No. NM_001042440.2.
  • the rs3214019 polymorphic site is located at chromosomal postion 96103110 (build 37.3; assembly GRCh37.p5), which is position 4416982 of Contig NT_034772.6.
  • the phrase “rs3214019 response allele” as used herein refers to the the "G” allele ("C” allele, in the case of the complementary strand) at the rs3214019 polymorphic site.
  • the patient has at least one rs3214019 response allele.
  • rs 72773968 refers to an A/G SNP located in an exon of the human ERAP1 gene (position 15254 of GeneBank Accession No. NG_027839.1).
  • the rs72773968 polymorphic site is located at chromosomal postion 96139595 (build 37.3; assembly
  • GRCh37.p5 which is position 4453467 of Contig NT_034772.6.
  • the rs72773968 polymorphic site is located at position 382 ⁇ ERAPl mRNA GeneBank Accession No. NM_001040458.1 and position 140 of GeneBank Accession No. NM 001198541.1.
  • the resulting missense mutation is a Thr - He at position 12 of the ERAPl protein (position 12 of NP_001035548.1 and NP_001185470.1).
  • rs72773968 response allele refers to the the "A" allele (T allele, in the case of the complementary strand) at the rs 72773968 polymorphic site.
  • the patient has at least one rs72773968 response allele.
  • rs 17482078 refers to a C/T SNP located in an exon of the human ERAPl gene (position 35983 of GeneBank Accession No. NG_027839.1).
  • the rsl7482078 polymorphic site is located at chromosomal postion 96118866 (build 37.3; assembly GRCh37.p5), which is position 4432738 of Contig NT_034772.6.
  • the rsl7482078 polymorphic site is located at position 2521 of ERAPl mRNA GeneBank Accession No. NM_001040458.1 and position 2279 of GeneBank Accession No. NM_001198541.1.
  • rsl 7482078 response allele refers to the the "T” allele ("A” allele, in the case of the complementary strand) at the rsl 7482078 polymorphic site.
  • the patient has at least one rsl7482078 response allele. Bettencourt et al.
  • rsl7482078 "A" allele (Arg725Gln) provided a protective effect from AS in HLA-B27 positive individuals.
  • rsl 937154 refers to a C/T SNP located near the human IL-17A gene.
  • rsl937154 polymorphic site is located at chromosomal postion 52057735 (build 37.3; assembly GRCh37.p5), which is position 51997735 of Contig NT_007592.15.
  • rsl937154 response allele refers to the the "T” allele (A allele, in the case of the
  • the patient has at least one rsl937154 response allele.
  • rs2241046 refers to a C/T SNP located in an intron of the human IL- 17RA gene (position 25621 of GeneBank Accession No. NG_028257.1).
  • the IL17RA gene encodes the IL-17 cytokine receptor, which is involved in the Thl7 pathway, and various autoimmine and inflammatory responses.
  • the sequence of IL-17RA mRNA is found in
  • rs2241046 polymorphic site is located at chromosomal postion 17586471 (build 37.3; assembly GRCh37.p5), which is position 738621 of Contig NT_011519.10.
  • rs2241046 response allele refers to the the "C” allele (G allele, in the case of the complementary strand) at the rs2241046 polymorphic site.
  • the patient has at least one rs2241046 response allele.
  • rsl 1465770 refers to a C/T SNP located in an intron of the human IL- 23R gene (position 6795 of GeneBank Accession No. NG 011498.1).
  • the IL23R gene encodes the IL-23 cytokine receptor, which is involved in the Thl7 pathway, and various autoimmine and inflammatory responses.
  • the sequence of IL-23R mRNA is found in GeneBank Accession No. NM_144701.2.
  • the rsl 1465770 polymorphic site is located at chromosomal postion 67633963 (build 37.3; assembly GRCh37.p5), which is position 37605881 of Contig NT_032977.9.
  • rsl 1465770 response allele refers to the the "T” allele (A allele, in the case of the complementary strand) at the rsl 1465770 polymorphic site.
  • the patient has at least one rsl 1465770 response allele.
  • rs28096 refers to an A/G SNP located in an intron of the human ERAPl gene (position 45605 of GeneBank Accession No. NG_027839.1). The sequence of ERAPl mRNA is found in GeneBank Accession No. NM_016442.3.
  • the rs28096 polymorphic site is located at chromosomal postion 96109244 (build 37.3; assembly GRCh37.p5), which is position 4423116 of Contig NT_034772.6.
  • rs28096 response allele refers to the the "T” allele ("A" allele, in the case of the complementary strand) at the rs28096 polymorphic site.
  • the patient has at least one rs28096 response allele.
  • rs3819024 refers to an A/G SNP located in the promoter for the human IL-17 A gene.
  • the sequence of IL-17 A mRNA is found in GeneBank Accession No.
  • rs3819024 polymorphic site is located at chromosomal postion 52050786 (build 37.3; assembly GRCh37.p5), which is position 51990786 of Contig NT_007592.15.
  • the phrase "rs3819024 response allele” as used herein refers to the the "A” allele (T allele, in the case of the complementary strand) at the rs3819024 polymorphic site. In some embodiments of the disclosed methods, uses, and kits, the patient has two rs3819024 response alleles.
  • rs 17066096 refers to an A/G SNP located intergenic between the human IL20RA and IL22RA2 genes.
  • the rsl7066096 polymorphic site is located at chromosomal postion 137452908 (build 37.3; assembly GRCh37.p5), which is position 41622365 of Contig NT_025741.15.
  • rsl7066096-G is risk allele of MS (Patsopoulos NA, et al, 2011).
  • the phrase “rs 17066096 response allele” as used herein refers to the "A" allele ("T" allele, in the case of the complementary strand) at the rs 17066096 polymorphic site. In some embodiments of the disclosed methods, uses, and kits, the patient has two rs 17066096 response alleles.
  • rs2546890 refers to an A/G SNP located in an intron of the human IL- 12B gene.
  • the rs2546890 polymorphic site is located at chromosomal postion 158759900 (build 37.3; assembly GRCh37.p5), which is position 3571173 of Contig NT_023133.13, which is also position 2582 of GeneBank Accession No. NG 009618.1 and position 634 of GeneBank
  • rs2546890-A is risk allele of MS (Patsopoulos NA et al, 2011).
  • the IL12B gene encodes the IL-12B cytokine, which is a cytokine that acts on T and natural killer cells, and has a broad array of biological activities.
  • the phrase "rs2546890 response allele” as used herein refers to the the "G” allele ("C" allele, in the case of the complementary strand) at the rs2546890 polymorphic site. In some embodiments of the disclosed methods, uses, and kits, the patient has two rs2546890 response alleles.
  • rsl 800693 refers to an A/G SNP located in splicing site of the human TNFRSFIA gene.
  • the rsl 800693 polymorphic site is located at chromosomal postion 6440009 (build 37.3; assembly GRCh37.p5), which is position 6380009 of Contig NT_009759.16, which is also position 16253 of GeneBank Accession No. NG_007506.1.
  • the TNFRSFIA gene encodes on of the major receptors for TNF-alpha. This receptor can activate NF-kappaB, mediate apoptosis, and function as a regulator of inflammation.
  • rsl800693-G is a risk allele of MS, directing expression of a novel, soluable form of TNFSR1 that can block TNF.
  • rsl800693-G mirrors the outcome of anti-TNF therapy in MS patients (Gregory et al 2012).
  • the phrase "rsl 800693 response allele” as used herein refers to the the "G” allele ("C" allele, in the case of the complementary strand) at the rsl 800693 polymorphic site.
  • the patient has at least one rsl 800693 response allele.
  • rs2863212 refers to a C/T SNP located in an intron of the human IL-23R gene.
  • the rs2863212 polymorphic site is located at chromosomal postion 67685116 (build 37.3; assembly GRCh37.p5), which is position 37657034 of Contig NT_032977.9, which is also position position 57948 of GeneBank Accession No. NG_011498.1.
  • the phrase “rs2863212 response allele” as used herein refers to the the "T” allele ("A" allele, in the case of the complementary strand) at the rs2863212 polymorphic site. In some embodiments of the disclosed methods, uses, and kits, the patient has two rs2863212 response alleles.
  • rs800292 refers to a C/T SNP located in an exon of the human complement factor H (CFH) gene (position 26093 of GeneBank Accession No. NG_007259.1). CFH is involved in the human complement system and is associated with dense deposit disease and age-realted macular degeneration. The rs800292 polymorphic site is located at
  • the rs800292 polymorphic site is located at position 424 of CFH mRNA GeneBank Accession No. NM_000186.3 and position 62 of GeneBank Accession No. NP_000177.2
  • a "A” replaces a "G” at this position
  • the resulting missense mutation is a Val - He at position 52 of the CFH protein (position 12 of NP 000177.2).
  • the phrase "rs800292 response allele” as used herein refers to the the "C” allele (G allele, in the case of the complementary strand) at the rs800292 polymorphic site.
  • the patient has at least one rs800292 response allele.
  • Yang et al. (2012) Mol. Vis. 18: 1865-72 reports that carriers of G allele for CFH-rs800292 is higher in patients with non-infectious intermediate and posterior uveitis than in controls.
  • the rsl 0484879 response allele, the rs4711998 response allele, the rs27689 response allele, the rs3214019 response allele, the rs 72773968 response allele, the rsl 937154 response allele, the rs2241046 response allele, and the rsl 1465770 response allele are collectively "RA response markers.”
  • the patient is an RA patient that has at least one RA response marker.
  • the rs28096 response allele and two rs3819024 response alleles are collectively "PsA response markers".
  • the patient is a PsA patient that has at least one PsA response marker.
  • the rsl0484879 response allele, the rsl937154 response allele, two rs3819024 response alleles, and the rsl 1465770 response allele are collectively "AS response markers".
  • the patient is an AS patient that has at least one AS response marker.
  • the rs2241046 response allele is also referred to as the "psoriasis response marker".
  • the patient is a psoriasis patient that has the psoriasis response marker.
  • the rs72773968 response allele, the rsl 7066096 response allele, the rs2546890 response allele, the rsl800693 response allele, and the rs2863212 response allele are collectively "MS response markers.”
  • the patient is an MS patient that has an MS response marker.
  • the rs72773968 response allele, the rs2241046 response allele, the rsl 1465770 response allele, the rsl7482078 response allele, and the rs800292 response allele are collectively "uveitis response markers.”
  • the patient is a uveitis patient that has a uveitis response marker.
  • the rs72773968 response allele is useful for predicting uveitis, RA and MS patient's response to IL-17 antagonism
  • the rsl937154 response allele is useful for predicting both RA and AS patient's response to IL-17 antagonism, etc.
  • each of these response alleles are members of two response marker families, e.g., the rsl 937154 response allele is an RA response marker and an AS response marker.
  • AI response marker(s) collectively refers to the uveitis response markers, RA response markers, PsA response markers, AS response markers, psoriasis response marker and MS response markers disclosed herein.
  • the patient has at least one AI response marker.
  • nucleic acid samples containing a particular SNP may be complementary double stranded molecules and thus reference to a particular site on the sense strand refers as well to the corresponding site on the complementary antisense strand.
  • a particular genotype obtained for a SNP on both copies of one strand of a chromosome is equivalent to the complementary genotype obtained for the same SNP on both copies of the other strand.
  • an A/G genotype for the rs3819024 polymorphic site on one strand is equivalent to a T/C genotype for that polymorphic site on the complementary strand.
  • genomic sequence refers to a DNA sequence present in a genome, and includes a region within an allele, an allele itself, or a larger DNA sequence of a chromsome containing an allele of interest.
  • Products of the AI response markers include nucleic acid products and polypeptide products.
  • Polypeptide product refers to a polypeptide encoded by an AI response markers (i.e., products of uveitis response markers, RA response markers, PsA response markers, AS response markers, psoriasis response marker and MS response markers) and fragments thereof.
  • Nucleic acid product refers to any DNA (e.g., genomic, cDNA, etc.) or RNA (e.g., pre-mRNA, mRNA, miRNA, etc.) products of an AI response markers (i.e., products of uveitis response markers, RA response markers, PsA response markers, AS response markers, psoriasis response marker and MS response markers) and fragments thereof.
  • an AI response markers i.e., products of uveitis response markers, RA response markers, PsA response markers, AS response markers, psoriasis response marker and MS response markers
  • an “equivalent genetic marker” refers to a genetic marker that is correlated to an allele of interest, e.g., it displays linkage disequilibrium (LD) or is in genetic linkage with the allele of interest. Equivalent genetic markers may be used to determine if a patient has an AI response marker, rather than directly interrogating a biological sample from the patient for the allele per se.
  • Various programs exist to help determine LD for particular SNPs e.g, HaploBlock (available at bioinfo.cs.technion.ac.il/haploblock/), HapMap, WGA Viewer.
  • probe refers to any composition of matter that is useful for specifically detecting another substance, e.g., a substance related to an AI response marker.
  • a probe can be an oligonucleotide (including a conjugated oligonucleotide) that specifically hybridizes to a genomic sequence of an AI response marker, or a nucleic acid product of an AI response marker.
  • a conjugated oligonucleotide refers to an oligonucleotide covalently bound to chromophore or molecules containing a ligand (e.g., an antigen), which is highly specific to a receptor molecule (e.g., an antibody specific to the antigen).
  • the probe can also be a PCR primer, e.g., together with another primer, for amplifying a particular region within an AI response marker.
  • the probe can be an antibody that specifically binds to polypeptide products of these alleles.
  • the probe can be any composition of matter capable of detecting (e.g., binding or hybridizing) an equivalent genetic marker of an AI response marker.
  • the probe specifically hybridizes to a nucleic acid sequence (preferably genomic DNA) or specifically binds to a polypeptide sequence of an allele of interest.
  • the probe is at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35 nucleic acids in length.
  • stringent hybridization is used to refer to hybrization under stringent hybridization conditions.
  • Stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. Aqueous and nonaqueous methods are described in that reference and either can be used.
  • One example of stringent hybridization conditions is hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by at least one wash in 0.2X SSC, 0.1% SDS at 50°C.
  • SSC sodium chloride/sodium citrate
  • a second example of stringent hybridization conditions is hybridization in 6X SSC at about 45°C, followed by at least one wash in 0.2X SSC, 0.1% SDS at 55°C.
  • Another example of stringent hybridization conditions is hybridization in 6X SSC at about 45°C, followed by at least one wash in 0.2X SSC, 0.1% SDS at 60°C.
  • a further example of stringent hybridization conditions is hybridization in 6X SSC at about 45°C, followed by at least one wash in 0.2X SSC, 0.1% SDS at 65°C.
  • High stringent conditions include hybridization in 0.5 M sodium phosphate, 7% SDS at 65°C, followed by at least one wash at 0.2X SSC, 1% SDS at 65°C.
  • a region of a nucleic acid is used to indicate a smaller sequence within a larger sequence of nucleic acids.
  • a gene is a region of a chromosome, an exon is a region of a gene, etc.
  • polypeptide target e.g., a polypeptide product an AI response marker
  • specifically binds does not exclude some cross reactivity with undesireable polypeptides, as long as that cross reactivity does not interfere with the capability of the probe to provide a a useful measure of the presence of the given polypeptide target.
  • a probe that is capable of detecting the presence of a particular substance means that the probe may be used to detect the particular substance.
  • oliogonucelotide refers to a short sequence of nucleotides, e.g., 2-100 bases.
  • biological sample refers to a sample from a patient, which may be used for the purpose of identification, diagnosis, prediction, or monitoring.
  • Preferred samples include synovial fluid, blood, blood-derived product (such as buffy coat, serum, and plasma), lymph, urine, tear, saliva, hair bulb cells, cerebrospinal fluid, buccal swabs, feces, synovial fluid, synovial cells, sputum, or tissue samples (e.g., cartilage samples).
  • blood-derived product such as buffy coat, serum, and plasma
  • lymph urine
  • tear saliva
  • hair bulb cells cerebrospinal fluid
  • buccal swabs buccal swabs
  • feces synovial fluid
  • synovial cells synovial cells
  • sputum e.g., cartilage samples
  • AI agent refers to any therapeutic agent for treating a particular AI disease (i.e., uveitis, RA, PsA, AS, psoriasis or MS), provided that such agent is not an IL-17 antagonist.
  • AI agents consist of therapeutic agents for treating RA (“RA agent”), therapeutic agents for treating PsA (“PsA agent”), therapeutic agents for treating AS (“AS agent”), therapeutic agents for treating psoriasis (“psoriasis agent”), and therapeutic agents for treating MS (“MS agent”).
  • an RA agent, PsA agent, and AS agent may be, inter alia, an immunosuppressive agent, a DMARD, a pain-control drug, a steroid, a non-steroidal antiinflammatory drug (NSAID), a cytokine antagonist, a bone anabolic, a bone anti-resorptive, and combinations thereof.
