WO2020036833A1 - TRAITEMENT DE MALADIES OU DE TROUBLES DE LA PEAU PAR ADMINISTRATION D'UN ANTICORPS ANTI-OSMRβ - Google Patents

TRAITEMENT DE MALADIES OU DE TROUBLES DE LA PEAU PAR ADMINISTRATION D'UN ANTICORPS ANTI-OSMRβ Download PDF

Info

Publication number
WO2020036833A1
WO2020036833A1 PCT/US2019/046065 US2019046065W WO2020036833A1 WO 2020036833 A1 WO2020036833 A1 WO 2020036833A1 US 2019046065 W US2019046065 W US 2019046065W WO 2020036833 A1 WO2020036833 A1 WO 2020036833A1
Authority
WO
WIPO (PCT)
Prior art keywords
approximately
antibody
subject
dose
weeks
Prior art date
Application number
PCT/US2019/046065
Other languages
English (en)
Inventor
John PAOLINI
Rohan GANDHI
Zamaneh MIKHAK
Original Assignee
Kiniksa Pharmaceuticals, Ltd.
Kiniksa Pharmaceuticals Corp.
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 Kiniksa Pharmaceuticals, Ltd., Kiniksa Pharmaceuticals Corp. filed Critical Kiniksa Pharmaceuticals, Ltd.
Publication of WO2020036833A1 publication Critical patent/WO2020036833A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • Atopic dermatitis is a chronic inflammatory ' skin disorder primarily characterized by extreme itching, leading to scratching and rubbing that in turn results in the typical lesions of eczema.
  • Various diseases and disorders are accompanied by pruritus (itch).
  • ESRD end-stage renal disease
  • uremic pruritus severe pruritus
  • Prurigo nodularis (PN) also known as nodular prurigo is a skin disease characterized by itchy nodules that usually appear in the arms and legs. Patients often present with multiple excoriating lesions caused by scratching. Severe pruritus is a seriously debilitating condition.
  • the uncomfortable and often painful symptoms associated with atopic dermatitis and uremic pruritus include itching, swelling, redness, blisters, crusting, ulceration, pain, scaling, cracking, harr loss, scarring, or oozing of fluid involving the skin, eye, or mucosal membranes.
  • Other debilitating skin conditions that are accompanied by pruritus include Chronic Idiopathic Pruritus, Chronic Idiopathic Urticaria, Chronic Spontaneous Urticaria, Cutaneous Amyloidosis, Lichen Simplex Chronicus, Plaque Psoriasis, Lichens Planus, Inflammatory Ichthyosis, Mastocytosis and Bullous Pemphigoid.
  • the present invention provides, among other things, methods of treating pruritic or inflammatory' skin diseases or disorders, or pruritus associated with a disease or disorder, with an anti-OSMRp antibody.
  • the present invention provides methods for treating prurigo nodularis, atopic dermatitis, uremic pruritus, and pruritus associated with Chronic Idiopathic Pruritus, Chrome Idiopathic Urticaria, Chronic Spontaneous Urticaria, Cutaneous Amyloidosis, Lichen Simplex Chronicus, Plaque Psoriasis, Lichen Planus, inflammatory Ichthyosis, Mastocytosis or Bullous Pemphigoid, to name hut a few.
  • the present invention provides a method of treating atopic dermatitis, comprising selecting a subject who has not received prior treatment with topical corticosteroid (TCS) and administering to the subject an anti-OSMRB antibody at a
  • the present invention provides a method of treating atopic dermatitis, comprising selecting a subject who has a serum IgE level lower than 300 lU/mL, and administering to the subject an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period to improve, stabilize or reduce a symptom associated with atopic dermatitis relative to a control.
  • the present invention provides a method of treating atopic dermatitis, comprising selecting a subject based on the subject’s blood eosinophil count, and administering to the subject an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period to improve, stabilize or reduce a symptom associated with atopic dermatitis relative to a control.
  • the subject is selected if the blood eosinophil counts is lower than 0.4xl0 9 /L. In some aspects, wherein the subject is selected if the blood eosinophil count is lower than 0.4xl0 10 /L, 0. lx10 9 /L, 0 4xlQ s /L, 0.4x10 7 /L, 0.4x10 6 /L, or 0.4xl0 5 /L.
  • the subject is selected if the serum eosinophil count is lower than 5% of the total leukocyte count. In some embodiments, the subject is selected if the serum eosinophil count is lower than 15% of the total leukocyte count. In some embodiments, z the subject is selected if the serum eosinophil count is lower than 12% of the total leukocyte count. In some embodiments, the subject is selected if the serum eosinophil count is lower than 10% of the total leukocyte count. In some embodiments, the subject is selected if the serum eosinophil count is lower than 8% of the total leukocyte count.
  • the subject is selected if the serum eosinophil count is lower than 6% of the total leukocyte count. In some embodiments, the subject is selected if the serum eosinophil count is lower than 4% of the total leukocyte count. In some aspects, the subject is selected if the serum eosinophil count is lower than 2% of the total leukocyte count.
  • the present invention comprising administering to a subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval and administering topical corticosteroid to the subject upon tire occurrence of an atopic dermatitis flare, and wherein the method improves, stabilizes or reduces a symptom associated with atopic dermatiti s relative to a control
  • the anti-OSMRB antibody is administered for at least 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 week, 8 weeks or 9 weeks. In some embodiments, the anti- OSMRB antibody is administered for at least 2 weeks, 10 weeks, 12 weeks, 15 weeks, 20 weeks, 24 weeks or 30 weeks.
  • the anti-OSMRB antibody is administered at a reduced dose concurrently with topical corticosteroid.
  • the reduced dose is at or lower than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10% of the therapeutically effective dose.
  • the anti-OSMRB antibody is administered at an increased administration interval concurrently with topical corticosteroid.
  • tire increased administration interval is about 1, 1.2, 1.5, 1.8, 2, 3, 4, 6, 8, or 10 fold greater than the therapeutically effective administration interval.
  • administration of the anti-OSMRB antibody is suspended after the occurrence of flare.
  • administration of the anti-OSMRB antibody is suspended after 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 12 weeks, or 16 weeks.
  • the atopic dermatitis flare is not pruritus
  • the moderate to severe atopic dermatitis is characterized with IGA of 3 or 4 and BSA involvement of approximately 10% or more.
  • the moderate to severe atopic dieatitis is accessed by
  • EASI Eczema Area and Severity Index
  • the symptom associated with atopic dermatitis is pruritus.
  • the symptom associated with atopic dermatitis is inflammation. In some embodiments, the symptom associated with atopic dermatitis is eczema.
  • a control is indicative of the one or more disease parameters of atopic dermatitis without the treatment.
  • a control is the respective value of a disease parameter of a subject with comparable disease status, but without treatment.
  • a control is the respective value of a disease parameter of a subject with comparable disease status but treated with a placebo.