  • an immunosuppressive agent e.g., a DMARD, a pain-control drug, a steroid, a non-steroidal antiinflammatory drug (NSAID), a cytokine antagonist, a bone anabolic, a bone anti-resorptive, and combinations thereof.
  • NSAID non-steroidal antiinflammatory drug
  • Representative agents include cyclosporin, retinoids, corticosteroids, propionic acid derivative, acetic acid derivative, enolic acid derivatives, fenamic acid derivatives, Cox-2 inhibitors, lumiracoxib, ibuprophen, cholin magnesium salicylate, fenoprofen, salsalate, difunisal, tolmetin, ketoprofen, flurbiprofen, oxaprozin, indomethacin, sulindac, etodolac, ketorolac, nabumetone, naproxen, valdecoxib, etoricoxib, MK0966; rofecoxib, acetominophen, Celecoxib, Diclofenac, tramadol, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefanamic acid, meclofenamic acid, flufenamic acid
  • fludrocortisone deoxycorticosterone, aldosterone, SB-681323, Rob 803, AZD5672, AD 452, SMP 114, HZT-501, CP-195,543, Doxycycline, vancomycin, CRx-102, AMG108, pioglitazone, SBI-087, SCIO-469, Cura-100, Oncoxin + Viusid, TwHF, PF-04171327, AZD5672,
  • LY2127399 E-551S-551, H-551, GSK3152314A, RWJ-445380, Tacrolimus (Prograf®), RADOOl, rapamune, rapamycin, fostamatinib, Fentanyl, XOMA 052, CNTO 136, JNJ 38518168, Imatimb, ATN-103, ISIS 104838, folic acid, folate, TNFa kinoid, MM-093, type II collagen, VX-509, AMG 827 70, masitimb (ABIOIO), LY2127399, cyclosponne, SB-681323, MK0663, NNC 0151-0000-0000, ATN-103, CCX 354-C, CAM3001, LX3305, Cetrore x, MDX-1342, TMI-005, MK0873, CDP870, Tranilast, CFlOl, mycophenolic acid (and esters thereof), VX-702
  • Preferred RA agents are DMARDs, such as methotrexate, and TNF alpha antagonists.
  • Preferred AS agents are NSAIDs, DMARDS, such as sulfasalazine, and TNF alpha antagonists.
  • Preferred PsA agents are DMARDS, such as cyclosporine, CTLA-4 blockers (e.g., CLTA4-Ig), alefacept, and TNF alpha antagonists.
  • psoriasis agents include, inter alia, topical and systemic agents, such as DMARDs, immunosuppressants, cytokine antagonists, antibiotics, and steroids.
  • Representative psoriasis agents include, e.g., salicylic acid, coal tar, Dovonex® (calcipotriene), Taclonex® (calcipotriene and betamethasone dipropionate), Tazorec® (tazarotene), pimecrolimus, tacrolimus, Vectical® (calcitriol), Zithranol-RR® (anthralin) and topical steroids (e.g., corticosteroids), retionoids such as Acitretin (Soriatane ®), cyclosporine, methotrexate, Hydrea® (hydroxyurea), isotretinoin, mycophenolate mofetil, mycophenolic acid, sulfasalazine, 6-thioguanine, fumarates
  • Preferred psoriasis agents are DMARDs (e.g., MTX and cyclosporine), IL-12/-23 antagonists (e.g., ustekinumab), CTLA-4 antagonists (e.g., CTLA4-Ig), and TNF-alpha antagonists.
  • DMARDs e.g., MTX and cyclosporine
  • IL-12/-23 antagonists e.g., ustekinumab
  • CTLA-4 antagonists e.g., CTLA4-Ig
  • TNF-alpha antagonists e.g., TNF-alpha antagonists.
  • MS agents are: interferon-beta [IFN- ⁇ ] la and lb: Betaferon®/ Avonex® /Rebif® /Extavia®; glatiramer acetate: Copaxone®; natalizumab: Tysabri® (selective adhesion molecule inhibitor); mitoxantrone: Novantrone® (chemotherapeutic); fingolimod
  • MS agents are available to modify the disease course, treat exacerbations (relapses), reduce the accumulation of lesions, prevent nerve damage, manage symptoms, and improve function and safety.
  • MS agents include masitinib, ELND002, atorvastatin, vitamin A, Cholecalciferol (Vitamin D3), alemtuzmab, Cyclophosphamide, Methylprednisolone, MIS416, RTL1000, Topamax, Duloxetine Hydrochloride (HCI),
  • Dalfampridine Ampyra, methylprednisolone, Daclizumab, CNTO 1275, Memantine, ATX-MS- 1467, GNbACl, SB683699 (firategrast), abatacept, L-Carnitine, Rituximab, BAF312, Inosine, Mangafodipir (Teslascan), Alemtuzumab; Fludarabine; Idebenone, levetiracetam, Androgel, Dalfampridine, MBP8298, CGP 77116, Rolipram, Ritalin, C105, Danfenacin (BAY79-4998), Lubiprostone, Dronabinol, Lithium Carbonate, donepezil, ONO-4641, LY2127399, Sativex, Myfortic® (mycophenolate sodium salt), Mycophenolate Mofetil, BIIB033, BIIB017, 3,4- diaminopyridine, ACTH, ML
  • glatiramer acetate natalizumab; mitoxantrone; fingolimod; and teriflunomide.
  • uveitis agents include corticosteroids, such as rimexolone, loteprednol, prednisolone, triamcinolone, fluocinolone, prednisolone, solumedrol, preferably loteprednol or flucinolone.
  • Aother uveitis agents include steroid-spering drugs such as calcineurin inhibitors (cyclosporine, tacrolimus, peimcrolimus), antimetabolities (methotrexate, myccophenolate mofetil), alkylating agents (cyclophosphamide, chlorambucil) and biologicals, (e.g., TNF antagonists (e.g., anti-TNF antibodies)).
  • Preferred uveitis agents are corticosteroids, calcineurin inhibitors (e.g., cyclosporine), antimetabolities (e.g., MTX), alkylating agents (e.g.,
  • compositions, regimens, processes, uses, methods and kits utilze an IL-17 antagonist, e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen- binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 receptor antibody or antigen-binding portion thereof).
  • IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen- binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 receptor antibody or antigen-binding portion thereof).
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises at least one immunoglobulin heavy chain variable domain (V H ) comprising hypervariable regions CDRl , CDR2 and CDR3, said CDRl having the amino acid sequence SEQ ID NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3.
  • V H immunoglobulin heavy chain variable domain
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises at least one immunoglobulin light chain variable domain (1 ⁇ 4 ) comprising hypervariable regions CDRl ', CDR2' and CDR3', said CDRl ' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO: 5 and said CDR3' having the amino acid sequence SEQ ID NO:6.
  • IL-17 binding molecule e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL- 17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises at least one immunoglobulin heavy chain variable domain (V H ) comprising hypervariable regions CDRl -x, CDR2-X and CDR3-X, said CDRl-x having the amino acid sequence SEQ ID NO: l 1, said CDR2-X having the amino acid sequence SEQ ID NO: 12, and said CDR3-X having the amino acid sequence SEQ ID NO: 13.
  • V H immunoglobulin heavy chain variable domain
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises at least one immunoglobulin V H domain and at least one immunoglobulin V L domain, wherein: a) the immunoglobulin V H domain comprises (e.g., in sequence): i) hypervariable regions CDR1, CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; or ii) hypervariable regions CDRl-x, CDR2-X and CDR3-X, said CDRl-x having the amino acid sequence SEQ ID NO: l 1, said CDR2-x having the amino acid sequence SEQ ID NO: 12, and said CDR3-x having the amino acid sequence SEQ ID NO: 13; and b) the immunoglobulin
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises: a) an immunoglobulin heavy chain variable domain (V H ) comprising the amino acid sequence set forth as SEQ ID NO: 8; b) an immunoglobulin light chain variable domain (V L ) comprising the amino acid sequence set forth as SEQ ID NO: 10; c) an immunoglobulin V H domain comprising the amino acid sequence set forth as SEQ ID NO: 8 and an immunoglobulin V L domain comprising the amino acid sequence set forth as SEQ ID NO: 10; d) an immunoglobulin V H domain comprising the hypervariable regions set forth as SEQ ID NO: l, SEQ ID NO:2, and SEQ ID NO: 3; e) an immunoglobulin V L domain comprising the hypervariable regions set forth as SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO:
  • amino acid sequences of the hypervariable regions of the secukinumab monoclonal antibody based on the Kabat definition and as determined by the X- ray analysis and using the approach of Chothia and coworkers, is provided in Table 2, below.
  • Table 2 Amino acid sequences of the hypervariable regions of the secukinumab monoclonal antibodies based on the Kabat definition and as determined by the X-ray analysis, using the approach of Chothia and coworkers.
  • the constant region domains preferably also comprise suitable human constant region domains, for instance as described in "Sequences of Proteins of
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises the three CDRs of SEQ ID NO: 10.
  • the IL-17 antagonist comprises the three CDRs of SEQ ID NO:8.
  • the IL-17 antagonist comprises the three CDRs of SEQ ID NO: 10 and the three CDRs of SEQ ID NO: 8.
  • CDRs of SEQ ID NO: 8 and SEQ ID NO: 10, according to both the Chothia and Kabat definition, may be found in Table 2.
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) comprises the light chain of SEQ ID NO: 15.
  • the IL-17 antagonist comprises the heavy chain of SEQ ID NO: 17.
  • the IL-17 antagonist comprises the light chain of SEQ ID NO: 15 and the heavy domain of SEQ ID NO: 17.
  • the IL-17 antagonist comprises the three CDRs of SEQ ID NO: 15.
  • the IL-17 antagonist comprises the three CDRs of SEQ ID NO: 17.
  • the IL-17 antagonist comprises the three CDRs of SEQ ID NO: 15 and the three CDRs of SEQ ID NO: 17.
  • CDRs of SEQ ID NO: 15 and SEQ ID NO: 17, according to both the Chothia and Kabat definition, may be found in Table 2.
  • the DNA encoding the light chain of secukinumab is set forth as SEQ ID NO: 14.
  • the DNA encoding the heavy chain of secukinumab is set forth as SEQ ID NO: 16.
  • Hypervariable regions may be associated with any kind of framework regions, though preferably are of human origin. Suitable framework regions are described in Kabat E.A. et al, ibid.
  • the preferred heavy chain framework is a human heavy chain framework, for instance that of the secukinumab antibody. It consists in sequence, e.g. of FRl (amino acid 1 to 30 of SEQ ID NO: 8), FR2 (amino acid 36 to 49 of SEQ ID NO: 8), FR3 (amino acid 67 to 98 of SEQ ID NO: 8) and FR4 (amino acid 117 to 127 of SEQ ID NO:8) regions.
  • another preferred heavy chain framework consists in sequence of FRl -x (amino acid 1 to 25 of SEQ ID NO: 8), FR2-x (amino acid 36 to 49 of SEQ ID NO: 8), FR3-x (amino acid 61 to 95 of SEQ ID NO: 8) and FR4 (amino acid 119 to 127 of SEQ ID NO:8) regions.
  • the light chain framework consists, in sequence, of FRl ' (amino acid 1 to 23 of SEQ ID NO: 10), FR2' (amino acid 36 to 50 of SEQ ID NO: 10), FR3' (amino acid 58 to 89 of SEQ ID NO: 10) and FR4' (amino acid 99 to 109 of SEQ ID NO: 10) regions.
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) is selected from a human anti IL- 17 antibody which comprises at least: a) an immunoglobulin heavy chain or fragment thereof which comprises a variable domain comprising in sequence the hypervariable regions CDR1, CDR2 and CDR3 and the constant part or fragment thereof of a human heavy chain; said CDR1 having the amino acid sequence SEQ ID NO: l, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; and b) an immunoglobulin heavy chain or fragment thereof which comprises a variable domain comprising in sequence the hypervariable regions CDR1, CDR2 and CDR3 and the constant part or fragment thereof of a human heavy chain; said CDR1 having the amino acid sequence SEQ ID NO: l, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said
  • immunoglobulin light chain or fragment thereof which comprises a variable domain comprising in sequence the hypervariable regions CDR1 ', CDR2', and CDR3' and the constant part or fragment thereof of a human light chain, said CDR1 ' having the amino acid sequence SEQ ID NO: 4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6.
  • the IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) is selected from a single chain binding molecule which comprises an antigen binding site comprising: a) a first domain comprising in sequence the hypervariable regions CDR1, CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: l, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; and b) a second domain comprising the hypervariable regions CDR1', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6; and c) a peptide linker which is bound either to the N-terminal extremity of the
  • an IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) for use in the disclosed methods may comprise a derivative of the IL-17 binding molecules set forth herein by sequnence (e.g., a pegylated version of secukinumab).
  • the V H or V L domain of an IL-17 antagonist, e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) for use in the disclosed methods may have V H or V L domains that are substantially identical to the the V H or V L domains set forth herein (e.g., those set forth in SEQ ID NO: 8 and 10).
  • a human IL-17 antibody disclosed herein may comprise a heavy chain that is substantially identical to that set forth as SEQ ID NO: 17 and/or a light chain that is substantially identical to that set forth as SEQ ID NO: 15.
  • a human IL-17 antibody disclosed herein may comprise a heavy chain that comprises SEQ ID NO : 17 and a light chain that comprises SEQ ID NO : 15.
  • a human IL- 17 antibody disclosed herein may comprise: a) one heavy chain which comprises a variable domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 8 and the constant part of a human heavy chain; and b) one light chain which comprises a variable domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 10 and the constant part of a human light chain.
  • an IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) for use in the disclosed methods may be an amino acid sequence variant of the reference IL-17 binding molecules set forth herein.
  • the inhibition of the binding of IL-17 to its receptor may be conveniently tested in various assays including such assays as described in WO 2006/013107.
  • assays including such assays as described in WO 2006/013107.
  • the reference and the derivative molecules exhibit, on a statistical basis, essentially identical IL-17 inhibitory activity in one of the assays referred to herein (see Example 1 of WO 2006/013107).
  • the IL-17 binding molecules disclosed herein typically have IC50S for the inhibition of human IL-17 on IL-6 production induced by human IL-17 in human dermal fibroblasts which are below about 10 nM, more preferably about 9, 8, 7, 6, 5, 4, 3, 2, or about 1 nM of that of, preferably substantially the same as, the IC50 of the corresponding reference molecule when assayed as described in Example 1 of WO 2006/013107.
  • the assay used may be an assay of competitive inhibition of binding of IL-17 by soluble IL-17 receptors (e.g. the human IL-17 R/Fc constructs of Example 1 of WO 2006/013107) and the IL- 17 antagonists of the disclosure.
  • the disclosure also includes IL-17 antagonists, e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) in which one or more of the amino acid residues of CDR1, CDR2, CDR3, CDRl-x, CDR2-X, CDR3-X, CDR1 , CDR2' or CDR3' or the frameworks, typically only a few (e.g., 1-4), are changed; for instance by mutation, e.g., site directed mutagenesis of the corresponding DNA sequences.
  • the disclosure includes the DNA sequences coding for such changed IL-17 antagonists.
  • IL-17 binding molecules e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab
  • IL-17 antibodies capable of inhibit
  • the IL-17 antagonist e.g., IL-17 antibody, e.g., secukinumab
  • IL-17 antibody binds to an epitope of mature human IL-17 comprising Leu74, Tyr85, His86, Met87, Asn88, Vall24, Thrl25, Prol26, Ilel27, Vall28, Hisl29.
  • the IL-17 antibody e.g., secukinumab
  • the IL-17 antibody e.g., secukinumab
  • the residue numbering scheme used to define these epitopes is based on residue one being the first amino acid of the mature protein (ie., IL-17A lacking the 23 amino acid N-terminal signal peptide and beginning with Glycine).
  • the sequence for immature IL-17A is set forth in the Swiss-Prot entry Q 16552.
  • the IL-17 antibody has a KD of about 100-200 pM.
  • the IL-17 antibody has an IC 50 of about 0.4 nM for in vitro neutralization of the biological activity of about 0.67 nM human IL-17 A.
  • the absolute bioavailability of subcutaneously (s.c.) administered IL-17 antibody has a range of about 60 - about 80%, e.g., about 76%.
  • the IL-17 antagonist e.g., an IL-17 binding molecule (e.g., an IL-17 antibody, such as secukinumab) or an IL-17 receptor binding molecule (e.g., an IL-17 receptor antibody) has an elimination half-life of about 4 weeks (e.g., about 23 to about 35 days, about 23 to about 30 days, e.g., about 30 days).
  • the IL-17 antagonist e.g., an IL-17 binding molecule (e.g., an IL-17 antibody, such as secukinumab) or an IL-17 receptor binding molecule (e.g., an IL-17 receptor antibody) has a Tmax of about 7-8 days.
  • IL-17 antagonists e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof) for use in the disclosed methods, uses, kits, etc. are human antibodies, especially secukinumab as described in Examples 1 and 2 of WO 2006/013107.
  • Secukinumab is a recombinant high-affinity, fully human monoclonal anti-human interleukin-17A (IL-17A, IL-17) antibody of the IgGi/ ⁇ isotype that is currently in clinical trials for the treatment of immune-mediated inflammatory conditions.
  • Secukinumab (see, e.g., WO2006/013107 and WO2007/117749) has a very high affinity for IL- 17, i.e., a KD of about 100-200 pM and an IC 50 for in vitro neutralization of the biological activity of about 0.67 nM human IL-17A of about 0.4 nM.
  • secukinumab inhibits antigen at a molar ratio of about 1 : 1.
  • This high binding affinity makes the secukinumab antibody particularly suitable for therapeutic applications.
  • secukinumab has a very long half life, i.e., about 4 weeks, which allows for prolonged periods between administration, an exceptional property when treating chronic life-long disorders, such as AI disorders.