  • a control is the respective value of a disease parameter of a subject prior to treatment (also referred to as baseline).
  • a control is a reference value indicative of a disease parameter without treatment based on collective knowledge, or historical data.
  • the control is baseline severity of the symptom associated with atopic dermatitis in the subject prior to the treatment.
  • control is indicative of se verity of the symptom associated with atopic dermatitis in a control subject with the same disease status that is administered a placebo. In some embodiments, the control is indicative of severity of the symptom associated with atopic dermatitis in a control subject with the same disease status without treatment.
  • the anti-OSMRB antibody is administered subcutaneously.
  • the anti-OSMRB antibody is administered intravenously.
  • the therapeutically effective dose is an initial loading dose, and wherein the method further comprises administering at least one maintenance dose.
  • the initial loading dose is two fold greater in dosage than tire dosage of the at least one maintenance dose.
  • a therapeutically effective dose (e.g., an initial dose and/or a maintenance dose) is a flat dose.
  • a flat dose As used herein, the terms“flat dose” and“fixed dose” are used inter-changeably.
  • a suitable flat dose is between about 10 mg and 800 mg.
  • a suitable flat dose is equal to or greater than about 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75mg, 80 mg, 85mg, 90 mg, 95 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg, 140 mg, 140 mg, 150 mg, 155 mg, 160 g, 165 mg, 170 mg, 175 mg, 180 mg, 185 g, 190 mg, 195 mg, 200 mg,
  • a suitable flat dose ranges from 50- 800 mg, 50-700 mg, 50-600 mg, 50-500 mg, 100-800 mg, 100-700 mg, 100-600 mg, 100-500 mg, 100-500 mg, 100-400 mg, 150-400 mg, 200-400 mg, 250-400 mg, 300-350 mg, 320-400 mg, or 350-400 mg.
  • a suitable initial bolus or loading flat dose is about 720 mg.
  • a suitable maintenance flat dose is about 360 mg.
  • the flat dose is about 720 mg initial bolus or loading dose, and is about 360 mg maintenance dose.
  • the administration interval is daily. In some embodiments, the administration interval is ever other day, multiple times a week, once every’ week, once every two weeks, once every three weeks, once every’ four weeks, or once every five weeks. In some embodiments, the administration interval is once every eight weeks, once every ' ten weeks, or once every 12 weeks.
  • the anti-OSMRB antibody comprises a light chain complementary-determining region 1 (LCDR1 ) defined by SEQ ID NO: 8, a light chain complementary-determining region 2 (LCDR2) defined by SEQ ID NO: 9, and a light chain complementary-determining region 3 (LCDR3) defined by SEQ ID NO: 10, and a heavy chain complementary-determining region 1 (HCDR1) defined by SEQ ID NO: 5, a heavy chain complementary-determining region 2 (HCDR2) defined by SEQ ID NO: 6, and a heavy chain complementary-determining region 3 (HCDR3) defined by SEQ ID NO: 7.
  • LCDR1 light chain complementary-determining region 1
  • LCDR2 light chain complementary-determining region 2
  • LCDR3 light chain complementary-determining region 3
  • the anti-OSMRB antibody comprises a light chain variable domain having an am o acid sequence at least 90% identical to SEQ ID NO: 4; and a heavy chain variable domain having an amino acid sequence at least 90% identical to SEQ ID NO: 3.
  • the light chain variable domain has the amino acid sequence set forth in SEQ ID NO: 4; and the heavy chain variable domain has the ammo acid sequence set forth in SEQ ID NO: 3.
  • the anti-OSMRB antibody comprises CHI , hinge and CH2 domains derived from an IgG4 antibody fused to a CH3 domain derived from an IgGl antibody.
  • the anti-OSMRB antibody comprises a light chain having an amino acid sequence at least 90% identical to SEQ ID NO: 2; and a heavy chain having an amino acid sequence at least 90% identical to SEQ ID NO: 1.
  • the light chain has the amino acid sequence set forth in SEQ ID NO: 2 and the heavy chain has the amino acid sequence set forth in SEQ ID NO: 1.
  • the one or more symptoms of atopic dermatitis are assessed by an Investigators’ Global Assessment (IGA) of atopic dermatitis. In some embodiments, the one or more symptoms of atopic dermatitis are assessed by an Eczema Area and Seventy Index (EASI). In some embodiments, the one or more symptoms of atopic dermatitis are assessed by SCORing Atopic Dermatitis. In some embodiments, the one or more symptoms of atopic dermatitis are assessed by atopic dermatitis area photographs.
  • IGA Global Assessment
  • EASI Eczema Area and Seventy Index
  • the one or more symptoms of atopic dermatitis are assessed by Body Surface Area Involvement (BSA) of Atopic Dermatitis in some embodiments, the one or more symptoms of atopic dermatitis are assessed by a Dermatology Life Quality Index (DLQI). In some embodiments, the one or more symptoms of atopic dermatitis are assessed by a Hospital Anxiety and Depression Scale (HADS). In some embodiments, the one or more symptoms of atopic dermatitis, such as sleep quality and sleep quantity', are assessed by actigraphy.
  • BSA Body Surface Area Involvement
  • DLQI Dermatology Life Quality Index
  • HADS Hospital Anxiety and Depression Scale
  • the one or more symptoms of atopic dermatitis, such as sleep quality and sleep quantity' are assessed by actigraphy.
  • the one or more symptoms of atopic dermatitis are assessed by a quantitative numerical pruritus scale, e.g., Pruritus Numerical Rating Scale (NRS), Visual Analogue Scale (VAS) or Verbal Rating Scale (VRS).
  • the administration of an anti-OSMRp antibody results in a statistically-significant drop on a quantitative numerical pruritus scale.
  • the administration of an anti-OSMRp antibody results in at least one of an improvement in the subject’s quality of life, quality of sleep and quantity of sleep.
  • OS MRJ3 antibody to a subject who has a pruritic or inflammatory skin disease or disorder results in a decrease in Numerical Rating Score (NRS) compared to a control
  • control is a NRS indicative of a subject with comparable disease status without treatment.
  • control is a NRS in the subject prior to the treatment.
  • a control is the respective value of a disease parameter of a subject with comparable disease status but treated with a placebo.
  • NRS is decreased by at least 2-points, or by at least 3-points, or by at least 4-points, or by at least 5-points, or by at least 6 points, or by at least 7 points, or by at least 8 points. In some embodiments, the NRS is decreased by greater than 4-points In some embodiments, the NRS is deceased by at least 8 points. In some embodiments, die NRS is decreased by approximately 10% or more, approximately 20% or more, approximately 30% or more, approximately 40% or more, approximately 50% or more, approximately 60% or more, approximately 70% or more, approximately 75% or more, or approximately 80% or more. In some embodiments, the decrease in NRS is approximately 4 or more points in approximately 30% or more,
  • the decrease in NRS is approximately 40% or more, approximately 50% or more, or approximately 60% or more , approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-QSMRp antibody.