  • IL-17 antibodies for use in the disclosed methods, kits and uses are those set forth in US Patent Nos: 8,057,794; 8,003,099; 8,110,191; and 7,838,638 and US Published Patent Application Nos: 20120034656 and 20110027290.
  • the disclosed methods are useful for the treatment, prevention, or amelioration of AI diseases, as well as predicting the likelihood of an AI disease patient's response to treatment with an IL-17 antagonist, e.g., secukinumab. These methods employ, inter alia, determining whether a patient has an AI response marker in a sample from the patient or determining the level of ERAP1 expression, level of ERAP1 protein or level of ERAP1 activity.
  • a biological sample from the patient may be assayed for the presence of an AI response marker by any applicable conventional means, which will be selected depending on whether the particular marker falls within an exon, an intron, a non-coding portion of mRNA or a non- conding genomic sequence.
  • a protein e.g., blood, synovial fluid, buffy coat, serum, plasma, lymph, feces, urine, tear, saliva, cerebrospinal fluid, buccal swabs, sputum, or tissue.
  • Various sources within a biological sample may be used in the disclosed methods, e.g., one may assay genomic DNA obtained from a biological sample to detect an AI response marker, or one may assay products of a an AI response marker, e.g., nucleic acid products (e.g., DNA, pre-mRNA, mRNA, micro RNAs, etc.) and polypeptide products (e.g., expressed proteins) obtained from a biological sample.
  • nucleic acid products e.g., DNA, pre-mRNA, mRNA, micro RNAs, etc.
  • polypeptide products e.g., expressed proteins
  • SNPs of Table 1 are useful for predicting certain patient's response to treatment by IL-17 antagonism (e.g., using secukinumab).
  • SNPs in Table 1 most are found in genomic DNA and introns, such that a patient's allelic status may be determined by interrogating, e.g., pre-mRNA or genomic DNA.
  • the presence of the rs72773968, rsl7482078 and rs800292 SNPs may be determined by assaying genomic DNA, RNA and/or protein sequence, given that these SNPs falls within exons (e.g., exonss of the ERAP1 gene and an exon of the CFH gene) and produce missense amino acid changes. Accordingly, a skilled artisan will understand that one may identify whether a subject has a given AI response marker by assaying a nucleic acid product of an AI response marker, a polypeptide product of an AI response marker, or an equivalent genetic marker of an AI response marker, as appropriate for a given SNP (see Table 1). In preferred embodiments, a genomic sequence of an AI response marker is analyzed to determine whether a subject has an AI response marker.
  • ERAP1 expression levels of ERAPl protein and/or levels of ERAPl activity may be useful to predict improved response to IL-17 antagonism (e.g., secukinumab) for uveitis, psoriasis, MS, PsA, and RA patients (especially uveitis, MS, PsA and RA patients), while increased levels of ERAPl expression, levels of ERAPl protein and/or levels of ERAPl activity may be useful to predict a weaker response to IL-17 antagonism (e.g., secukinumab) for uveitis, psoriasis, MS, PsA, and RA patients (especially uveitis, MS, PsA and RA patients).
  • IL-17 antagonism e.g., secukinumab
  • a weaker response to IL-17 antagonism e.g., secukinumab
  • Levels of ERAPl expression, ERAPl protein and/or ERAPl activity may be directly measured by various techniques disclosed herein.
  • any ERAPl polymorphism e.g., translocations, insertions, substitutions, deletions, SNP, etc., occurring in ERAP1 enhancers, exons, introns, promoters, 5' UTR, 3'UTR, etc.
  • a change in the level of ERAP1 expression, level of ERAP1 protein, and/or level of ERAP1 activity is also expected to be useful to predict an increased or decreased likelihood of an AI patient responding to treatment with an IL-17 antagonist, e.g., secukinumab.
  • genotyping techniques employ one or more oligonucleotides that are complementary to a region containing, or adjacent to, the polymorphic site (e.g., SNP) of interest.
  • the sequence of an oligonucleotide used for genotyping a particular polymorphic site of interest is typically designed based on a context sequence or a reference sequence.
  • DNA for SNP detection can be prepared from a biological sample by methods well known in the art, e.g., phenol/chloroform extraction, PUREGENE DNA® purification system from GentAS Systems (Qiagen, CA).
  • Detection of a DNA sequence may include examining the nucleotide(s) located at either the sense or the anti-sense strand within that region.
  • the presence of polymorphisms in a patient may be detected from DNA (genomic or cDNA) obtained from PCR using sequence-specific probes, e.g., hydrolysis probes from Taqman, Beacons, Scorpions; or hybridization probes that detect the marker or polymorphism.
  • sequence specific probes may be designed such that they specifically hybridize to the genomic DNA for the alleles of interest or, in some cases, an RNA of interest.
  • Primers and probes for polymorphic sites may be designed based on context sequences found in the NCBI SNP database available at: www.ncbi.nlm.nih.gov/snp. These probes may be labeled for direct detection or contacted by a second, detectable molecule that specifically binds to the probe.
  • the PCR products also can be detected by DNA-binding agents. Said PCR products can then be subsequently sequenced by any DNA sequencing method available in the art. Alternatively the presence of allele can be detected by sequencing using any sequencing methods such as, but not limited to, Sanger-based sequencing, pyrosequencing or next generation sequencing (Shendure J. and Ji, H., Nature Biotechnology (1998), Vol. 26, Nr 10, pages 1135-1145).
  • Optimised allelic discrimination assays for SNPs may be purchased from Applied Biosystems (Foster City, California, USA).
  • Various techniques can be applied to interrogate a particular polymorphism (e.g., SNP), including, e.g., hybridization-based methods, such as dynamic allele-specific hybridization (DASH) genotyping, polymorphic site (e.g., SNP) detection through molecular beacons
  • a particular polymorphism e.g., SNP
  • hybridization-based methods such as dynamic allele-specific hybridization (DASH) genotyping
  • DASH dynamic allele-specific hybridization
  • SNP polymorphic site detection through molecular beacons
  • RFLP restriction fragment length polymorphism
  • PCR-based methods e.g., Tetra-primer ARMS-PCR
  • Invader assays Olivier M. (2005) Mutat Res. 573(1-2): 103-10
  • primer extension assays incorporated into detection formats, e.g., MALDI-TOF Mass spectrometry, electrophoresis, blotting, and ELISA-like methods
  • TaqMan® assays oligonucleotide ligase assays
  • other post-amplification methods e.g., analysis of single strand conformation polymorphism (Costabile et al.
  • TGGE temperaure gradient gel electrophoresis
  • DNA mismatch-binding protein assays e.g., MutS protein from Thermus aquaticus binds different single nucleotide mismatches with different affinities and can be used in capillary electrophoresis to differentiate all six sets of mismatches
  • SNPLex® proprietary SNP detecting system available from Applied Biosystems
  • capillary electrophoresis mass spectrometry
  • various sequencing methods e.g., pyrosequencing and next generation sequencing, etc.
  • kits for SNP genotyping include, e.g., Fluidigm Dynamic Array® IFCs (Fluidigm), TaqMan® SNP Genotyping Assay (Applied Biosystems), MassARRAY® iPLEX Gold
  • the presence of a polymorphic site (e.g., SNP) in a patient is detected using a hybridization assay.
  • a hybridization assay the presence of the genetic marker is determined based on the ability of the nucleic acid from the sample to hybridize to a complementary nucleic acid molecule, e.g., an oligonucleotide probe.
  • a variety of hybridization assays are available.
  • hybridization of a probe to the sequence of interest is detected directly by visualizing a bound probe, e.g., a Northern or Southern assay. In these assays, DNA (Southern) or RNA (Northern) is isolated.
  • the DNA or RNA is then cleaved with a series of restriction enzymes that cleave infrequently in the genome and not near any of the markers being assayed.
  • the DNA or RNA is then separated, e.g., on an agarose gel, and transferred to a membrane.
  • a labeled probe or probes e.g., by incorporating a radionucleotide or binding agent (e.g., SYBR® Green), is allowed to contact the membrane under low-, medium- or high- stringency conditions. Unbound probe is removed and the presence of binding is detected by visualizing the labeled probe.
  • arrays e.g., the MassARRAY® system (Sequenom, San Diego, California, USA) may be used to genotype a subject.
  • Genoing methods may also be modified for use in genotyping.
  • Such traditional methods include, e.g., DNA amplification techniques such as PCR and variants thereof, direct sequencing, SSO hybridization coupled with the Luminex xMAP® technology, SSP typing, and SBT.
  • Sequence-Specific Oligonucleotide (SSO) typing uses PCR target amplification, hybridization of PCR products to a panel of immobilized sequence-specific oligonucleotides on the beads, detection of probe-bound amplified product by color formation followed by data analysis.
  • SSO Sequence-Specific Oligonucleotide
  • LABType® SSO is a reverse SSO (rSSO) DNA typing solution that uses sequence-specific oligonucleotide (SSO) probes and color-coded microspheres to identify HLA alleles.
  • SSO sequence-specific oligonucleotide
  • the target DNA is amplified by polymerase chain reactions (PCR) and then hybridized with the bead probe array.
  • PCR polymerase chain reactions
  • the assay takes place in a single well of a 96-well PCR plate; thus, 96 samples can be processed at one time.
  • Sequence Specific Primers (SSP) typing is a PCR based technique which uses sequence specific primers for DNA based typing.
  • the SSP method is based on the principle that only primers with completely matched sequences to the target sequences result in amplified products under controlled PCR conditions. Allele sequence-specific primer pairs are designed to selectively amplify target sequences which are specific to a single allele or group of alleles. PCR products can be visualized on agarose gel. Control primer pairs that matches non-allelic sequences present in all samples act as an internal PCR control to verify the efficiency of the PCR amplification.
  • low, medium and high resolution genotyping with the described sequence-specific primer typing may be performed using various commercially available kits, such as the Olerup SSPTM kits (Olerup, PA) or (Invitrogen) or Allset andTMGold DQA1 Low resolution SSP (Invitrogen).
  • Sequence Based Typing is based on PCR target amplification, followed by sequencing of the PCR products and data analysis.
  • RNA e.g., mature mRNA, pre-mRNA
  • RNA can also be used to determine the presence of particular polymorphisms (see Table 1).
  • Analysis of the sequence of mRNA transcribed from a given gene can be performed using any known method in the art including, but not limited, to Northern blot analysis, nuclease protection assays (NPA), in situ hybridization, reverse transcription-polymerase chain reaction (RT-PCR), RT-PCR ELISA, TaqMan-based quantitative RT-PCR (probe-based quantitative RT-PCR) and SYBR green-based quantitative RT-PCR.
  • NPA nuclease protection assays
  • RT-PCR reverse transcription-polymerase chain reaction
  • RT-PCR ELISA reverse transcription-polymerase chain reaction
  • TaqMan-based quantitative RT-PCR probe-based quantitative RT-PCR
  • SYBR green-based quantitative RT-PCR SYBR green-based quantitative RT-PCR.
  • detection of mRNA levels involves contacting the isolated mRNA with an oligonucleotide that can hybridize to mRNA encoded by an AI response marker.
  • the nucleic acid probe can typically be, for example, a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, or 100 nucleotides in length and sufficient to specifically hybridize under stringent conditions to the mRNA. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.
  • the RNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated RNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose.
  • Amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. In general, amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length.
  • PCR products can be detected by any suitable method including, but not limited to, gel electrophoresis and staining with a DNA-specific stain or hybridization to a labeled probe.
  • the level of expression of a gene may be determined by measuring RNA (or reverse transcribed cDNA) levels using various techniques, e.g., a PCR-based assay, reverse- transcriptase PCR (RT-PCR) assay, Northern blot, etc. Quantitative RT-PCR with standardized mixtures of competitive templates can also be utilized.
  • the presence of a polymorphism in a patient can be determined by analyzing polypeptide products of the AI response markers (see Table 1). Detection of polypeptide products can be performed using any known method in the art including, but not limited, to immunocytochemical staining, ELISA, flow cytometry, Western blot,
  • decreased levels of ERAPl protein may be predictive of an improved response to IL-17 antagonism (e.g., secukinumab treatment) for uveitis, AS, MS, PsA, RA and psoriasis patients (preferably uveitis, MS, PsA, RA and psoriasis patients, most preferably uveitis, MS, PsA, and RA patients).
  • IL-17 antagonism e.g., secukinumab treatment
  • uveitis, AS, MS, PsA, RA and psoriasis subjects preferably uveitis, MS, PsA, RA and psoriasis patients, most preferably uveitis, MS, PsA, and RA patients
  • IL-17 antagonists e.g., secukinumab
  • One method for detecting polypeptide products in a sample is by means of a probe that is a binding protein capable of interacting specifically with a marker protein (e.g., an antibody capable of binding ERAPl protein).
  • a marker protein e.g., an antibody capable of binding ERAPl protein.
  • labeled antibodies, binding portions thereof, or other binding partners can be used.
  • the antibodies can be monoclonal or polyclonal in origin, or may be biosynthetically produced.
  • the binding partners may also be naturally occurring molecules or synthetically produced.
  • the amount of complexed proteins is determined using standard protein detection methodologies described in the art. A detailed review of
  • Direct labels include fluorescent or luminescent tags, metals, dyes, radionucleides, and the like, attached to the antibody.
  • Indirect labels include various enzymes well known in the art, such as alkaline phosphatase, hydrogen peroxidase and the like.
  • polypeptide products if present, are immobilized and incubated with a labeled antibody. The labeled antibody binds to the immobilized target molecule. After washing to remove unbound molecules, the sample is assayed for the label.
  • immobilized antibodies specific for the proteins or polypeptides is also contemplated by the present disclosure.
  • the antibodies can be immobilized onto a variety of solid supports, such as magnetic or chromatographic matrix particles, the surface of an assay place (such as microtiter wells), pieces of a solid substrate material (such as plastic, nylon, paper), and the like.
  • An assay strip can be prepared by coating the antibody or a plurality of antibodies in an array on solid support. This strip can then be dipped into the test sample and then processed quickly through washes and detection steps to generate a measurable signal, such as a colored spot.
  • immobilized polypeptide products of an AI response marker or ERAP1 protein may be incubated with an unlabeled antibody.
  • the unlabeled antibody complex if present, is then bound to a second, labeled antibody that is specific for the unlabeled antibody.
  • the sample is washed and assayed for the presence of the label.
  • the choice of marker used to label the antibodies will vary depending upon the application. However, the choice of the marker is readily determinable to one skilled in the art.
  • the antibodies may be labeled with a radioactive atom, an enzyme, a chromophoric or fluorescent moiety, or a colorimetric tag. The choice of tagging label also will depend on the detection limitations desired.
  • Enzyme assays typically allow detection of a colored product formed by interaction of the enzyme-tagged complex with an enzyme substrate.
  • radioactive atoms include 32 P, 125 1, 3 H, and 14 P.
  • enzymes include horseradish peroxidase, alkaline phosphatase, beta- galactosidase, and glucose-6-phosphate dehydrogenase.
  • chromophoric moieties include fluorescein and rhodamine. The antibodies may be conjugated to these labels by methods known in the art.
  • enzymes and chromophoric molecules may be conjugated to the antibodies by means of coupling agents, such as dialdehydes, carbodiimides, dimaleimides, and the like.
  • conjugation may occur through a ligand-receptor pair.
  • suitable ligand-receptor pairs include, for example, biotin-avidin or -streptavidin, and antibody-antigen.
  • the present disclosure contemplates the use of a sandwich technique for detecting polypeptide products in biological samples.
  • the technique requires two antibodies capable of binding the protein of interest: e.g., one immobilized onto a solid support and one free in solution, but labeled with some easily detectable chemical compound.
  • chemical labels that may be used for the second antibody include but are not limited to radioisotopes, fluorescent compounds, and enzymes or other molecules which generate colored or
  • the presence of polypeptide products in a sample is detected by
  • radioimmunoassays or enzyme-linked immunoassays or enzyme-linked immunoassays, competitive binding enzyme-linked immunoassays, dot blot, Western blot, chromatography, preferably high performance liquid chromatography (HPLC), or other assays known in the art.