  • the decrease in NRS is approximately 5 points in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRJ3 antibody.
  • the decrease in NRS is 6 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody.
  • the decrease in NRS is approximately 7 points or more in approximately 30% or more,
  • the decrease in NRS is approximately 8 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS is approximately 9 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the sub j ects administered the anti-OSMRp antibody.
  • the decrease in NRS is approximately 10 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS occurs less than 5 weeks, or less than 4 weeks, or less than 3 weeks, or less than 2 weeks, or less than 1 week after the subject’s initial dose of the anti-OSMRp antibody. In some embodiments, the decrease in NRS is approximately 30 % or more, approximately 40% or more, approximately 50% or more, or approximately 60 % or more, approximately 70% or more, or approximately 80% or more, about 4 weeks after the subject’s initial dose of the anti-OSMRp antibody.
  • the method of treatment results in a decrease in pruritus
  • the method of treatment results in a decrease in EASI by at least 20% compared to a control EASI. In some embodiments, the method of treatment results in a decrease in sleep- loss VAS by at least 20% compared to a control VAS. in some embodiments, the method of treatment results in an improvement in Scoring of Active Dermatitis (SCORAD) compared to a control SCORAD. In some embodiments, the method of treatment results in an improvement in Dermatology Life Quality Index (DLQI) compared to a control DLQI. In some embodiments, the method of treatment results in an improvement in Hospital Anxiety and Depression Scale (HADS) compared to a control HADS1.
  • SCORAD Scoring of Active Dermatitis
  • DLQI Dermatology Life Quality Index
  • HADS Hospital Anxiety and Depression Scale
  • tire NRS is w'orst itch NRS (WI-NRS).
  • the NRS value is calculated as a weekly average. [0037] In various aspects and embodiments described herein, administering the anti-
  • OSMRp antibody results in improved sleep in a subject as evidenced by a decrease in sleep-loss VAS from a compared to a control.
  • control is a sleep-loss
  • the control is a sleep-loss VAS in the subject prior to the treatment.
  • the baseline is a sleep-loss VAS in the subject prior to the treatment.
  • the decrease in the sleep- loss VAS from the baseline is by at least 10%, or by at least 20%, or by at least 30%, or by at least 40%, or by at least 50%, or by at least 60%, or by at least 70%, or by at least 80%, or by at least 90%.
  • the decrease in the sleep-loss VAS occurs less than 5 weeks, or less than 4 weeks, or less than 3 weeks, or less than 2 weeks, or less than 1 week after the subject’s initial dose of the anti-OSMR-b antibody.
  • the sleep-loss VAS value is calculated as a weekly average.
  • OSMRp antibody results in a decrease in EASI compared to a control.
  • the control is an EASI indicative of a subject with comparable disease status without treatment.
  • the control is an EASI in the subject prior to tire treatment.
  • the control is an EASI in the subject prior to the treatment.
  • the decrease in EASI from the baseline is by at least 10%, or by at least 20%, or by at least 30%, or by at least 40%, or by at least 50%, or by at least 60%, or by at least 70%, or by at least 80%, or by at least 90%.
  • the decrease in EASI occurs less than 5 weeks, or less than 4 weeks, or less than 3 weeks after the subject’s initial dose of the a ti-OSMRp antibody.
  • the EASI value is calculated as a weekly average.
  • OSMRp antibody results in two or more of: a decrease in pruritus Numerical Rating Score (NRS) by at least 4-points compared to a control NRS; a decrease in EASI by at least 20% compared to a control EASI: a decrease in sleep-loss VAS by at least 20% compared to a control VAS; an improvement in Scoring of Active Dermatitis (SCORAD) compared to a control S CORAD; an improvement in Dermatology Life Quality Index (DLQI) compared to a control DLQ1: and an improvement in Hospital Anxiety and Depression Scale (HADS) compared to a control HADS.
  • administering the anti-OSMRB antibody results in three or more, four or more, five or more, or six or more of the above identified decreases and improvements.
  • OSMRp antibody results in a decrease in pruritus Numerical Rating Score (NRS) by at least 4- points compared to a control NRS, and a decrease in EASI by at least 20% compared to a control EASE
  • NRS pruritus Numerical Rating Score
  • OSMRp antibody results in a decrease in pruritus Numerical Rating Score (NRS) by at least 4- points compared to a control NRS, and a decrease in sleep-loss VAS by at least 20% compared to a control VAS.
  • NRS pruritus Numerical Rating Score
  • OSMRp antibody results in a decrease in sleep-loss VAS by at least 20% compared to a control VAS, and a decrease in EASI by at least 20% compared to a control EASI.
  • OSMRp antibody results in a decrease in pruritus Numerical Rating Score (NRS) by at least 4- points, 5-points, 6-points, 7-points, 8-points, or 9-points compared to the control NRS.
  • NRS pruritus Numerical Rating Score
  • OSMRB antibody results in a decrease in EASI by at least 30%, or by at least 40%, or by at least 50%, or by at least 60%, or by at least 70%, by at least 75%, or by at least 80%, or by at least 90% compared to the control EASE
  • administering the anti-OSMRB antibody to subjects results in a decrease in EASI score by 50% (i.e., EASI-50) in approximately 30% or more of the subjects, in approximately 35% or more of the subjects, approximately 40% or more of the subjects, in approximately 45% or more of the subjects, approximately 50% or more of the subjects, in approximately 55% or more of the subjects, approximately 60% or more of the subjects, in approximately 65% or more of the subjects, approximately 70% or more of the subjects, in approximately 75% or more of the subjects, in approximately 80% or more of the subjects, or approximately 85% or more of the subjects.
  • administering the anti-OSMRB antibody to subjects results in a decrease in EASI score by 75% (i.e., EASI-75) in approximately 30% or more of the subjects, in approximately 35% or more of the subjects, approximately 40% or more of the subjects, in approximately 45% or more of the sub j ects, approximately 0% or more of the subjects, in approximately 55% or more of the subjects, approximately 60% or more of the subjects, in approximately 65% or more of the subjects, approximately 70% or more of the subjects, in approximately 75% or more of the subjects, in approximately 80% or more of the subjects, or approximately 85% or more of the subjects.
  • OSMRB antibody results in a decrease in sleep-loss VAS by at least 30%, or by at least 40%, or by at least 50%, or by at least 60%, or by at least 70%, or by at least 80%, or by at least 90% compared to the control VAS.
  • control is a value indicative of a respective parameter (e.g., NRS, EASI, VAS, SCORAD, DLQI, or HADS) in a subject with comparable disease status without treatment.
  • control is a value indicative of a respective parameter (e.g., NRS, EASI, VAS, SCORAD, DLQI, or HADS) in a subject prior to the treatment.
  • control is a value indicative of a respective parameter (e.g., NRS, EASI, VAS, SCORAD, DLQI, or HADS) in a subject with comparable disease status but treated with a placebo
  • a respective parameter e.g., NRS, EASI, VAS, SCORAD, DLQI, or HADS
  • the present invention provides methods for treating prurigo nodularis, comprising a step of administering to a subject in need of treatment an anti ⁇ OSMR antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptom s of prurigo nodul ari s relative to a control.
  • the subject presents with pruritic hyperkeratotic nodules.
  • the prurigo nodularis is idiopathic. In some embodiments, the prurigo nodularis is not associated with any other underlying co-morbidities.
  • the prurigo nodularis is associated with one or more underlying co-morbidities.
  • 1L-31 expression level is elevated in the subject relative to a control. In some embodiments, IL-31 expression level is not elevated in the subject relative to a control. In some embodiments, IL-31 expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the IL-31 expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy sub j ect, who is not diagnosed with a pruritic disease or condition. In some embodiments, IL-31Ra expression level is elevated in the subject relative to a control.
  • OSM expression level is elevated in the subject relative to a control.
  • OSMRB expression level is elevated in the subject relative to a control.
  • OSMRfl expression level is not elevated in the subject relative to a control.
  • OSMRji expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the OSMRp expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not di agnosed with a pruritic disease or condition.
  • OSMRp in the subject is determined via skin biopsy from hyperkeratotic nodules.
  • the control is a healthy subject who is not diagnosed with a pruritic disease.
  • the subject in need of treatment has a score on a pruritus NRS greater than or equal to 5. in some embodiments, the subject in need of treatment has a score on a pruritus NRS greater than or equal to 7.
  • the subject in need of treatment has elevated MCP-
  • 1/CCL2 levels in comparison to a control subject.
  • treating results in a reduction of MCP-1/CCL2 levels in the subject.
  • treating results in a reduction of MCP-1/CCL2 levels in the subject equivalent to levels in a healthy subject. In some embodiments, treating results in a reduction of MCP-1/CCL2 levels in the subject equivalent to levels in a control subject who does not have the disease.
  • the invention provides method s of treating atopi c dermatitis comprising a step of administering to a subject in need of treatment an anti-OSMRp antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of atopic dermatitis relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • the subcutaneous administration is through a subcutaneous pump.
  • the therapeutically effective dose comprises an initial bolus or loading dose.
  • the therapeutically effective dose comprises a maintenance dose. In some embodiments, the therapeutically effective dose comprises an initial bolus or loading dose, followed by at least one maintenance dose. In some embodiments, the therapeutically effective dose is an initial bolus or loading dose, and wherein the method further comprises administering at least one maintenance dose. In some embodiments, the step of administering comprises an initial bolus or loading dose, followed by at least one maintenance dose. In some embodiments, tire initial bolus or loading dose is greater than the at least one maintenance dose. In some embodiments, the initial bolus or loading dose is at least one fold, two fold, three fold, four fold or five fold greater in dosage than the dosage of the at least one maintenance dose. In some embodiments, the initial bolus or loading dose is two fold greater in dosage than the dosage of the at least one maintenance dose.
  • the one or more symptoms of atopic dermatitis in the subject before the treatment comprises a score on a pruritus NRS greater than or equal to 4, or an equivalent assessment using a quantitative numerical pruritus scale. In some embodiments, the one or more symptoms of atopic dermatitis in the subject before the treatment comprises a score on a pruritus NRS greater than or equal to 7, or an equivalent assessment using a quantitative numerical pruritus scale. In some embodiments, the subject m need of treatment has been diagnosed with moderate to severe atopic dermatitis, wherein moderate to severe atopic dermatitis comprises IGA of 3 or 4 and BSA involvement of approximately 10% or more.
  • control is indicative of the one or more symptoms of atopic dermatitis in a control subject with the same disease status without treatment. In some embodiments, the control is indicative of the one or more symptoms of atopic dermatitis in a control subject with the same disease status that was administered a placebo.
  • the administration results in no serious adverse effects in the subject. In some embodiments, the administration does not result in one or more of peripheral edema, exacerbation of atopic dermatitis, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, and dry eye.
  • the present invention provides methods of treating uremic pruritus, comprising a step of administering to a subject in need of treatment an anti ⁇ OSMRJ3 antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of uremic pruritus relative to a control .
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection. In some embodiments, the subcutaneous administration is through a subcutaneous pump. In some embodiments, the step of administering comprises an initial bolus or loading dose, followed by at least one maintenance dose. In some embodiments, the initial bolus or loading dose is greater than the at least one maintenance dose. In some embodiments, the initial bolus or loading dose is at least one fold, two fold, three fold, four fold or five fold greater in dosage than the dosage of the at least one maintenance dose. In some embodiments, the initial bolus or loading dose is two fold greater in dosage than the dosage of the at least one maintenance dose. As used herein, an initial bolus or loading dose, an initial loading dose and an initial dose are terms used interchangeably.
  • the one or more symptoms of uremic pruritus are assessed by a Dermatology Life Quality Index (DLQI). In some embodiments, the one or more symptoms of uremic pruritus are assessed by a Hospital Anxiety and Depression Scale (HADS). In some embodiments, the one or more symptoms of atopic dermatitis, such as sleep quality and sleep quantity, are assessed by actigraphy. In some embodiments, the administration of an anti- OSMRB antibody results in at least one of an improvement in the subject’s quality of life, quality of sleep and quantity of sleep.
  • the control is indicative of the one or more symptoms of uremic pruritus in the subject before the treatment.
  • the one or more symptoms of uremic pruritus in the subject before the treatment comprises a score on a pruritus NRS greater than or equal to 5, or an equivalent assessment using a quantitative numerical pruritus scale.
  • the one or more symptoms of uremic pruritus in the subject before the treatment comprises a score on a pruritus NRS greater than or equal to 7, or an equivalent assessment using a quantitative numerical pruritus scale.
  • the subject in need of treatment has end stage renal disease.
  • the subject in need of treatment is undergoing a hemodialysis regimen of at least one time-per-week.
  • the subject in need of treatment is undergoing a three-times-per ⁇ week
  • the control is indicative of the one or more symptoms of uremic pruritus in a control subject with the same disease status without treatment. In some embodiments, the control is indicative of the one or more symptoms of uremic pruritus in a control subject with the same disease status that was administered a placebo.
  • the present invention provides methods and compositions for treating pruritus in a subject suffering from a kidney disease.
  • the subject suffers from chronic kidney disease.
  • the subject having chronic kidney disease has not undergone dialysis.
  • the present invention provides a method and compositions for use in treating chronic kidney disease-associated pruritus in predialysis subjects.
  • the method comprises a step of administering to a subject in need of treatment an anti-OSMRp antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of chronic kidney disease associated pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • the subcutaneous administration is through a subcutaneous pump.
  • the step of administering comprises an initial bolus or loading dose, followed by at least one maintenance dose.