  • Specific immunological binding of the antibody to the protein or polypeptide can be detected directly or indirectly.
  • Dot blotting is routinely practiced by the skilled artisan to detect a desired protein using an antibody as a probe (Promega Protocols and Applications Guide, Second Edition, 1991, Page 263, Promega Corporation). Samples are applied to a membrane using a dot blot apparatus. A labeled probe is incubated with the membrane, and the presence of the protein is detected.
  • Western blot analysis is well known to the skilled artisan (Sambrook et al, Molecular Cloning, A Laboratory Manual, 1989, Vol. 3, Chapter 18, Cold Spring Harbor Laboratory).
  • the sample is separated by SDS-PAGE.
  • the gel is transferred to a membrane.
  • the membrane is incubated with labeled antibody for detection of the desired protein.
  • the assays described above involve steps such as but not limited to, immunoblotting, immunodiffusion, Immunoelectrophoresis, or immunoprecipitation.
  • an automatic analyzer is used to determine the presence of an AI response marker.
  • the level ERAPl activity may be assayed by various methods disclosed in the art, e.g., via the methods set forth in Kochan et al. (2011) Proc Natl Acad Sci U S A. 108(19):7745-50.
  • the level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity from a patient may be compared to the level of ERAPl expression, level of ERAPl protein, and level of ERAPl activity from a control.
  • the control may be a reference level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity derived from subjects (e.g., uveitis, PsA, RA, psoriasis, RA patients) known to respond well to treatment with an IL-17 antagonist (e.g., secukinumab) or subjects known to respond poorly to treatment with an IL-17 antagonist (e.g., secukinumab), as the case may be.
  • subjects e.g., uveitis, PsA, RA, psoriasis, RA patients
  • an IL-17 antagonist e.g., secukinumab
  • subjects known to respond poorly to treatment with an IL-17 antagonist e.g., secukinumab
  • a control level of expression may be derived from biological samples from reference subjects (i.e., uveitis, PsA, RA, psoriasis, RA subjects known to respond well to treatment with an IL-17 antagonist (e.g., secukinumab) or subjects known to respond poorly to treatment with an IL-17 antagonist (e.g., secukinumab)), or may simply be a numerical standard (e.g., mean, median, range, [+/- standard deviation]) previously derived from reference subjects.
  • control is a reference level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity derived from a subject known to respond poorly to treatment with an IL-17 antagonist and the level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity (as the case may be) from the patient is compared to this control, wherein a decreased level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity in the patient (relative to a control) provides an indication that the patient will have an increased likelihood of responding to treatment with the IL-17 antagonist (e.g., secukinumab).
  • IL-17 antagonist e.g., secukinumab
  • control is a reference level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity derived from a subject known to respond well to treatment with an IL-17 antagonist and the level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity from the patient to be treated is compared to this control, wherein a similar (e.g., statistically similar) level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity in the patient (relative to a control) provides an indication that the patient will have an increased likelihood of responding to treatment with the IL-17 antagonist (e.g., secukinumab).
  • a similar (e.g., statistically similar) level of ERAPl expression, level of ERAPl protein, or level of ERAPl activity in the patient provides an indication that the patient will have an increased likelihood of responding to treatment with the IL-17 antagonist (e.g., secukinumab).
  • An AI response marker or an ERAPl polymorphism that results in decreased level of ERAPl expression, level of ERAPl protein and/or level of ERAPl activity can also be identified by detecting an equivalent genetic marker thereof, which can be, e.g., another polymorphic site (e.g., SNP), a microsatellite marker, another allele or other kinds of genetic polymorphisms.
  • an equivalent genetic marker thereof can be, e.g., another polymorphic site (e.g., SNP), a microsatellite marker, another allele or other kinds of genetic polymorphisms.
  • the presence of a genetic marker on the same haplotype as an AI response marker or an ERAPl polymorphism, rather than an AI response marker or an ERAPl polymorphism per se may be indicative of a patient's likelihood for responding to treatment with an IL-17 antagonist.
  • Two particular alleles at different loci on the same chromosome are said to be in linkage disequilibrium (LD) if the presence of one of the alleles at one locus tends to predict the presence of the other allele at the other locus.
  • Such variants which are referred to herein as linked variants, or proxy variants, may be any type of variant (e.g., a SNP, insertion or deletion) that is in high LD with the better response allele of interest.
  • the candidate linked variant may be an allele of a polymorphism that is currently known.
  • Other candidate linked variants may be readily identified by the skilled artisan using any technique well-known in the art for discovering polymorphisms.
  • the degree of LD between alleles of interest and a candidate linked variant may be determined using any LD measurement known in the art.
  • LD patterns in genomic regions are readily determined empirically in appropriately chosen samples using various techniques known in the art for determining whether any two alleles (e.g., between nucleotides at different PSs) are in linkage disequilibrium (see, e.g., GENETIC DATA ANALYSIS II, Weir, Sineuer Associates, Inc. Publishers, Sunderland, MA 1996). The skilled artisan may readily select which method of determining LD will be best suited for a particular population sample size and genomic region.
  • r is the measure of how well an allele X at a first locus predicts the occurrence of an allele Y at a second locus on the same chromosome. The measure only reaches 1.0 when the prediction is perfect (e.g. X if and only if Y).
  • the locus of the linked variant is in a genomic region of about 200 kilobases, more preferably 100 kilobases, more preferably about 10 kb that spans one of the polymorphic sites disclosed herein.
  • Other linked variants are those in which the LD with the better response allele has a r2 value, as measured in a suitable reference population, of at least 0.75, more preferably at least 0.80, even more preferably at least 0.85 or at least 0.90, yet more preferably at least 0.95, and most preferably 1.0.
  • the reference population used for this r measurement may be the general population, a population using an IL-17 antagonist, a population diagnosed with a particular condition for which the IL-17 antagonists shows efficacy (such as an RA, PsA, AS, psoriasis, or MS patient) or a population whose members are self-identified as belonging to the same ethnic group, such as Caucasian, African American, Hispanic, Latino, Native American and the like, or any combination of these categories.
  • the reference population reflects the genetic diversity of the population of patients to be treated with the IL-17 antagonist.
  • an array is provided to which probes that correspond in sequence to gene products, e.g., genomic DNA, cDNAs, mRNAs, cRNAs, polypeptides and fragments thereof, can be specifically hybridized or bound at a known position.
  • probes that correspond in sequence to gene products e.g., genomic DNA, cDNAs, mRNAs, cRNAs, polypeptides and fragments thereof, can be specifically hybridized or bound at a known position.
  • the level of ERAPl expression, the level of ERAPl protein, or the level of ERAPl activity may be made by consulting a data repository that contains sufficient information on the patient's genetic composition to determine whether the patient has the marker of interest.
  • the data repository lists the genotype present (or absent) in the individual.
  • the data repository could include the individual's patient records, a medical data card, a file (e. g., a flat ASCII file) accessible by a computer or other electronic or non-electronic media on which appropriate information or genetic data can be stored.
  • a medical data card is a portable storage device such as a magnetic data card, a smart card, which has an on-board processing unit and which is sold by vendors such as Siemens of Kunststoff Germany, or a flash-memory card.
  • the data repository is a file accessible by a computer; such files may be located on various media, including: a server, a client, a hard disk, a CD, a DVD, a personal digital assistant such as a smart phone, Palm Pilot, a tape recorder, a zip disk, the computer's internal ROM (read-only-memory) or the internet or worldwide web.
  • Other media for the storage of files accessible by a computer are available to skilled artisans.
  • ERAPl expression Typically, once levels of ERAPl expression, levels of ERAPl protein/activity, or the presence of an AI response marker or ERAPl polymorphism is determined, physicians or genetic counselors or patients or other researchers may be informed of the result. Specifically the result can be cast in a transmittable form of information that can be communicated or transmitted to other researchers or physicians or genetic counselors or patients. Such a form can vary and can be tangible or intangible.
  • the result in the individual tested can be embodied in descriptive statements, diagrams, photographs, charts, images or any other visual forms. For example, images of gel electrophoresis of PCR products can be used in explaining the results. Diagrams showing where a variant occurs in an individual's allele are also useful in indicating the testing results.
  • Statements regarding levels ⁇ ERAPl expression, levels of ERAPl protein/activity, or the presence of an AI response marker or ERAPl polymorphism are also useful in indicating the testing results.
  • These statements and visual forms can be recorded on a tangible media such as papers, computer readable media such as floppy disks, compact disks, etc., or on an intangible media, e.g., an electronic media in the form of email or website on internet or intranet.
  • the result can also be recorded in a sound form and transmitted through any suitable media, e.g., analog or digital cable lines, fiber optic cables, etc., via telephone, facsimile, wireless mobile phone, internet phone and the like.
  • the information and data on a test result can be produced anywhere in the world and transmitted to a different location.
  • the information and data on a test result may be generated and cast in a transmittable form as described above.
  • the test result in a transmittable form thus can be imported into the U.S.
  • the present disclosure also encompasses a method for producing a transmittable form of information containing levels of ERAPl expression, levels of ERAPl protein/activity, or the presence of an AI response marker or ERAPl polymorphism in an individual.
  • This form of information is useful for predicting the responsiveness of a patient having uveitis, RA, AS, PsA, psoriasis, or MS to treatment with an IL-17 antagonist, for selecting a course of treatment based upon that information, and for selectively treating a patient based upon that information.
  • a) the presence of the at least one AI response marker is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist; and b) the absence of the at least one AI response marker is indicative of a decreased likelihood that the patient will respond to treatment with the IL-17 antagonist, wherein the AI disease is selected from the group consisting of RA, PsA, AS, psoriasis, and MS, and further wherein: i) if the AI disease is RA, then the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs72773968 response
  • the method further comprises the step of obtaining the biological sample from the patient, wherein the step of obtaining is performed prior to the step of assaying.
  • the at least one AI response marker is detected by assaying the biological sample for a nucleic acid product of the at least one AI response marker, a polypeptide product of the at least one AI response marker, or an equivalent genetic marker of the at least one AI response marker.
  • the AI response marker is detected by assaying the biological sample for a genomic sequence of the at least one AI response marker.
  • the biological sample is selected from the group consisting of synovial fluid, blood, serum, feces, plasma, urine, tear, saliva, cerebrospinal fluid, a leukocyte sample and a tissue sample.
  • a patient having an AI disease selected from uveitis, RA, PsA, AS, psoriasis, and MS preferably selected from uveitis, MS, RA, PsA and AS patients
  • an IL-17 antagonist e.g., secukinumab
  • detecting the level of ERAP1 expression e.g., mRNA, cDNA, etc.
  • the level of ERAPl protein, and/or the level of ERAPl activity in a biological sample from the patient relative to a control wherein a decreased level of ERAPl expression, decreased level of ERAPl protein, and/or a decreased level of ERAPl activity relative to the control is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist (e.g., secukinumab).
  • the control is a reference level of ERAPl expression, level of ERAPl protein,
  • a patient having an AI disease selected from uveitis, RA, PsA, AS, psoriasis, and MS preferably selected from uveitis, MS, RA, PsA and AS patients
  • an IL-17 antagonist e.g., secukinumab
  • detecting the level of ERAPl expression e.g., mRNA, cDNA, etc.
  • a similar level of ERAPl expression, similar level of ERAPl protein, or a similar level of ERAPl activity relative to the control is indicative of an increased likelihood that the patient will respond to treatment with the IL-17 antagonist (e.g., secukinumab).
  • the control is a reference level of ERAPl expression, level of ERAPl protein, or level
  • the level of ERAPl expression, the level of ERAPl protein, or the level of ERAPl activity is measured by assaying the biological sample from the patient for an ERAPl polymorphism that results in decreased ERAPl expression, decreased level of ERAPl protein, and/or decreased level of ERAPl activity relative to the control.
  • the presence of the at least one AI response marker, the level of ERAPl expression, or level of ERAPl protein/activity is detected by a technique selected from the group consisting of Northern blot analysis, polymerase chain reaction (PCR), reverse transcription-polymerase chain reaction (RT-PCR), TaqMan-based assays, direct sequencing, dynamic allele-specific hybridization, high-density oligonucleotide SNP arrays, restriction fragment length polymorphism (RFLP) assays, primer extension assays, oligonucleotide ligase assays, analysis of single strand conformation polymorphism, temperature gradient gel electrophoresis (TGGE), denaturing high performance liquid chromatography, high-resolution melting analysis, DNA mismatch-binding protein assays, SNPLex®, capillary electrophoresis, Southern Blot, immunoassays, immunohistochemistry, ELISA, flow cytometry, Western blot, HPLC, and mass spectrome
  • the AI disease is RA. In some embodiments, the AI disease is PsA. In some embodiments, the AI disease is AS. In some embodiments, the AI disease is psoriasis. In some embodiments, the AI disease is MS. In some embodiments, the AI disease is uveitis.
  • the IL-17 antagonist is an IL-17 binding molecule or an IL-17 receptor binding molecule. In some embodiments, the IL-17 binding molecule or IL-17 receptor binding molecule is an IL-17 binding molecule. In some embodiments, the IL-17 binding molecule is an IL-17 antibody or antigen-binding portion thereof. In some embodiments of the disclosed methods and uses, the IL-17 antibody is a human antibody. In some embodiments of the disclosed methods and uses, the human IL-17 antibody is secukinumab.
  • the disclosed methods allow clinicians to provide a personalized therapy for AI disease (i.e., uveitis, RA, PsA, AS, MS, and psoriasis) patients, i.e., they allow determination of whether to selectively treat the patient with an IL-17 antagonist (e.g., secukinumab) or whether to selectively treat the patient with an AI agent (e.g., NSAIDs, TNF alpha antagonists, DMARDs or corticosteroids).
  • an IL-17 antagonist e.g., secukinumab
  • an AI agent e.g., NSAIDs, TNF alpha antagonists, DMARDs or corticosteroids.
  • a clinician can maximize the benefit and minimize the risk of IL-17 antagnoism in the entire population of patients afflicted with AI disease (i.e., uveitis, RA, PsA, AS, MS, and psoriasis).
  • AI disease i.e., uveitis, RA, PsA, AS, MS, and psoriasis.
  • IL-17 antagonists e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof) are useful for the treatment, prevention, or amelioration of AI disease (i.e., uveitis, RA, PsA, AS, MS, and psoriasis) (e.g., signs and symptoms & structural changes, preventing further joint erosion, improving joint structure, etc.) as disclosed herein, particularly in patients that have an AI response marker.
  • AI disease i.e., uveitis, RA, PsA, AS, MS, and psoriasis
  • the IL-17 antagonists e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen- binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof), may be used in vitro, ex vivo, or incorporated into pharmaceutical compositions and administered to individuals (e.g., human patients) in vivo to treat, ameliorate, or prevent AI, e.g., in patients who have an AI response marker, or decreased levels oiERAPl expression and/or levels of ERAP1 protein/activity.
  • individuals e.g., human patients
  • AI e.g., in patients who have an AI response marker, or decreased levels oiERAPl expression and/or levels of ERAP1 protein/activity.
  • a pharmaceutical composition will be formulated to be compatible with its intended route of administration (e.g., oral compositions generally include an inert diluent or an edible carrier).
  • routes of administration include parenteral (e.g., intravenous), intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • parenteral e.g., intravenous
  • intradermal e.g., subcutaneous
  • oral e.g., inhalation
  • transdermal topical
  • transmucosal e.g., transmucosal
  • rectal administration e.g., rectal administration.
  • the pharmaceutical compositions compatible with each intended route are well known in the art.
  • the IL-17 antagonists may be used as a pharmaceutical composition when combined with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier may contain, in addition to an IL-17 antagonist, carriers, various diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The characteristics of the carrier will depend on the route of administration.
  • the pharmaceutical compositions for use in the disclosed methods may also contain additional therapeutic agents for treatment of the particular targeted disorder.
  • a pharmaceutical composition may also include antiinflammatory agents. Such additional factors and/or agents may be included in the pharmaceutically acceptable carrier.
  • IL-17 binding molecules e.g., IL- 17 antibody or antigen-binding portion thereof, e.g., secukinumab
  • IL-17 receptor binding molecules e.g., IL-17 receptor antibody or antigen-binding portion thereof
  • compositions for use in the disclosed methods may be manufactured in conventional manner.
  • the pharmaceutical composition is provided in lyophilized form.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • albumin a suitable concentration is from 0.5 to 4.5% by weight of the saline solution.
  • Other formulations comprise liquid or lyophilized formulation.
  • Antibodies e.g., antibodies to IL-17
  • the IL-17 antagonist e.g., IL-17 antibody, e.g., secukinumab
  • Suitable lyophilisate formulations can be reconstituted in a small liquid volume (e.g., 2ml or less) to allow subcutaneous administration and can provide solutions with low levels of antibody aggregation.
  • the use of antibodies as the active ingredient of pharmaceuticals is now widespread, including the products HERCEPTIN® (trastuzumab), RITUXAN® (ntuximab), SYNAGIS®
  • IL-17 antagonist e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof)
  • the IL-17 antagonist will be in the form of a pyrogen-free, parenterally acceptable solution.
  • compositions for intravenous, cutaneous, or subcutaneous injection may contain, in addition to the IL-17 antagonist, an isotonic vehicle such as sodium chloride, Ringer's, dextrose, dextrose and sodium chloride, lactated Ringer's, or other vehicle as known in the art.
  • an isotonic vehicle such as sodium chloride, Ringer's, dextrose, dextrose and sodium chloride, lactated Ringer's, or other vehicle as known in the art.
  • the appropriate dosage will, of course, vary depending upon, for example, the particular IL-17 antagonists, e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof) to be employed, the host, the mode of administration and the nature and severity of the condition being treated, and on the nature of prior treatments that the patient has undergone.
  • the attending health care provider will decide the amount of the IL-17 antagonist with which to treat each individual patient.
  • the attending health care provider may administer low doses of the IL-17 antagonist and observe the patient's response.
  • the initial dose(s) of IL-17 antagonist administered to a patient are high, and then are titrated downward until signs of relapse occur. Larger doses of the IL-17 antagonist may be administered until the optimal therapeutic effect is obtained for the patient, and the dosage is not generally increased further.
  • a therapeutically effective amount of an IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) is administered to a patient, e.g., a mammal (e.g., a human).
  • a mammal e.g., a human
  • the disclosed methods provide for selective treatment of patients (i.e., patients having a AI selected from uveitis, RA, PsA, AS, MS and psoriasis) depending on the presence of an AI response marker, this does not preclude that, if the patient is ultimately treated with an IL-17 antagonist, such IL-17 antagonist therapy is necessarily a monotherapy.
  • the IL-17 antagonist e.g., secukinumab
  • the IL-17 antagonist may be administered in accordance with the method of the disclosure either alone or in combination with other therapeutics for treating AI disease in patients, e.g., in combination with an immunosuppressive agent, a disease-modifying anti-rheumatic drug (DMARD) (e.g., MTX), a pain-control drug (e.g., tramadol or paracetamol), a steroid (e.g., prednisone), a non-steroidal anti-inflammatory drug (NSAID), a cytokine antagonist, a bone anabolic, a bone anti-resorptive, and combinations thereof (e.g., dual and tripple therapies).
  • DMARD disease-modifying anti-rheumatic drug
  • a pain-control drug e.g., tramadol or paracetamol
  • a steroid e.g., prednisone
  • NSAID non-ster
  • an IL-17 antagonist When coadministered with one or more additional therapeutics, an IL-17 antagonist may be administered either simultaneously with the other therapeutic, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering the IL-17 antagonist in combination with other therapeutics, as well as the appropriate dosages for co-delivery.
  • IL-17 antagonist e.g., secukinumab
  • an IL-17 antagonist include, propionic acid derivative, acetic acid derivative, enolic acid derivatives, fenamic acid derivatives, Cox inhibitors, e.g., lumiracoxib, ibuprophen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin, indomethacin, sulindac, etodolac, ketorolac, nabumetone, aspirin, naproxen, valdecoxib, etoricoxib, MK0966; rofecoxib, acetominophen, Celecoxib, Diclofenac, tramadol, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefanamic acid, meclofenamic acid, flufenamic acid, tolfenamic,
  • DMARDs useful in combination with an IL-17 antagonist include, methotrexate (MTX), antimalarial drugs (e.g., hydroxychloroquine and chloroquine),
  • methotrexate MTX
  • antimalarial drugs e.g., hydroxychloroquine and chloroquine
  • Steroids useful in combination with an IL-17 antagonist, e.g., secukinumab, include Prednisolone, Prednisone, dexamethasone, Cortisol, cortisone, hydrocortisone,
  • methylprednisolone betamethasone, triamcinolone, beclometasome, fludrocortisone,
  • deoxycorticosterone aldosterone.
  • Other therapeutics useful in combination with an IL-17 antagonist include, ADALIMUMAB (Humira®), ETANERCEPT (Enbrel®), INFLIXIMAB (Remicade®; TA-650), CERTOLIZUMAB PEGOL (Cimzia®;
  • CDP870 GOLIMUMAB (Simpom®; CNT0148), ANAKINAS (Kineret®), RITUXIMAB (Rituxan®; MabThera®), ABATACEPT (Orencia®), TOCILIZUMAB (RoActemAS
  • IL-17 antagonists LY2439821, RG4934, AMG827, SCH900117, R05310074, MEDI-571, CAT-2200
  • IL-23 antagonists IL-20 antagonists
  • IL-6 antagonists TNF alpha antagonists (e.g., TNF alpha antagonists or TNF alpha receptor antagonsits, e.g., pegsunercept, etc.)
  • BLyS antagonists e.g., Atacicept, Benlysta®/ LymphoStat-B® (belimumab)
  • p38 Inhibitors CD20 antagonists (Ocrelizumab, Ofatumumab (Arzerra®)), Interferon gamma antagonists (Fontolizuma
  • Tysabri® selective adhesion molecule inhibitor
  • mitoxantrone mitoxantrone: Novantrone®
  • fingolimod (Gilenya®/FTY720) (sphingosine- 1 phosphate [SIP] receptor modulator); and teriflunomide (Aubagio®/ HMR1726).
  • An IL-17 antagonist e.g., secukinumab
  • the duration of intravenous (i.v.) therapy using a pharmaceutical composition of the present disclosure will vary, depending on the severity of the disease being treated and the condition and personal response of each individual patient. Also contemplated is subcutaneous (s.c.) therapy using a pharmaceutical composition of the present disclosure. The health care provider will decide on the appropriate duration of i.v. or s.c. therapy and the timing of administration of the therapy, using the pharmaceutical composition of the present disclosure.
  • Preferred dosing and treatment regimens for treating AI disease patients (i.e., patients having uveitis, RA, PsA, AS, MS or psoriasis) who have an AI response marker are provided in PCT Application No. PCT/US2011/064307, PCT/EP2011/069476, and PCT/EP2011/067522, which are incoporated by reference herein in their entirety.
  • Doses may be delivered based on weight, e.g., 3 mg/kg, 10 mg/kg, 15 mg/kg, or as a fixed amount, e.g., 75 mg, 150 mg, 300 mg, depending on the disease.
  • RA, PsA, AS patients may use about 75 mg, about 150 mg or 300 mg of secukinumab s.c. for maintenance dosing, while psoriasis patients typically use about 150 mg or about 300 mg secukinumab s.c. for maintenance dosing.
  • dose escalation may be required (e.g., during the induction and/or maintenance phase) for certain patients, e.g., patients that display inadequate response to treatment with the IL-17 antagonists, e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof).
  • IL-17 antagonists e.g., IL-17 binding molecules (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecules (e.g., IL-17 receptor antibody or antigen-binding portion thereof).
  • dosages of secukinumab may be greater than about 75 mg to about 300 mg s.c, e.g., about 80 mg, about 100 mg, about 125 mg, about 175 mg, about 200 mg, about 250 mg, about 350 mg, about 400 mg, etc.; similarly, i.v. dosages may be greater than about 10 mg/kg, e.g., about 11 mg/kg, 12 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 35 mg/kg, etc.
  • dosages of secukinumab may be less than about 75 mg to about 300 mg s.c, e.g., about 25 mg, about 50 mg, about 80 mg, about 100 mg, about 125 mg, about 175 mg, about 200 mg, 250 mg, etc.; similarly, i.v.
  • dosages may be less than about 10 mg/kg, e.g., about 9 mg/kg, 8 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg etc. Delivery of the doses may be weekly, every two weeks, every three weeks, monthly (every 4 weeks), every other month (every 8 weeks), or quarterly (every 3 months).
  • the IL-17 antagonist e.g., secukinumab
  • Preferred regimens are as follows:
  • Tab e 3 Preferred dosing regimens.
  • autoimmune disease comprising either: a) selectively administering a therapeutically effective amount of an IL-17 antagonist to the patient on the basis of the patient having at least one AI response marker; or b) selectively administering a therapeutically effective amount of an AI agent to the patient on the basis of the patient not having the at least one AI response marker, wherein the AI disease is selected from the group consisting of uveitis, RA, PsA, AS, psoriasis, and MS, and further wherein: i) if the AI disease is RA, then the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl 0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs 72773968 response allele, an rsl0484879 response allele, an rs
  • Disclosed herein are methods of selectively treating a patient having an AI disease with an IL-17 antagonist comprising: a) assaying a biological sample from the patient for at least one AI response marker selected; and b) thereafter, selectively administering to the patient either: i) a therapeutically effective amount of an IL-17 antagonist on the basis of the biological sample from the patient having the at least one AI response marker; or ii) a therapeutically effective amount of an AI agent on the basis of the biological sample from the patient not having the at least one AI response marker, wherein the AI disease is selected from the group consisting of uveitis, RA, PsA, AS, psoriasis, and MS, and further wherein: i.
  • the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs72773968 response allele, an rsl0484879 response allele, an rsl 937154 response allele, an rs2241046 response allele, an rsl 1465770 response allele, and combinations thereof; and the AI agent is an RA agent selected from the group consisting of DMARDs and TNF alpha antagonists; ii.
  • the at least one AI response marker is at least one PsA response marker selected from the group consisting of an rs28096 response allele, two rs3819024 response alleles, and a combination thereof; and the AI agent is a PsA agent selected from the group consisting of NSAIDs, DMARDS, CTLA-4 antagonists, alefacept, and TNF alpha antagonists; iii.
  • the at least one AI response marker is at least one AS response marker selected from the group consisting of an rsl0484879 response allele, an rsl937154 response allele, two rs3819024 response alleles, an rsl 1465770 response allele, and combinations thereof; and the AI agent is an AS agent selected from the group consisting of NSAIDs, DMARDS, and TNF alpha antagonists; iv.
  • the at least one AI response marker is an rs2241046 response allele; and the AI agent is a psoriasis agent selected from the group consisting of DMARDs, CTLA-4 antagonists, IL-12/-23 antagonists, and TNF-alpha antagonists; v.
  • the at least one AI response marker is at least one MS response marker selected from the group consisting of an rs 72773968 response allele, two rsl 7066096 response alleles, two rs2546890 response alleles, an rsl 800693 response allele, and two rs2863212 response alleles; and the AI agent is an MS agent selected from the group consisting of IFN-b la and lb, glatiramer acetate, natalizumab, mitoxantrone, fingolimod, and teriflunomide; and vi.
  • the at least one AI response marker is at least one uveitis response marker selected from the group consisting of an rs72773968 response allele, an rs2241046 response allele, an rsl 1465770 response allele, an rsl 7482078 response allele, an rs800292 response allele, and combinations thereof; and the AI agent is a uveitis agent selected from the group consisting of a corticosteroid, NSAIDs, DMARDS, and TNF alpha antagonists.
  • IL-17 antagonists for use in treating a patient having an AI disease, characterized in that a therapeutically effective amount of the IL-17 antagonist is to be administered to the patient on the basis of said patient having at least one AI response marker, wherein the AI disease is selected from the group consisting of uveitis, RA, PsA, AS, psoriasis, and MS, and further wherein: i) if the AI disease is RA, then the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl 0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs 72773968 response allele, an rsl 0484879 response allele, an rsl 937154 response allele, an rs2241046 response allele, an rsl 1465770 response allele, and combinations thereof; ii)
  • IL-17 antagonists for use in treating a patient having an AI disease, characterized in that: a) the patient is to be selected for treatment with the IL-17 antagonist on the basis of a the patient having at least one AI response marker; and b) thereafter a
  • the AI disease is selected from the group consisting of uveitis, RA, PsA, AS, psoriasis, and MS, and further wherein: i) if the AI disease is RA, then the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl 0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs 72773968 response allele, an rsl 0484879 response allele, an rsl 937154 response allele, an rs2241046 response allele, an rsl 1465770 response allele, and combinations thereof; ii) if the AI disease is PsA, then the at least one AI response marker is at least one PsA response marker selected from the group consisting of an rs28096 response allele,
  • IL-17 antagonists for use in treating a patient having an AI disease, characterized in that: a) a biological sample from the patient is to be assayed for at least one AI response marker; and b) a therapeutically effective amount of the IL-17 antagonist is to be selectively administered to the patient on the basis of the biological sample from the patient having the at least one AI response marker, wherein the AI disease is selected from the group consisting of uveitis, RA, PsA, AS, psoriasis, and MS, and further wherein: i) if the AI disease is RA, then the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs 72773968 response allele, an rsl0484879 response allele, an rsl9371
  • IL-17 antagonists for use in treating a patient having an AI disease, characterized in that: a) a biological sample from the patient is assayed for at least one AI response marker; b) the patient is selected for treatment with the IL-17 antagonist on the basis of the biological sample from the patient having the at least one AI response marker; and c) a therapeutically effective amount of the IL-17 antagonist is to be selectively administered to the patient, wherein the AI disease is selected from the group consisting of uveitis, RA, PsA, AS, psoriasis, and MS, and further wherein: i) if the AI disease is RA, then the at least one AI response marker is at least one RA response marker selected from the group consisting of an rsl 0484879 response allele, an rs4711998 response allele, two rs27689 response alleles, an rs3214019 response allele, an rs 72773968 response allele, an rsl93
  • the method further comprises the step of obtaining the biological sample from the patient, wherein the step of obtaining is performed prior to the step of assaying.
  • the at least one AI response marker is detected by assaying the biological sample for a nucleic acid product of the at least one AI response marker, a polypeptide product of the at least one AI response marker, or an equivalent genetic marker of the at least one AI response marker.
  • the at least one AI response marker is detected by assaying the biological sample for a genomic sequence of the at least one AI response marker.