  • the administration interval is daily. In some embodiments, the administration interval is every other day. In some embodiments, the administration interval is multiple times a week. In some embodiments, the administration interval is once every week. In some embodiments, the administration interval is once every two weeks. In some
  • the administration interval is once every three weeks. In some embodiments, the administration interval is once every four weeks. In some embodiments, the adminisiraiiori interval is once every five weeks.
  • the treatment period is for as long as the subject is on hemodialysis.
  • the step of administering occurs one day before the subject undergoes hemodialysis. In other embodiments, the step of administering occurs during hemodialysis. In other embodiments, the step of administering occurs on the day of
  • the step of administering occurs within one day after hemodialysis.
  • the one or more symptoms of uremic pruritus are assessed by a quantitative numerical pruritus scale, e.g., Pruritus Numerical Rating Scale (NRS), Visual Analogue Scale (VAS) or Verbal Rating Scale (VRS).
  • a quantitative numerical pruritus scale e.g., Pruritus Numerical Rating Scale (NRS), Visual Analogue Scale (VAS) or Verbal Rating Scale (VRS).
  • the administration of an anti-OSMR antibody results in a statist! call y-significant drop on a quantitative numerical pruritus scale.
  • the present invention provides a method for treating pruritus in a subject having a disease or a condition selected from Chronic Idiopathic Pruritus (CIP), Chronic Idiopathic Urticaria (CIU), Chronic Spontaneous Urticaria (CSU), Cutaneous
  • the method comprising a step of administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce pruritus relative to a control.
  • the subject has CIP. In some embodiments, the subject has CSU. In some embodiments, the subject has CIU. In some embodiments, the subject has CA. In some embodiments, the subject has LSC. In some embodiments, the subject has PPs. In some embodiments, the subject has LP. In some embodiments, the subject has II. In some embodiments, the subject has MA. In some embodiments, the subject has BP.
  • the present invention provides a method of treating CIU, the method comprising administering to the subject in need of treatment an anti-OSMR antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce urticaria relative to a control.
  • the administration results in no serious adverse effects in the subject.
  • the administration does not result in one or more of peripheral edema, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, and dry eye.
  • the anti-OSMRB antibody comprises a light chain complementary-determining region 1 (LCDR1) defined by SEQ ID NO: 8, a light chain complementary-determining region 2 (LCDR2) defined by SEQ ID NO: 9, and a light chain complementary-determining region 3 (LCDR3) defined by SEQ ID NO: 10; and a heavy chain complementary -determining region 1 (HCDR1) defined by SEQ ID NO: 5, a heavy chain complementar -determining region 2 (HCDR2) defined by SEQ ID NO: 6, and a heavy chain complementary -determining region 3 (HCDR3) defined by SEQ ID NO: 7.
  • LCDR1 light chain complementary-determining region 1
  • HCDR2 light chain complementary-determining region 2
  • HCDR3 light chain complementary-determining region 3
  • the anti-OSMRB antibody comprises a light chain variable domain having an amino acid sequence at least 90% identical to SEQ ID NO: 4; and a heavy chain variable domain having an amino acid sequence at least 90% identical to SEQ ID NO: 3
  • the light chain variable domain has the amino acid sequence set forth in SEQ ID NO: 4
  • the heavy chain variable domain has the amino acid sequence set forth in SEQ ID NO: 3.
  • the anti-OSMRp antibody comprises CHI , hinge and CH2 domains derived from an IgG4 antibody fused to a CHS domain derived from an IgGl antibody.
  • the anti-OSMRp antibody comprises a light chain having an amino acid sequence at least 90% identical to SEQ ID NO: 2; and a heavy chain having an amino acid sequence at least 90% identical to SEQ ID NO: 1.
  • the light chain has the amino acid sequence set forth in SEQ ID NO: 2; and the heavy chain has the amino acid sequence set forth in SEQ ID NO: 1.
  • the invention provided herein allows for treating a pruritic or inflammatory skin disease or disorder by using a therapeutically effective dose of anti-OSMRJ3 antibody.
  • the invention allows for treating a pruritic or inflammatory skin disease or disorder by using a therapeutically effective dose of anti-OSMRJ3 antibody.
  • therapeutically effective dose is equal to or greater than about 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 0.6 mg/kg, 0.8 mg/kg, 0.9 mg/kg, 1 mg/kg, 1.2 mg/kg, 1.5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 8.5 mg/kg, 9 mg/kg, 9.5 mg/kg, 10 mg/kg, 10.5 mg/kg, 11 mg/kg, 1 1.5 mg/kg, 12 mg/kg, 12.5 mg/kg, 13 mg/kg, 13.5 mg/kg, 14 mg/kg, 14.5 mg/kg, 15 mg/kg, 15.5 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg or 20 mg/kg.
  • the therapeutically effective dose is about between about 20 mg/kg and about 50 mg/kg. In some embodiments, the therapeutically effective dose is about 50 mg/kg and about 75 mg/kg. In some embodiments, the therapeutically effective dose is about between 75-100 mg/kg. In some embodiments, the therapeutically effective dose is about between 100 mg/kg and 125 mg/kg. In some embodiments, the therapeutically effective dose is about between 125 mg/kg and about 150 mg/kg. In some embodiments, the therapeutically effective dose is about between 175 mg/kg and 200 mg/kg.
  • the therapeutically effective dose is approximately 3-20 mg/kg, approximately 4-20 mg/kg, approximately 5-20 mg/kg, approximately 6-20 mg/kg, approximately 7-20 mg/kg, approximately 8-20 mg/kg, approximately 9-20 mg/kg,
  • a pharmaceutically acceptable amount approximately 3-11 mg/kg, approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mgkg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • a pharmaceutically acceptable amount approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mgkg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • therapeutically effective dose is about 5 mg/kg. In some embodiments, a therapeutically effective dose is about 10 mg/kg.
  • the therapeutically effective dose is equal to or greater than 20 mg/kg, 25 mg/kg, 30 mg/kg, 35 mg/kg, 40 mg/kg, 45 mg/kg, or 50 mg/kg.
  • the therapeutically effective dose is equal to or greater than 50 mg/kg, 100 mg/kg, 150 mg/kg, 200 mg/kg, 250 mg/kg, 300 mg/kg, 350 mg/kg, 400 mg/kg, 450 mg/kg, 500 mg/kg, 550 mg/kg, 600 mg/kg, 650 mg/kg, 700 mg/kg, 750 mg/kg, 800 mg/kg, 850 mg kg, 900 mg/kg, 950 mg/kg, or 1000 mg/kg. [0081] In some embodiments, the therapeutically effective dose is approximately 50-
  • a loading dose is about 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 21 mg/kg, 22 mg/kg, 23 mg/kg, 24 mg/kg, or 25 mg/kg.
  • a maintenance dose is administered after administration of the loading dose.
  • a loading dose is between about 5 mg/kg and 25 mg/kg and a maintenance dose is between about 2.5 mg/kg and 7.5 mg/kg.
  • the maintenance dose is about 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 3.5 mg/kg, 4.0 mg/kg, 4.5 mg/kg, 5.0 mg/kg, 5.5 mg/kg, 6.0 mg/kg, 6.0 mg/kg, 6.5 mg/kg, 7.0 mg/kg, or 7.5 mg/kg.
  • a loading dose is about 10 mg/kg and maintenance dose is about 5 mg/kg
  • OSMRP antibody to a subject who has a pruritic or inflammatory ' skin disease or disorder results in a decrease in Numerical Rating Score (NRS) compared to a control.
  • NRS Numerical Rating Score
  • the control is a NRS indicative of a subject with comparable disease status without treatment.
  • the control is a NRS m the subject prior to the treatment.
  • a control is the respective value of a disease parameter of a subject with comparable disease status but treated with a placebo.
  • NRS is decreased by at least 2-points, or by at least 3 -points, or by at least 4-points, or by at least 5 -points, or by at least 6 points, or by at least 7 points, or by at least 8 points. In some embodiments, the NRS is decreased by greater than 4-points.
  • the NRS is deceased by at least 8 points. In some embodiments, the NRS is decreased by approximately 10% or more, approximately 20% or more, approximately 30% or more, approximately 40% or more, approximately 50% or more, approximately 60% or more, approximately 70% or more, approximately 75% or more, or approximately 80% or more. In some embodiments, the decrease in NRS is approximately 4 or more points in approximately 30% or more,
  • the decrease in NRS is approximately 40% or more, approximately 50% or more, or approximately 60% or more , approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS is approximately 5 points in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS is 6 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS is approximately 7 points or more in approximately 30% or more,
  • the decrease in NRS is approximately 8 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS is approximately 9 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody.
  • the decrease in NRS is approximately 10 points or more in approximately 30% or more, approximately 40% or more, approximately 50% or more, or approximately 60% or more, approximately 70% or more, or approximately 80% or more, of the subjects administered the anti-OSMRp antibody. In some embodiments, the decrease in NRS occurs less than 5 weeks, or less than 4 weeks, or less than 3 weeks, or less than 2 weeks, or less than 1 week after the subject’s initial dose of the anti-OSMRp antibody. In some embodiments, the decrease in NRS is approximately 30 % or more, approximately 40% or more, approximately 50% or more, or approximately 60 % or more, approximately 70% or more, or approximately 80% or more, about 4 weeks after the subject’s initial dose of the anti-OSMRp antibody.
  • the invention provides a method of treating inflammation, the method comprising administering to a subject need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period such that one or more symptoms associated with inflammation are reduced in intensity, severity, or frequency or has delayed in onset.
  • the inflammation is TH2 mediated inflammation.
  • the inflammation is independent of IL-31.
  • the subject is suffering from an inflammatory disease, disorder or condition.
  • the subject is suffering from a chronic inflammatory disease.
  • the chronic inflammator ' disease is Chronic Idiopathic Urticaria (CIU) and the symptom associated with inflammation that is reduced in intensity, severity, or frequency or has delayed in onset is urticaria.
  • CUA Chronic Idiopathic Urticaria
  • an anti-OSMRB antibody described herein is administered in conjunction with an additional therapeutic agent.
  • the additional therapeutic agent is a topical corticosteroid (e.g., TCS).
  • the additional therapeutic agent is a topical calcineurin inhibitor.
  • the additional therapeutic agent is a topical antimicrobial and/or antiseptic.
  • the additional therapeutic agent is a topical antihistamine.
  • Figure 1A depicts an exemplary graph of the percent inhibition of scratching behavior monkeys 1 hour after 1L-31 challenge at 2, 8, 15, 21 and 29 days after dosing with an anti-OSMR antibody.
  • Figure IB depicts graphs of scratching behavior and serum concentration of an anti-OSMRp antibody in monkeys 1 hour after IL-31 challenge at 2, 8, 15, 21 and 29 days after dosing with the anti-QSMR[3 antibody.
  • Figure 2 depicts study design for determining safety and efficacy of tire anti-
  • OSMR antibody in a single dose dose-escalation study in healthy volunteers and atopic dermatitis patients.
  • Figure 3A-3D depicts change in pruritus with anti-QSMR[3 antibody treatment.
  • Figure 3 A indicates change in mean VAS pruritus score (+/- SEM) from baseline over the indicated period.
  • Figure 3B indicates the mean percent VAS pruritus score change from baseline (+/- SEM) over the indicated period.
  • Figure 3C indicates mean weekly average worst itch MRS (WI-NRS) over the indicated period.
  • Figure 3D indicates the mean percent change in weekly average WI-NRS from baseline (+/- SEIM). The data indicate greater reduction in patients receiving the antibody as compared to PBO within the first 4 weeks after administration, which persisted up to 8 weeks.
  • Figure 4 depicts percentage of subjects having clinically meaningful reduction in weekly average NRS (>4 points) after receiving single intravenous dose of 7.5 mg/kg anti- OSMR antibody (anti-OSMR[3 Ab) or placebo (PBO).
  • Figures 5A-5D depicts the number of subjects who responded with a particular magnitude of NRS reduction from baseline (>4) over 9 weeks after single intravenous dose of 7.5 mg/kg anti-QSMR[3 antibody (anti-OSMR Ab) or placebo.
  • Figure 5A and 5C show results from the anti-OSMR[3 antibody recipient group, and
  • Figure 5B and 5D show the results from the placebo group.
  • Figure 6A-6B shows improvement from sleeplessness over the course of the indicated observation period in subjects receiving single intravenous dose of 7.5 mg/kg anti- OSMR[3 antibodv (anti-OSMR[3 Ab) or placebo.
  • Figure 6A show's mean ( ⁇ SEM) sleep-loss VAS score change
  • Figure 6B show's Mean ( ⁇ SEM) Percent sleep-loss VAS score change from baseline.
  • Figures 7A-7B depicts the changes in EASI score as a measure of the disease severity. Subjects received single intravenous dose of 7.5 mg/kg anti-OSMRfl antibody (anti- OSMR Ab) or placebo. Figure 7A shows the mean scores ( ⁇ SEM); Figure 7B shows mean percent ( ⁇ SEM) EASI change from baseline.
  • Figures 8.4-8 B depict percent of subjects showing of clinically meaningful response of having a reduction in disease severity as measured by EASI score.
  • Subjects received single intravenous dose of 7.5 mg/kg anti-OSMRf) antibody (anti-OSMRp Ab) or placebo.
  • Figure 8A shows results for responders having a EASI reduction of 50% or more compared to baseline (EASI-50 score);
  • Figure 8B show's results for responders having a EASI reduction of 75% or more compared to baseline (EASI-75) respectively, over the study period after single intravenous dose of 7.5 mg/kg anti-OSMRp antibody (anti-OSMRff Ab) or placebo.