  • the biological sample is selected from the group consisting of synovial fluid, blood, serum, feces, plasma, urine, tear, saliva, cerebrospinal fluid, a leukocyte sample and a tissue sample.
  • the at least one AI response marker is detected by a technique selected from the group consisting of Northern blot analysis, polymerase chain reaction (PCR), reverse transcription-polymerase chain reaction (RT-PCR), TaqMan-based assays, direct sequencing, dynamic allele-specific hybridization, high-density oligonucleotide SNP arrays, restriction fragment length polymorphism (RFLP) assays, primer extension assays, oligonucleotide ligase assays, analysis of single strand conformation polymorphism, temperature gradient gel electrophoresis (TGGE), denaturing high performance liquid chromatography, high-resolution melting analysis, DNA mismatch-binding protein assays, SNPLex®, capillary electrophoresis, Southern Blot, immunoassays, immunohistochemistry, ELISA, flow cytometry, Western blot, HPLC, and mass spectrometry.
  • PCR polymerase chain reaction
  • RT-PCR reverse transcription-polymerase chain reaction
  • a patient having an AI disease selected from uveitis, RA, PsA, AS, psoriasis, and MS comprising assaying a biological sample from the patient for the level of ERAPl expression (e.g., mRNA, cDNA, etc.), the level of ERAPl protein, and/or the level of ERAPl activity; and thereafter selectively administering a therapeutically effective amount of an IL-17 antagonist, e.g., secukinumab, to the patient if the patient has a decreased ERAPl expression, decreased level of ERAPl protein, and/or a decreased level of ERAPl activity relative to a control.
  • an AI disease selected from uveitis, RA, PsA, AS, psoriasis, and MS
  • the level of ERAPl expression, the level of ERAPl protein, or the level of ERAPl activity is measured by assaying the biological sample from the patient for an ERAPl polymorphism that results in decreased ERAPl expression, decreased level of ERAPl protein, and/or decreased level of ERAPl activity relative to the control.
  • the AI disease is RA. In some embodiments of the disclosed methods and uses, the AI disease is PsA. In some embodiments of the disclosed methods and uses, the AI disease is AS. In some embodiments of the disclosed methods and uses, the AI disease is psoriasis. In some embodiments of the disclosed methods and uses, the AI disease is MS. In some embodiments, the patient is an MS patient having relapsing remitting MS (RRMS), primary-progressive MS, secondary-progressive MS, or progressive-relapsing MS.
  • RRMS relapsing remitting MS
  • the patient is an RA patient that is a non- responder to TNF alpha antagonism and/or a DMARD (e.g, MTX), or the patient has an inadequate response to TNF alpha antagonism and/or a DMARD (e.g., MTX).
  • the AI disease is uveitis.
  • the patient is a uveitis patient having non-infectious uveitis.
  • the uveitis patient has active uveitis. In some embodiments, the uveitis patient has anterior, intermediate, posterior, or pan uveitis.
  • the IL-17 antagonist is an IL-17 binding molecule or an IL-17 receptor binding molecule. In some embodiments, the IL-17 binding molecule or an IL-17 receptor binding molecule is an IL-17 binding molecule. In some embodiments, the IL-17 binding molecule is an IL-17 antibody or antigen-binding portion. In some embodiments of the disclosed methods and uses, the IL-17 antibody is a human antibody. In some embodiments of the disclosed methods and uses, the human IL-17 antibody is secukinumab.
  • kits for detecting an AI response marker orERAPl polymorphism, or the level ofERAPl expression, level of ERAPl protein or level of ERAPl activity in a biological sample (a test sample) from a patient can be used to predict if a patient having an AI disease selected from uveitis, RA, PsA, AS, MS and psoriasis is likely to respond (or have a higher response) to treatment with an IL-17 antagonist, e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof).
  • an IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL
  • the kit can comprise a probe (e.g., an oligonucleotode, antibody, labeled compound or other agent) capable of detecting an AI response marker orERAPl polymorphism, or the level of ERAPl expression, level of ERAPl protein or level of ERAPl activity, products of those alleles and/or an equivalent genetic marker of those alleles in a biological sample.
  • a probe e.g., an oligonucleotode, antibody, labeled compound or other agent
  • the kit may also comprise instructions for providing a prediction of the likelihood that the patient will respond to treatment with the IL-17 antagonist.
  • Probes may specifically hybridize to genomic sequences, nucleic acid products, or polypeptide products.
  • Exemplary probes are oligonucleotides or conjugated oligonucleotides that specifically hybridizes to the response alleles of Table 1 (e.g., from DNA, cDNA, mRNA, etc.); an antibody that is capable of differentiating between polypeptide products encoded by the disclosed alleles (e.g., an antibody that is capable of binding the CFH or ERAPl protein);
  • the kit can contain a probe that targets an internal control allele, which can be any allele presented in the general population. Detection of an internal control allele is designed to assure the performance of the kit.
  • the disclosed kits can also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent.
  • the kit can also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate).
  • the kit can also contain a control sample or a series of control samples that can be assayed and compared to the test sample contained. Each component of the kit is usually enclosed within an individual container, and all of the various containers are within a single package along with instructions for use.
  • kits may also comprise an IL-17 antagonist, e.g., IL-17 binding molecule (e.g., IL- 17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) (e.g., in liquid or lyophilized form) or a pharmaceutical composition comprising the IL-17 antagonist (described supra).
  • an IL-17 antagonist e.g., IL-17 binding molecule (e.g., IL- 17 antibody or antigen-binding portion thereof, e.g., secukinumab) or IL-17 receptor binding molecule (e.g., IL-17 antibody or antigen-binding portion thereof) (e.g., in liquid or lyophilized form) or a pharmaceutical composition comprising the IL-17 antagonist (described supra).
  • an IL-17 antagonist e.g., secukinumab
  • kits may comprise means for administering the IL-17 antagonist (e.g., a syringe and vial, a prefilled syringe, a prefilled pen) and instructions for use.
  • kits may contain additional therapeutic agents (described supra) for treating an AI disease, e.g., for delivery in combination with the enclosed IL-17 antagonist, e.g., secukinumab.
  • phrases "means for administering” is used to indicate any available implement for systemically administering a drug top a patient, including, but not limited to, a pre-filled syringe, a vial and syringe, an injection pen, an autoinjector, an i.v. drip and bag, a pump, etc.
  • a patient may self-administer the drug (i.e., administer the drug on their own behalf) or a physician may administer the drug.
  • an artisan may analyze more than one marker.
  • a clinician may choose to analyze one or many of the SNPs in Table 1 in a single patient.
  • the haplotype of the patient may be analysed for 1, 2, 3 or more of the alleles in Table 1 , e.g., all (or some) of the ERAPl alleles in Table 1.
  • a biological sample from an RA patient may be analyzed for the TGA haplotype at ERAPl SNPs rs27689/rs3214019/rs72773968 or a sample from a uveitis patient may be analyzed for the AT hapolotype aX ERAPl SNPs rs72773968/rsl7482078.
  • biomarkers may be analyzed, e.g., additional genetic markers (e.g., HLA alleles), transcription markers (e.g., mRNA/miRNA derived form blood, PBMCs, biopsies, etc.), and protein and cellular markers (e.g., protein biomarkers in serum or feces and Thl7 and Treg cells.
  • additional biomarkers for use in the disclosed methods with the AI respose markers disclosed herein are found in WO2012059598, WO2012/082573, U.S. Provisional Application No.
  • a preferred ERAP1 SNP additional marker is an rs30187 "T" allele (US Provisional Patent Application 61/636,062).
  • the IL-17 binding molecule is selected from the group consisting of: a) an IL-17 antibody that binds to an epitope of IL-17 comprising Leu74, Tyr85, His86, Met87, Asn88, Vall24, Thrl25, Prol26, Ilel27, Vall28, Hisl29; b) an IL-17 antibody that binds to an epitope of IL-17 comprising Tyr43, Tyr44, Arg46, Ala79, Asp80; c) an IL-17 antibody that binds to an epitope of an IL-17 homodimer having two mature IL-17 protein chains, said epitope comprising Leu74, Tyr85, His86, Met87, Asn88, Vall24, Thrl25, Prol26, Ilel27, Vall28, Hisl29 on one chain and Tyr43, Tyr44, Arg46, Ala79, Asp80 on the other chain; d) an IL-17 antibody that
  • immunoglobulin V H domain comprising the amino acid sequence set forth as SEQ ID NO: 8 and an immunoglobulin V L domain comprising the amino acid sequence set forth as SEQ ID NO: 10; iv) an immunoglobulin V H domain comprising the hypervariable regions set forth as SEQ ID NO: l, SEQ ID NO: 2, and SEQ ID NO: 3; v) an immunoglobulin V L domain comprising the hypervariable regions set forth as SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; vi) an immunoglobulin V H domain comprising the hypervariable regions set forth PsA SEQ ID NO: 11, SEQ ID NO: 12 and SEQ ID NO: 13; vii) an immunoglobulin V H domain comprising the hypervariable regions set forth as SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 and an immunoglobulin V L domain comprising the hypervariable regions set forth as SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6; and
  • a 52 week extension study CAIN457A2206E1 to this core study in 28 patients dosed at 3 mg/kg i.v. every 4 weeks showed no safety signal. Confirmatory phase III trials are currently ongoing.
  • Example 2 -Secukinumab for Ankylosing Spondylitis CAIN457A2209 was a 28-week placebo controlled double-blind proof-of-concept study to determine the efficacy of secukinumab (2 x 10 mg/kg i.v. three weeks apart) in 30 patients with moderate to severe ankylosing spondylitis (AS) refractory to NSAID therapy with or without previous use of TNF-a blocking therapy.
  • An additional 30 patients were recruited into Part 2 of the study and were randomized 2:2: 1 into 0.1 mg/kg, 1 mg/kg and 10 mg/kg.
  • the primary endpoint was met in Part 1 , as secukinumab induced statistically significant AS AS 20% improvements compared with placebo at week 6.
  • the Bath AS disease activity index (BASDAI) change from baseline was similar between the secukinumab 10 mg/kg and 1.0 mg/kg dose arms. Responses were induced rapidly and seen as early as week 1 after 2 x 10 mg/kg secukinumab. Reductions of spinal inflammation as measured by MRI scoring were also noted in subsets of patients. Confirmatory phase III trials are currently ongoing.
  • BASDAI Bath AS disease activity index
  • ACR20 response rates at week 12 were 48% (10 mg/kg i.v. x3 q 2 weeks followed by 150 mg s.c. q 4 weeks), 55% (150 mg s.c. q week followed by 150 mg s.c. q 4 weeks) and 88% (10 mg/kg i.v. q 2 weeks x 6) respectively on secukinumab.
  • CAIN457F2208 was a randomized, double-blind, placebo-controlled trial, patients with RA (2010 criteria), TJC > 6, SJC > 6, CRP > 10 mg/L, who were biologic-naive and DMARD incomplete responders or DMARD naive, were randomized 2: 1 to secukinumab 6x10 mg/kg i.v. or placebo every other week (wk). Associations of treatment effect of secukinumab vs.
  • HLA-DRB1*04 primary endpoint
  • HLA-DRB1 *SE protein markers RF and anti-CCP were assessed at wk 12 in 96 evaluable patients, using change from baseline in DAS28-CRP by ANCOVA, or ACR20 response by logistic regression.
  • HLA-DRB1*04 allelic group and HLA-DRB 1*0401 did not demonstrate significant associations with response to secukinumab vs. placebo.
  • the small number of HLA-DRB 1*SE non-carriers within the CCP/RF positive sub-cohort precluded assessment of associations in this group.
  • CAIN457B2201 was a Phase II randomized, multi-center, double-blind, placebo- controlled proof-of- concept study aimed at determining if secukinumab can reduce the disease activity in Relapsing-Remitting Multiple Sclerosis (RRMS), as measured by the occurrence of new active brain magnetic resonance imaging (MRI) lesions during a 24 week treatment period.
  • RRMS Relapsing-Remitting Multiple Sclerosis
  • a phase III clinical development program for secukinumab in uveitis patients with quiescen non-infectious posterior segment uveitis (CAIN457C2301), active non-infectious posterior segment uveitis (CAIN457C2302), and Behcet's uveitis (CAIN457C2303) was commenced in 2009 using dose regimens of 150 mg s.c. every 4 weeks (q4wk), 300 mg s.c. q4wk and 300 mg s.c every 2 weeks (q2wk).
  • the available results did not demonstrate any significant difference between secukinumab and placebo for the primary efficacy endpoint.
  • the proportion of responders in the i.v. -treated patients was approximately double that of the s.c.-treated patients (62% in the 10 mg/kg i.v. q2wk group, 73% in the 30 mg/kg iv q4wk group, 33% in the 300 mg s.c. q2wk group).
  • the 30 mg/kg i.v. q4wk group met superiority criteria compared to the 300 mg s.c. q2wk dose. Therefore, further clinical studies evaluating the effects of secukinumab in non-infectious posterior uveitis are planned.
  • a PG analysis was run using consented PG samples from secukinumab RA, PsA, AS (also referred to as “ASp”), psoriasis (also referred to as "Pso”), and MS trials with the obejctives to find new secukinumab response predictive genetic biomarkers in single indications and to find genetic biomarkers associating with secukinumab response across indications.
  • Table 4 shows sample size, dosing, efficacy endpoints, response rate, and adjusted covartiates.
  • the subjects included in the PG analysis need to satisfy the following four criteria: (1) Consented Caucasian patients treated by secukinumab; (2) Call rate >80%; (3) No gender discrepancy. Thirty extreme Responder Non-responders were selected from the psoriasis trial to have comparable sample size with AS, PsA and MS.
  • ⁇ IL17A secukinumab target; plays major role in inflammatory and autoimmune diseases;
  • ⁇ IL17RA/C receptor of IL17A
  • ⁇ IL23R relevant pathway, associated with multiple autoimmune diseases
  • TRAF3IP2 regulates IL-17 pathway; association with secukinumab response observed in PsA (International Patent Application PCT/US2012/041310, which is incorporated by reference herein in its entirety); risk factor for Pso and PsA;
  • ⁇ IL12B strong association with secukinumab response observed in MS (unpublished data); risk factor for multiple autoimmune diseases; plays a major role in IL23R signalling and Thl7 cell differentiation;
  • ⁇ ERAP1 certain alleles show strong association with secukinumab response observed in AS (US Provisional Patent Application 61/636062, which is incorporated by reference herein in its entirety); associated with AS (Evans et al. (2011) Nat Genet 43(8) 761-67, psoriasis (Strange et al. (2010) Nat Genet 42:982-990), and MS (Guenni et al. (2012) PLoS One 7(l)e29931), involved in trimming peptides for MHC presentation and cytokine receptor cleavage; risk factor for multiple autoimmune disease;
  • ⁇ TNFSF15 strong association with secukinumab response observed in Crohn's Disease and association also observed in PsA (International Patent Application PCT/US2012/041310, which is incorporated by reference herein in its entirety); plays with a major role in IL23R signalling and Thl7 cell differentiation.
  • Tests for asscoiation with secukinumab response were conducted in secukinumab-treated patients were used for the genetic analysis.
  • the null hypothesis was that the coefficient for the genotype variable was equal to zero, and the corresponding p- value was presented. Rejecting the null hypothesis would mean concluding that genotype was a predictor of response to secukinumab as measured by the specific clinical endpoint Variants showing association with secukinumab response was also tested in placebo arm to see whether the association is specific to secukinumab.
  • Example 7.2 Samples and Processing of Pharmacogenetic (PG) study in AIN457F2208 (RA)
  • HLA-DRB1 *04 allelic group HLA-DRB1*SE allelic group and HLA-DRB1 position 11 V/L allelic group were assessed as primary and secondary objectives of the trial.
  • exploratory PG analysis was performed in 53 sero-positive Secukinumab-treated patients who are Caucasians based on self-classification with the goal to identify genetic variants that may associate with the change in DAS28-CRP and ACR50 response to Secukinumab to at week 10.
  • the following SNPs as selected from the WGS microarray data were tested as candidate genetic markers.
  • RA (F2201): IL17A/rs 10484879; IL17A/rs4711998
  • PsA (A2206): ERAPl/rs28096
  • RA (F2201 & A2101): ERAPl/rs27689; ERAPl/rs3214019
  • IL4R/rsl 805010 reported to associate with Increased Thl7 Cell Frequency and Poor Clinical Outcome in Rheumatoid Arthritis
  • TRAF3IP2/rs33980500 reported to abolish IL-17 signaling but by doing so somehow increases Thl7 and may drive Psoriasis through a non-Il-17A pathway. They claim that these patients should not respond to anti-Il- 17 treatment but rather p40 or pi 9.
  • ⁇ CCR6/rs3093023 reported to be associated with increased RA risk (in multiple populations) and elevated serum IL17 levels in Japanese
  • Exploratory PG analysis was performed in 146 Caucasian RA patients in AIN457 arm in the AIN457F2206 study with the goal to find genetic variants that may associate with response to secukinumab. All are seropositive as defined by inclusion criteria of the study.