  • % indicated above each data point indicates the percent of subjects in the population of the group. Empty bars denote placebo subjects, solid bars denote anti-OSMRp Ab recipient subjects in Figures 8A and 8B
  • Figure 9A-9B shows SCQRAD values as an over clinical evaluation of change in the extent and severit of atopic dermatitis after subjects received single intravenous dose of 7.5 mg/kg anti-OSMRj) antibody (anti-OSMRfr Ab) or placebo.
  • Figure 9A show's mean SC OR AD values (+/- SEM) change from baseline over the indicated period.
  • Figure 9B shows mean percent SCORAD (+/- SEIM) change from baseline over the indicated period.
  • Figure 10A-10C depicts modeled PK parameters for subcutaneous
  • Figure 10A depicts simulated median values of anti-OSMR Ab concentration in plasma in various dosing regimens indicated in the inset.
  • Figure I0B depicts concentration profiles of anti-OSMRj) Ab in plasma over indicated time period after subcutaneous administered to atopic dermatitis patients.
  • HV healthy volunteers
  • AD atopic dermatitis patients
  • IV intravenous administration
  • SC subcutaneous administration.
  • Figure 10C depicts a range of simulations for various SC dosing regimens.
  • Figure 11 is a schematic that depicts IL-3 IRa, OSMRJ3 and L1FR signaling pathways.
  • Figure 12 is a series of graphs that depict MCP-1 protein levels in the supernatants of human epidermal keratinocytes (HEK) and human dermal fibroblasts (HDF) following treatment with OSM (50 ng/niL) for 6 hours and 24 hours (panel A).
  • Panel B shows MCP-1 mRNA levels relative to the housekeeping gene 18S mRNA. The data show strong upregulation of MCP-1 levels following addition of OSM.
  • Figure 13 is a series of graphs that depict MCP-1 protein levels in the supernatants of cultured HEK and HDF cells following addition of 50 ng/rnL OSM, 50 ng/mL LIF, or 100 ng/mL of IL-31 in combination with increasing concentrations of IL-4 (panel A) or IL-13 (panel B).
  • Figure 14 is a series of graphs that shows the mRNA expression levels of 1L-
  • Figure 15 is a series of graphs that show' the effect of adding either anti-OSMRp antibody (panel A), anti-IL-3 IRa antibody (panel B) or an isotype control (panel C) at increasing concentrations to cultured HEK cells that had been treated with OSM at 50 ng/mL.
  • Figure 16 is a series of graphs that show the effect of adding either anti-OSMRp antibody (panel A), anti-IL-3 IRa antibody (panel B) or an isotype control (panel C) at increasing concentrations to cultured HEK cells that had been stimulated with OSM at 50 ng/mL and IL-4 (at either 5 or 20 ng/mL concentrations).
  • Figure 17 is a series of graphs that depict the results of IL-31 mRNA expression measurements obtained from non-lesional (NL) and lesional (LS) skin biopsies of subjects who have prurigo nodularis (PN) or atopic dermatitis (AD).
  • NL non-lesional
  • LS lesional
  • PN prurigo nodularis
  • AD atopic dermatitis
  • Figure 18 is a series of graphs that depict the results of IL-31 mRNA expression measurements (panel A) or OSM expression measurements (panel B) obtained from PN, AD or from healthy control subject (HC) skin biopsies.
  • Figure 19 is a series of graphs that show the results of OSM (panel A) and IL-31
  • Figure 20 is a series of graphs that show' quantitation of immunohistochemistry observations in skin samples obtained from PN subjects.
  • panels A-D show quantitation of cells (cells/iim2) found in the dermis that are positive for OS RB (panel A), OSM (panel B), IL-31 (panel C), or IL-3 IRa (panel D) in samples obtained from PN subjects in comparison to healthy controls.
  • panels E-FI are graphs that show' percent positivity for IL-31 (panel E), OSM (panel (F), IL-31a (panel G), or OSMR[3 (panel H) in skin samples obtained from NL or LS skin biopsies of PN subjects.
  • Figure 21 is a series of graphs that show quantitation of immunohistochemistry observations (IL-31, panel A; OSM, panel B; IL-31Ra, panel C; 08MKb, panel D) obtained from NL skin biopsies, and from LS skin biopsies from subjects who had either WI-NRS ⁇ 7 or WI-NRS>7
  • FIG 22 is a series of graphs that show OSMR mRNA (panels A and B) or protein (panel C) expression levels obtained from control skin samples or skin samples obtained from chronic idiopathic urticaria patients.
  • Panels A and B show OSMRJ3 mRNA expression levels as detected using RNAscope® or NanoString® technologies, respectively.
  • Panel C shows QSMRf ⁇ protein expression levels as determined by immunohistochemistry.
  • FIG 23 is a series of graphs that show OSMRfi mRNA levels in subjects who have Lichen Simplex Chronicus (LSC). OSMRfi mRNA levels in samples obtained from LSC patients was assessed by NanoString (panel A) and RNAscope (panel B) technology.
  • Figure 24 is a graph that show's OSMRj3 mRNA levels in subjects who have
  • Figure 25 is a graph that shows QSMRjl mRNA levels in subjects who have
  • OSMRji mRNA levels in samples obtained from CIP patients was assessed using NanoString technology.
  • Figure 26 depicts a study design for determining safety and efficacy of the anti-
  • OSMR antibody in a placebo-controlled repeated-single-dose Phase lb clinical trial in patients with moderate-to-severe atopic dermatitis.
  • Figure 27A-27D depicts change in pruritus and sleep loss with anti-OSMRp antibody treatment in atopic dermatitis patients.
  • Figure 27A is a graph that shows percent change in weekly average worst itch NRS (Wl-NRS) from the baseline over the indicated period.
  • Figure 27B depicts the number of subjects who responded with a particular magnitude of NRS reduction from baseline (>4) over a 12 week period with a weekly 360 mg dose anti-OSMRJ3 antibody (left panel) or placebo (right panel).
  • Figure 27C is a graph that shows percent change in mean VAS pruritus score from baseline over the indicated period.
  • Figure 27D is a graph that show's the mean percent sleep loss VAS pruritus score change from baseline over the indicated
  • Figure 28A-28B shows bar graphs of baseline subject characteristics by retrospective groupings.
  • Figure 28A show's percent of patients with prior TCS use in placebo group, treated group (OSMRB Ab; All), treated group who did not flare (OSMRB Ab; subjects who did not require rescue TCS), and treated group who flared (OSMRB Ab; subjects who were rescued with TCS).
  • Figure 28B shows percent of subjects with serum IgE levels greater than 158 IU/ml (Upper Limit Normal; ULN) placebo group, treated group (OSMRB Ab: All), treated group who did not flare (OSMRB Ab; subjects who did not require rescue TCS), and treated group who flared (OSMRB Ab; subjects who were rescued with TCS). Subjects who flared tended to use TCS prior to treatment or have IgE levels above ULN.