  • HLA-DRBl allelic groups as well as the SNPs as selected from the WGS microarray data were tested as candidate genetic markers.
  • HLA-DRB 1 SE allelic group major risk alleles (*0401, *0404, *0405, *0408, *1001, each allele separately, as well as pooled together as an allelic group)
  • CXCL10/rs4859588 full LD with rs3921: change of CXCL10 associates with AIN response in F2208; This SNP was reported to associate with the development of invasive aspergillosis after allogeneic hematopoietic stem-cell transplantation (HSCT) and the disease progression of multiple sclerosis in European populations.
  • HSCT allogeneic hematopoietic stem-cell transplantation
  • Exploratory PG analysis was performed in Uveitis patients in AIN457 arm in the ⁇ 457 ⁇ 2208 study with the goal to find genetic variants that may associate with response to secukinumab.
  • PG samples from Cohorts 2,3,5,6 were combined for the PG analysis, with all the 10 mg/kg groups being combined as one dosing arm, and all the 30 mg/kg groups being combined as another dosing arm. The 1 mg/kg and 3 mg/kg groups were excluded since response rate is known to be very low in these arms.
  • o risk factor for Bechet's disease ERAPl / rsl 7482078 (R725Q)
  • o risk factor for anterior Uveitis CFH-rs800292
  • polymorphisms in IL17A were assessed by Sanger sequencing.
  • the complete coding region of the gene, including all three exons, as well as the majority of the 5' and 3' UTR regions were sequenced.
  • PCR was performed for the regions described using unique primers carrying a universal Ml 3 sequence tag.
  • PCR amplification was optimized using AmpliTaq Gold Master Mix (Applied Biosystems, 43270590).
  • PCR products were cleaned up with AMPure Xp magnetic beads (Beckman, A63882) on a Biomek FX. After cleanup, products were sequenced in both forward and reverse directions by Sanger sequencing with M13 universal primers and BigDye Terminator v3.1 (Life Technologies, 4337456).
  • Sequencing products were cleaned up with CleanSEQ (Beckman, A29154) and then run on an Applied Biosystems 3730x1 with POP-7 for sequence detection.
  • traces were aligned to reference sequences for each amplicon from NCBI using PhredPhrap (University of Washington). The traces were visually inspected for any discrepancies from canonical sequence using Consed (University of Washington, version 19.0). All sequence variations were noted and any corresponding protein coding changes characterized where possible.
  • the final allele calls for each sample at each identified polymorphism position were captured with Polyphred (University of Washington, Version 6.18), manually QCed, and uploaded to the CGL Genotype Database.
  • the common Hapmap SNPs with minor allele frequency (MAF)>0.01 (exons and 5kb beyond 5' and 3') as well as missense, nonsense, and frame-shift SNPs (as reported in public database HG19) of the candidate genes were selected from IL17RA/RC, IL23R, TRAF3IP2, IL12B, ERAP1, TNFSF15. SNPs that were reported in GWAS studies and were linked to these candidate genes were included as well. The genotyping of selected variants was done using Fluidigm SNP genotyping platform.
  • Fluidigm SNPtype Assays was based on allele-specific PCR SNP detection chemistry, which employs tagged, allele- specific PCR primers and a common reverse primer.
  • the assay was run in Fluidigm dynamic array, a nanofluidic system that has an integrated network of channels, chambers and valves that automatically combine the reaction of individual assay and sample. Combination of both SNPtype assay and dynamic array allows up to 9216 individual reactions to be run in a single experiment, providing high-throughput, low-cost SNP genotyping solution. A total of 640 variants were selected for genotyping.
  • HLA-DRB1 high resolution genotyping was conducted by IMGM.
  • Each sample was analyzed using SBT (Sequencing Based Typing), SSO (Sequence Specific Oligonucleotide), SSP (Sequence Specific Primer) or combinations of those methods, without considering rare alleles that were estimated to be less frequent than 1 : 100,000 according to NMDP (National Marrow Donor Program) rare allele library Version 3.7.0.
  • SBT Sequencing Based Typing
  • SSO Sequence Specific Oligonucleotide
  • SSP Sequence Specific Primer
  • HLA-DRB1 high resolution genotyping was determined using SSO (Sequence Specific Oligonucleotide) or combinations of SSO and SSP (Sequence Specific Primer), without considering rare alleles that were estimated to be less frequent than 1 : 100,000 according to NMDP (National Marrow Donor Program) rare allele library Version 3.9.0.
  • SSO Sequence Specific Oligonucleotide
  • SSP Sequence Specific Primer
  • AIN457F2208 and AIN457F2206 the WGS microarray data generation and quality control (QC) were conducted in this study.
  • Single nucleotide polymorphisms (SNPs) from genomic DNA (gDNA) samples were analyzed using the Illumina HumanOMNI5Exome microarray, which contains 4,511,703 loci from HumanOMNI5 BeadChip, and 210,371 additional SNPs selected from Exome chip which were not on the commercial HumanOmni5 array.
  • Sample quality control (QC) was conducted before genotyping, and data QC was conducted after genotyping.
  • SNPs were identified by Sanger sequencing in IL17A and ERAPl . PCR was performed using unique primers carrying Ml 3 universal sequence tags. Amplicons for each coding region were bi-directionally sequenced with universal primers and BigDye Terminator v3.1 (Applied Biosystems, 4337456). Sequencing products were cleaned up with CleanSEQ (Beckman, A29154) and run on a 3730x1 Genetic Analyzer (Applied Biosystems) with POP-7 for sequence detection. Traces were aligned to reference sequences for each amplicon from NCBI using PhredPhrap (University of Washington) and were then visually inspected for discrepancies from canonical sequence using Consed (University of Washington, version 19.0).
  • Genotyping was performed using TaqMan Assays-by-Design and Assays-on-Demand (Life Technologies, Carlsbad, CA). Genotyping was performed according to manufacturer's protocol PN 4332856D, using Taqman Universal Master Mix (PN 4326614), with the following exceptions: 2 ⁇ 1 of DNA sample per reaction, for a total DNA input of 20ng, and 0.25 ⁇ 1 of water added per reaction. PCR reactions were conducted on the GeneAmp 9700 (Applied Biosystems) using the program specified in the vendor protocol with the following exception: standard thermal profile is used with 50 cycles, rather than 40. Endpoint reads were conducted on the ABI PRISM 7900HT Sequence Detection System, with data analysis by SNP Manager and SDS 2.2.2 (Applied Biosystems).
  • a SNP rsl0484879 has the best p value, with nominal p-value of 0.00017 in additive mode of inheritance (Figure 1) for association with percentage percentage ACR20 at week 16 in RA Phil trial (F2201) combining all secukinumab dosing groups adjusting for dosing.
  • the presence of at least one 'C allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab, and the presence of two 'C alleles is indicative of a further increased likelihood that the patient will respond to treatment with secukinumab.
  • Such trend of association was not observed in the placebo arm.
  • SNP rs 10484879 is in an intron of IL17A.
  • a SNP rs4711998 has nominal p-value of 0.00094 in dominant mode of inheritance (Figure 2) for association with reduction of DAS28 at week 16 from baseline in RA Phil trial (F2201) combining all secukinumab dosing groups adjusting for dosing. The presence of at least one 'G' allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. Such trend of association was not observed in the placebo arm.
  • SNP rs4711998 is near IL17A.
  • a SNP rs28096 has nominal p-value of 0.00054 in dominant mode of inheritance (Figure 3) for association with percentage ACR50 at week 6 in secukinumab-treated PsA patients.
  • the presence of at least one 'T' allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. It was difficult to check for association in the placebo arm with only 3 patients having the CT/TT genotype and 1 ACR50 / ACR20 responder. SNP rs28096 is in an intron of ERAP1.
  • a SNP rs27689 has nominal p-value of 0.038 in dominant mode of inheritance for association with percentage ACR50 at week 16 in RA Phil trial (F2201) combining all secukinumab dosing groups adjusting for dosing (Figure 4). Such trend of association was also observed in RA PoC trial (A2101) with nominal p-value of 0.050 ( Figure 4). In both RA trials the presence of at least one ' ⁇ ' allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. There was no such trend of association observed in the placebo arm of F2201 trial. It was difficult to check for association in the placebo arm of A2101 trial with only 3 patients carrying ' ⁇ ' allele. SNP rs27689 is in an intron of ERAP1.
  • a SNP rs3214019 has nominal p-value of 0.045 in dominant mode of inheritance for association with percentage ACR50 at week 16 in RA Phil trial (F2201) combining all secukinumab dosing groups adjusting for dosing (Figure 5). Such trend of association was also observed in RA PoC trial (A2101) with nominal p-value of 0.009 ( Figure 5). In both RA trials the presence of at least one 'G' allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. There was no such trend of association observed in the placebo arm of the F2201 trial. It was difficult to check for association in the placebo arm of the A2101 trial with only 2 patients carrying 'G' allele. SNP rs3214019 is in an intron of ERAP1.
  • a SNP rsl 7066096 has nominal p-value of 0.00090 in dominant mode of inheritance for association with percentage responders in MS PoC trial (B2201) ( Figure 12). The presence of two "A" alleles is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. There was no such trend of association observed in the placebo arm of the B2201 trial. SNP rsl 7066096 is in intergenic between IL20RA and IL22RA2.
  • a SNP rs2546890 has nominal p-value of 0.0049 in dominant mode of inheritance for association with percentage responders in MS PoC trial (B2201) ( Figure 13). The presence of two "G" alleles is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. There was no such trend of association observed in the placebo arm of the B2201 trial. SNP rs2546890 is in intron otIL12B.
  • a SNP rsl 800693 has nominal p-value of 0.055 in dominant mode of inheritance for association with percentage responders in MS PoC trial (B2201) ( Figure 14). The presence of at least one "G" allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab. There was no such trend of association observed in the placebo arm of the B2201 trial. SNP rsl800693 is in splicing site of TNFRSF1A.
  • a SNP rs2863212 has nominal p-value of 0.0023 in dominant mode of inheritance for association with percentage responders in MS PoC trial (B2201) ( Figure 15). The presence of two "T' allelesis indicative of an increased likelihood that the patient will respond to treatment with secukinumab. There was no such trend of association observed in the placebo arm of the B2201 trial. SNP rs2863212 is in intron oiIL23R.
  • a SNP rs 17482078 has nominal p-value of 0.037 in dominant mode of inheritance for association with percentage responders in Uveitis PoC trial combing all races (A2208) (Figure
  • SNP rs 17482078 is a missense SNP in ERAP1 and a risk factor for Behcet's disease, especially in HLA-B51 carriers (Yohei Kirino et al, Nature Genetics 45:202-207, 2013).
  • a SNP rs800292 has nominal p-value of 0.040 in additive mode of inheritance for association with percentage remission in Uveitis PoC trial combing all races (A2208) (Figure
  • SNP rs 72773968 is coding nonsynonymous in ERAP1. It has nominal p-value of 0.027 in additive mode of inheritance for association with percentage ACR20 at week 16 in the RA trial F2201 combining all secukinumab dosing groups adjusting for dosing (Figure 6). Such trend of association was also observed for response rate in MS PoC trial (B2201) with nominal p-value of 0.023 ( Figure 6); as well as for response rate in Uveitis PoC trial (A2208) combining all races with nominal p-value of 0.016 ( Figure 16). A similar trend was observed in the subcohort of Caucasians (data not shown).
  • a SNP rsl0484879 is in an intron oiIL17A. It has nominal p-value of 0.00017 in additive mode of inheritance for association with percentage ACR20 at week 16, as well as nominal p- value of 0.006 in additive mode of inheritance for association with change of DAS28 from baseline at week 16 in the RA trial F2201 combining all secukinumab dosing groups adjusting for dosing (Figure 7). Such trend of association was also observed for ASAS40 at week 6 in the AS trial A2209 with nominal p-value of 0.020 ( Figure 7).
  • SNP rsl937154 is near IL17A. It has nominal p-value of 0.013 in additive mode of inheritance for association with percentage ACR20 at week 16 in the RA trial F2201 combining all secukinumab dosing groups adjusting for dosing (Figure 8). Such trend of association was also observed for ASAS5/6 at week 6 in the AS trial A2209 with nominal p-value of 0.087 ( Figure 8). In both trials the presence of at least one ' ⁇ ' allele is indicative of an increased likelihood that the patient will respond to treatment with secukinumab, and the presence of two ' ⁇ ' alleles is indicative of a further increased likelihood that the patient will respond to treatment with secukinumab. It was difficult to check for association in the placebo arm due to the small sample size carrying the ' ⁇ ' allele.
  • SNP rs3819024 is in the promoter of IL17A. It has nominal p-value of 0.074 in dominant mode of inheritance for association with percentage ACR20 at week 6 in the PsA trial A2206 ( Figure 9). Such trend of association was also observed for ASAS56 at week 6 in the AS trial A2209 with nominal p-value of 0.036, as well as for ASAS40 at week 6 with nominal p-value of 0.058 ( Figure 9). In both trials the presence of 'AA' genotype is indicative of an increase likelihood that the patient will respond to treatment with secukinumab. It was difficult to check for association in the placebo arm due to the small sample size carrying the 'G' allele.
  • SNP rs2241046 is in an intron of IL17RA. It has nominal p-value of 0.032 in additive mode of inheritance for association with change of DAS28 from baseline at week 16 in the RA trial F2201 combining secukinumab 150/300 mg dosing groups (Figure 10). Such trend of association was also observed for PASI score at week 12 with nominal p-value of 0.054, as well as PASI75 at week 12 with nominal p-value of 0.017 in the psoriais A2211 trial ( Figure 10). What's more, similar trend of association was also observed for response rate in Uveitis PoC trial (A2208) combining all races with nominal p-value of 0.006 ( Figure 17).
  • a SNP rs 11465770 has nominal p-value of 0.039 in additive mode of inheritance for association with change of DAS28 from baseline at week 16 in the RA trial F2201 combining secukinumab 150/300 mg dosing groups (Figure 11).
  • Such trend of association was also observed for ASAS40 at week 6 with nominal p-value of 0.012, as well as for ASAS20 at week 6 with nominal p-value of 0.10 in the AS trial A2209 ( Figure 11).
  • Thirdly, such trend of association was also observed for ACR50 at week 10 in US/EU Caucasian seropositive RA patientsin in the trial F2208 with with nominal p-value of 0.0074 (data not shown).
  • Example 9 Decreased ERAP1 expression, levels of ERAP1 protein and/or levels of ERAP1 activity may be useful to predict improved response to IL-17 antagonism
  • the rs30187 "C” allele encodes a K528R variant of ERAPl with impaired catalytic properties, and is associated with reduced incidence of AS. (Kochan et al. (2011) Proc Natl Acad Sci U S A. 108(19):7745-50).
  • AS patients having an rs30187 "T” allele which encodes fully active ERAPl, display decreased secukinumab response, and that AS patients who carry this "T" allele typically showed higher levels of ERAPl expression
  • US Provisional Patent Application 61/636,062 which is incorporated by reference herein in its entirety. This suggested to us that increased ERAPl activity and expression might predict decreased response to IL-17 antagonism for AS patients, and by correlation, that decreased ERAPl expression could therefore predict improved response to IL-17 antagonism for AS patients.
  • ERAPl plays a role common to AI diseases that involve dysregulation of the IL-17 pathway (e.g., uveitis, AS, PsA, RA, MS, psoriasis, etc.).
  • levels of ERAPl protein and/or levels of ERAPl activity may be useful to predict improved response to IL-17 antagonism (e.g., secukinumab) for psoriasis, uveitis, MS, PsA, and RA patients (especially uveitis, MS, PsA and RA patients), while increased levels of ERAPl expression, levels of ERAPl protein and/or levels of ERAPl activity may be useful to predict a weaker response to IL-17 antagonism (e.g., secukinumab) for psoriasis, uveitis, MS, PsA, and RA patients (especially MS, PsA and RA patients).
  • IL-17 antagonism e.g., secukinumab
  • IL-17 blockade is currently being explored as a therapeutic strategy in a variety of immune-mediated inflammatory diseases, including rheumatoid arthritis, anklyosing spondylitis, uveitis, psoriasis, psoriatic arthritis and multiple sclerosis.
  • IL-17 antagonism e.g., an IL-17 antibody, e.g., secukinumab
  • the predictive methods and personalized therapies disclosed herein are useful to maximize the benefit and minimize the risk of IL-17 antagonism in patients having various AI diseases by identifying those patients likely to respond prior to treatment with an IL-17 antagonist, such as secukinumab.
  • HLA-DRB1 alleles encoding the shared epitope (SE) confer higher risk for RA development (Gonzalez-Gay et al. (2002) Sem. Arthritis. Rheum. 31 :355-60; Fries et al. (2002) Arthritis and Rheumatism 46:2320-29; van der Helm-van Mil et al. (2006) Arthritis and Rheum. 54: 1117-21).

Abstract

L'invention concerne de nouvelles méthodes prédictives et thérapies personnalisées destinées au traitement d'une maladie auto-immune (AI) choisie parmi le psoriasis, l'uvéite, la polyarthrite rhumatoïde (PR), le rhumatisme psoriasique (RP), la spondylarthrite ankylosante (SA) et la sclérose en plaques (SEP) Plus particulièrement, l'invention concerne des méthodes de traitement d'un patient atteint d'une maladie AI par administration sélective d'un antagoniste de l'IL-17, par exemple, d'un anticorps anti-IL-17, tel que le sécukinumab, audit patient sur la base du fait que ce patient présente une prédisposition génétique à développer une réponse favorable au traitement par l'antagoniste de l'IL-17. L'invention concerne en outre des sources d'information sous forme transmissible, des méthodes diagnostiques et des trousses servant à prédire la probabilité qu'un patient atteint d'uvéite, de PR, RP, SA, psoriasis ou SEP développe une réponse au traitement par un antagoniste de l'IL-17, par exemple, un anticorps anti-IL-17, tel que le sécukinumab.
PCT/IB2014/060110 2013-03-26 2014-03-24 Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'il -17 WO2014155278A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361805257P 2013-03-26 2013-03-26
US61/805,257 2013-03-26

Publications (2)

Publication Number Publication Date
WO2014155278A2 true WO2014155278A2 (fr) 2014-10-02
WO2014155278A3 WO2014155278A3 (fr) 2014-12-18

Family

ID=50513390

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/060110 WO2014155278A2 (fr) 2013-03-26 2014-03-24 Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'il -17

Country Status (1)

Country Link
WO (1) WO2014155278A2 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017221174A1 (fr) * 2016-06-22 2017-12-28 Novartis Ag Méthodes de traitement du vitiligo à l'aide d'anticorps de l'interleukine-17 (il -17)
WO2021053591A1 (fr) * 2019-09-20 2021-03-25 Novartis Ag Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'interleukine-17 (il-17)
US11492396B2 (en) 2015-10-27 2022-11-08 UCB Biopharma SRL Methods of treatment using anti-IL-17A/F antibodies
US20230250434A1 (en) * 2021-10-14 2023-08-10 Regeneron Pharmaceuticals, Inc. Treatment Of Uveitis With Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) Inhibitors

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000032773A1 (fr) 1998-11-27 2000-06-08 Darwin Discovery Ltd. Compositions et methodes d'augmentation de la mineralisation de la substance osseuse
US20030229041A1 (en) 2002-03-01 2003-12-11 Sutherland May S. Kung Methods to increase or decrease bone density
WO2003106657A2 (fr) 2002-06-14 2003-12-24 Stowers Institute For Medical Research Sequences nucleotidiques et sequences d'acides amines wise/sost
WO2005003158A2 (fr) 2003-06-16 2005-01-13 Celltech R & D, Inc. Compositions et methodes d'augmentation de la mineralisation osseuse
WO2005014650A2 (fr) 2003-06-16 2005-02-17 Celltech R & D, Inc. Anticorps specifiques de la sclerostine et methodes permettant d'accroitre la mineralisation osseuse
WO2006013107A1 (fr) 2004-08-05 2006-02-09 Novartis Ag Anticorps antagonistes de il-17
WO2006102070A2 (fr) 2005-03-18 2006-09-28 Enzo Biochem, Inc. Sclerostine et inhibition de la signalisation par le wnt, et formation osseuse
WO2006119107A2 (fr) 2005-05-03 2006-11-09 Ucb Pharma S.A. Agents de fixation
WO2006119062A2 (fr) 2005-05-03 2006-11-09 Ucb Pharma S.A. Epitopes
WO2007117749A2 (fr) 2006-01-31 2007-10-18 Novartis Ag Anticorps antagonistes il-17
WO2008061013A2 (fr) 2006-11-10 2008-05-22 Amgen Inc. Diagnostic et thérapie basés sur des anticorps
WO2008115732A2 (fr) 2007-03-20 2008-09-25 Eli Lilly And Company Anticorps antisclérostine
WO2008133722A2 (fr) 2006-11-10 2008-11-06 Ucb Pharma S.A. Anticorps et diagnostics
WO2009039175A2 (fr) 2007-09-17 2009-03-26 Amgen Inc. Procédé pour inhiber la résorption osseuse
WO2009047356A1 (fr) 2007-10-12 2009-04-16 Novartis Ag Compositions et procédés pour l'utilisation d'anticorps contre la sclérostine
WO2009079471A1 (fr) 2007-12-14 2009-06-25 Amgen Inc. Procédé de traitement d'une fracture osseuse à l'aide d'anticorps anti-sclérostine
US20100028335A1 (en) 2007-02-02 2010-02-04 Novartis Ag Compositions and Methods to Treat Bone Related Disorders
US7838638B2 (en) 2005-12-13 2010-11-23 Eli Lilly And Company Anti-IL-17 antibodies
US8003099B2 (en) 2008-09-29 2011-08-23 Roche Palo Alto Llc Antibodies against human IL17 and uses thereof
US8057794B2 (en) 2006-06-29 2011-11-15 Ucb Pharma S.A. Antibody molecules which bind to human IL-17
US20120034656A1 (en) 2004-11-19 2012-02-09 Ucb Pharma S.A. Neutralising Antibody Molecules Having Specificity for Human IL-17
WO2012059598A2 (fr) 2010-11-05 2012-05-10 Novartis Ag Méthodes de traitement de la polyarthrite rhumatoïde au moyen d'antagonistes d'il-17
WO2012082573A1 (fr) 2010-12-13 2012-06-21 Novartis Ag Procédés prédictifs et procédés de traitement de l'arthrite en utilisant des antagonistes de l'il-17

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1983000T1 (sl) * 2003-11-21 2015-12-31 Ucb Biopharma Sprl Postopek za zdravljenje multiple skleroze z zaviranjem aktivnosti il-17
US20150125462A1 (en) * 2012-04-20 2015-05-07 Stephan Bek Methods of treating ankylosing spondylitis using il-17 antagonists

Patent Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000032773A1 (fr) 1998-11-27 2000-06-08 Darwin Discovery Ltd. Compositions et methodes d'augmentation de la mineralisation de la substance osseuse
US20080227138A1 (en) 2002-03-01 2008-09-18 Celltech R & D, Inc. Methods to Increase or Decrease Bone Density
US20030229041A1 (en) 2002-03-01 2003-12-11 Sutherland May S. Kung Methods to increase or decrease bone density
WO2003106657A2 (fr) 2002-06-14 2003-12-24 Stowers Institute For Medical Research Sequences nucleotidiques et sequences d'acides amines wise/sost
WO2005003158A2 (fr) 2003-06-16 2005-01-13 Celltech R & D, Inc. Compositions et methodes d'augmentation de la mineralisation osseuse
WO2005014650A2 (fr) 2003-06-16 2005-02-17 Celltech R & D, Inc. Anticorps specifiques de la sclerostine et methodes permettant d'accroitre la mineralisation osseuse
WO2006013107A1 (fr) 2004-08-05 2006-02-09 Novartis Ag Anticorps antagonistes de il-17
US20120034656A1 (en) 2004-11-19 2012-02-09 Ucb Pharma S.A. Neutralising Antibody Molecules Having Specificity for Human IL-17
WO2006102070A2 (fr) 2005-03-18 2006-09-28 Enzo Biochem, Inc. Sclerostine et inhibition de la signalisation par le wnt, et formation osseuse
US7872106B2 (en) 2005-05-03 2011-01-18 Amgen Inc. Sclerostin-binding antibodies
US7592429B2 (en) 2005-05-03 2009-09-22 Ucb Sa Sclerostin-binding antibody
WO2006119107A2 (fr) 2005-05-03 2006-11-09 Ucb Pharma S.A. Agents de fixation
WO2006119062A2 (fr) 2005-05-03 2006-11-09 Ucb Pharma S.A. Epitopes
US8110191B2 (en) 2005-12-13 2012-02-07 Eli Lilly And Company Anti-IL-17 antibodies
US20110027290A1 (en) 2005-12-13 2011-02-03 Eli Lilly And Company Anti-il-17 antibodies
US7838638B2 (en) 2005-12-13 2010-11-23 Eli Lilly And Company Anti-IL-17 antibodies
WO2007117749A2 (fr) 2006-01-31 2007-10-18 Novartis Ag Anticorps antagonistes il-17
US8057794B2 (en) 2006-06-29 2011-11-15 Ucb Pharma S.A. Antibody molecules which bind to human IL-17
WO2008061013A2 (fr) 2006-11-10 2008-05-22 Amgen Inc. Diagnostic et thérapie basés sur des anticorps
WO2008133722A2 (fr) 2006-11-10 2008-11-06 Ucb Pharma S.A. Anticorps et diagnostics
US20100028335A1 (en) 2007-02-02 2010-02-04 Novartis Ag Compositions and Methods to Treat Bone Related Disorders
US7744874B2 (en) 2007-03-20 2010-06-29 Eli Lilly And Company Anti-sclerostin antibodies
WO2008115732A2 (fr) 2007-03-20 2008-09-25 Eli Lilly And Company Anticorps antisclérostine
WO2009039175A2 (fr) 2007-09-17 2009-03-26 Amgen Inc. Procédé pour inhiber la résorption osseuse
WO2009047356A1 (fr) 2007-10-12 2009-04-16 Novartis Ag Compositions et procédés pour l'utilisation d'anticorps contre la sclérostine
US7879322B2 (en) 2007-10-12 2011-02-01 Novartis Ag Compositions and methods for use for antibodies against sclerostin
WO2009079471A1 (fr) 2007-12-14 2009-06-25 Amgen Inc. Procédé de traitement d'une fracture osseuse à l'aide d'anticorps anti-sclérostine
US8003099B2 (en) 2008-09-29 2011-08-23 Roche Palo Alto Llc Antibodies against human IL17 and uses thereof
WO2012059598A2 (fr) 2010-11-05 2012-05-10 Novartis Ag Méthodes de traitement de la polyarthrite rhumatoïde au moyen d'antagonistes d'il-17
WO2012082573A1 (fr) 2010-12-13 2012-06-21 Novartis Ag Procédés prédictifs et procédés de traitement de l'arthrite en utilisant des antagonistes de l'il-17

Non-Patent Citations (47)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Molecular Biology", 1989, JOHN WILEY & SONS
"GENETIC DATA ANALYSIS II", 1996, WEIR, SINEUER ASSOCIATES, INC. PUBLISHERS
"Promega Protocols and Applications Guide, Second Edition,", 1991, PROMEGA CORPORATION, pages: 263
ABRAVAYA K. ET AL., CLIN CHEM LAB MED., vol. 41, 2003, pages 468 - 474
ALTSHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALZABIN, EXPERT REV. CLIN. IMMUNOL., vol. 8, no. 2, 2012, pages 111 - 113
BARALIAKOS ET AL., ARTHRITIS RHEUM, vol. 53, 2005, pages 856 - 63
BETTENCOURT ET AL., RHEUMATOLOGY (OXFORD, vol. 52, no. 12, 2013, pages 2168 - 76
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BRAUN ET AL., RHEUMATOLOGY, vol. 44, 2005, pages 670 - 6
BRAUN J ET AL., LANCET, vol. 359, 2002, pages 1187 - 93
BUTT, W. R.,: "Practical Immunology", 1984, MARCEL DEKKER
COSTABILE ET AL., HUM. MUTAT., vol. 27, no. 12, 2006, pages 1163 - 73
DEVLIN ET AL., GENOMICS, vol. 29, no. 2, 1995, pages 311 - 22
E. MEYERS; W. MILLER, CABIOS, vol. 4, 1989, pages 11 - 17
ELLINGHAUS ET AL., NAT. GENET, vol. 42, no. 11, 2010, pages 991 - 5
EMERY; DORNER, ANN. RHEM. DIS., vol. 70, 2011, pages 2063 - 2070
EVANS ET AL., NAT GENET, vol. 43, no. 8, 2011, pages 761 - 67
EVANS ET AL., NAT. GENET., vol. 43, no. 8, 2011, pages 761 - 67
FERRANDIZ ET AL., CLINICS IN DERMATOLOGY, vol. 28, 2010, pages 81 - 87
FRIES ET AL., ARTHRITIS AND RHEUMATISM, vol. 46, 2002, pages 2320 - 29
GONZALEZ-GAY ET AL., SEM. ARTHRITIS. RHEUM., vol. 31, 2002, pages 355 - 60
GUERINI ET AL., PLOS ONE, vol. 7, no. 1, 2012, pages E29931
HARVEY ET AL., HUM. MOL. GENET., vol. 18, no. 21, 2009, pages 4204 - 4212
HUFFMEIER ET AL., NAT. GENET, vol. 42, no. 11, 2010, pages 996 - 9
HUSTON ET AL., PROC. NATL. ACAD. SCI., vol. 85, 1988, pages 5879 - 5883
HWANG ET AL., ARTHRITIS RES THER, vol. 6, 2004, pages R120 - 128
KABAT E.A. ET AL.: "Sequences of Proteins of Immunological Interest", US DEPARTMENT OF HEALTH AND HUMAN SERVICES, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTE OF HEALTH
KOCHAN ET AL., PROC NATL ACAD SCI U S A., vol. 108, no. 19, 2011, pages 7745 - 50
LIN ET AL., J RHEUMATOL., vol. 38, no. 2, 2011, pages 317 - 21
M. BROWN, RHEUMATOLOGY, vol. 47, no. 2, 2008, pages 132 - 7
MEASE PJ ET AL., LANCET, vol. 356, 2000, pages 385 - 90
MEASE PJ ET AL.: "Primer on Rheumatic Diseases", 2008, SPRINGER SCIENCE, article "Psoriatic Arthritis", pages: 170 - 192
MEYERS ET AL., COMPUT. APPL. BIOSCI., vol. 4, 1988, pages 11 - 17
MING-MING YANG ET AL., MOLECULAR VISION, vol. 18, 2012, pages 1865 - 1873
NEEDLEMAN ET AL., J. MOL. BIOL., vol. 48, 1970, pages 443 - 453
OLIVIER M., MUTAT RES., vol. 573, no. 1-2, 2005, pages 103 - 10
POTTER ET AL., ANN. RHEUM. DIS., vol. 68, 2009, pages 69 - 74
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", vol. 3, 1989, COLD SPRING HARBOR LABORATORY, article ", Chapter 18,"
SHENDURE J.; JI, H., NATURE BIOTECHNOLOGY, vol. 26, no. 10, 1998, pages 1135 - 1145
STRANGE ET AL., NAT GENET, vol. 42, 2010, pages 982 - 990
STRANGE ET AL., NAT. GENET., vol. 42, no. 11, 2010, pages 985 - 990
VAN DER HELM-VAN ET AL., ARTHRITIS AND RHEUM., vol. 54, 2006, pages 1117 - 21
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WELLCOME TRUST ET AL., NAT GENET., vol. 39, no. 11, 2007, pages 1329 - 37
YANG ET AL., MOL. VIS., vol. 18, 2012, pages 1865 - 72
YOHEI KIRINO ET AL., NATURE GENETICS, vol. 45, 2013, pages 202 - 207

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11492396B2 (en) 2015-10-27 2022-11-08 UCB Biopharma SRL Methods of treatment using anti-IL-17A/F antibodies
WO2017221174A1 (fr) * 2016-06-22 2017-12-28 Novartis Ag Méthodes de traitement du vitiligo à l'aide d'anticorps de l'interleukine-17 (il -17)
WO2021053591A1 (fr) * 2019-09-20 2021-03-25 Novartis Ag Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'interleukine-17 (il-17)
US20230250434A1 (en) * 2021-10-14 2023-08-10 Regeneron Pharmaceuticals, Inc. Treatment Of Uveitis With Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) Inhibitors

Also Published As

Publication number Publication date
WO2014155278A3 (fr) 2014-12-18

Similar Documents

Publication Publication Date Title
AU2012341081B2 (en) Methods of treating psoriatic arthritis (PsA) using IL-17 antagonists and PsA response or non- response alleles
EP3129497B1 (fr) Méthodes de traitement sélectif de l'asthme au moyen d'antagonistes de l'il-13
US11725246B2 (en) Methods of treating ophthalmic disorders
TW201307845A (zh) 預測方法及利用il-17拮抗劑治療關節炎的方法
US20150125462A1 (en) Methods of treating ankylosing spondylitis using il-17 antagonists
WO2014155278A2 (fr) Méthodes de traitement de maladies auto-immunes à l'aide d'antagonistes de l'il -17
TW201343176A (zh) 使用il-17拮抗劑治療乾癬性關節炎之方法
US20190177407A1 (en) Methods of treating dry eye disease using tnf alpha antagonists

Legal Events

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

Ref document number: 14718170

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 14718170

Country of ref document: EP

Kind code of ref document: A2