  • Figure 29 shows a graphical representation of the median serum IgE levels at the indicated study period. Subjects who flared tend to have higher IgE levels.
  • Figure 30 shows graphical representations of median percent eosinophil count in the blood of subjects at the indicated study period, and mean and median eosinophil + leukocyte count in the blood of subjects at the indicated study period.
  • Figure 31 depicts percent change in weekly average worst itch NRS (WI-NRS) from the baseline over the indicated period for each of the following groups: Placebo group without prior TCS use, placebo group with prior TCS use, treated group without prior TCS use and treated group with TCS use.
  • WI-NRS weekly average worst itch NRS
  • Figure 32 depicts percent change in weekly average worst itch NRS (WI-NRS) from the baseline over the indicated period for each of the following groups: Placebo group with equal to or less than IgE level of 300 IU/ml at baseline, placebo group with greater than IgE level of 300 IU/ml at baseline, treated group with equal to or less than IgE level of 300 IU/ml at baseline and treated group with greater than IgE level of 300 IU/ml at baseline.
  • WI-NRS weekly average worst itch NRS
  • amino acid refers to any compound and/or substance that can be incorporated into a polypeptide chain.
  • an amino acid has the general structure 1 1L C(i !(!! ⁇ ( OOl l.
  • an amino acid is a naturally occurring amino acid.
  • an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a d-amino acid; in some embodiments, an amino acid is an 1-ammo acid.
  • Standard amino acid refers to any of the twenty standard 1-amino acids commonly found in naturally occurring peptides.
  • Nonstandard amino acid refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source.
  • synthetic amino acid encompasses chemically modified amino acids, including but not limited to salts, amino acid derivatives (such as amides), and/or substitutions.
  • Amino acids, including carboxyl- and/or amino-terminal amino acids in peptides, can be modified by methylation, amidation, acetylation, protecting groups, and/or substitution with other chemical groups that can change the peptide s circulating half-life without adversely affecting their activity.
  • Amino acids may participate in a disulfide bond.
  • Amino acids may comprise one or posttranslationa!
  • amino acid is used interchangeably with“amino acid residue,” and may refer to a free amino acid and/or to an amino acid residue of a peptide lt will be apparent from the context in which the term is used whether it refers to a free amino acid or a residue of a peptide
  • Amelioration is meant the prevention, reduction or palliation of a state, or impro vement of the state of a subject. Amelioration includes, but does not require complete recovery or complete prevention of a disease condition. In some embodiments, amelioration includes increasing levels of relevant protein or its activity that is deficient in relevant disease tissues.
  • the term“approximately” or“about,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value.
  • the term“approximately” or“about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3 %, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
  • Control ⁇ As used herein, the term control is a reference based on which a change is determined.
  • a control is the respective value of a disease parameter of a subject with comparable disease status, but without treatment.
  • a control is the respective value of a disease parameter of a subject with comparable disease status but treated with a placebo.
  • a control is tire respective value of a disease parameter of a subject prior to treatment (also referred to as baseline).
  • a control is a reference value indicative of a disease parameter without treatment based on collective knowledge, or historical data.
  • delivery As used herein, the term“deliver ⁇ ” encompasses both local and systemic deliver ⁇ 7 .
  • Half-life As used herein, the term“half-life” is the time required for a quantity such as nucleic acid or protein concentration or activity to fall to half of its value as measured at the beginning of a time period.
  • “reduce,” or grammatical equivalents indicate values that are relative to a baseline measurement, such as the respective value of a disease parameter of a subject with comparable disease status, but without a treatment described herein, or a measurement in a subject (or multiple control subjects) in the absence of the treatment described herein, e.g., a subject who is administered a placebo.
  • a control is a reference value indicative of a disease parameter without treatment, based on collective knowledge, or historical data.
  • Substantial identity is used herein to refer to a comparison between amino acid or nucleic acid sequences. As will be appreciated by those of ordinary skill in tire art, two sequences are generally considered to be “substantially identical” if they contain identical residues in corresponding positions. As is well known in this art, amino acid or nucleic acid sequences may be compared using any of a variety of algorithms, including those available in commercial computer programs such as BLASTN for nucleotide sequences and BLAST?, gapped BLAST, and PSI-BLAST for amino acid sequences. Exemplary such programs are described in Altschul, et ah, Basic local alignment search tool, JMal.
  • two sequences are considered to be substantially identical if at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of their corresponding residues are identical over a relevant stretch of residues.
  • the relevant stretch is a complete sequence.
  • the relevant stretch is at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500 or more residues.
  • Suitable for subcutaneous delivery As used herein, the phrase "suitable for subcutaneous delivery” or “formulation for subcutaneous delivery” as it relates to the pharmaceutical compositions of the present invention generally refers to the stability, viscosity, tolerability and solubility properties of such compositions, as well as the ability of such compositions to deliver an effective amount of antibody contained therein to the targeted site of delivery.
  • the term“patient” refers to any organism to which a provided composition may be administered, e.g., for experimental, diagnostic, prophylactic, cosmetic, and/or therapeutic purposes. Typical patients include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and/or humans). In some embodiments, a patient is a human. A human includes pre- and post-natal forms. A“patient” is used interchangeably with “subject” where the subject has a disease and is administered either the antibody or a placebo.
  • compositions that, within the scope of sound medical judgment, are suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • Subject refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate).
  • a human includes pre- and post-natal forms.
  • a subject is a human being.
  • a subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease.
  • the term“subject” is used herein interchangeably with“individual” or “patient.”
  • a subject can be afflicted with or is susceptible to a disease or disorder but may or may not display symptoms of the disease or disorder.
  • the term“substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest.
  • One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result.
  • the term“substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
  • Systemic distribution or delivery As used herein, the terms“systemic distribution,”“systemic delivery,” or grammatical equivalent, refer to a delivery or distribution mechanism or approach that affect the entire body or an entire organism. Typically, systemic distribution or delivery is accomplished via body’s circulation system, e.g., blood stream.
  • Target tissues refers to any tissue that is affected by a disease or disorder to be treated.
  • target tissues include those tissues that display disease-associated pathology, symptom, or feature.
  • therapeutically effective amount As used herein, the term“therapeutically effective amount” of a therapeutic agent means an amount that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, diagnose, prevent, and/or delay the onset of the symptom(s) of the disease, disorder, and/or condition. It will be appreciated by those of ordinary skill in the art that a therapeutically effective amount is typically administered via a dosing regimen comprising at least one unit dose.
  • Treating refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. Treatment may be administered to a subject who does not exhibit signs of a disease and/or exhibits only early signs of the disease for the purpose of decreasing the risk of developing pathology associated with the disease.
  • Treatment period refers to the period during which the drug is treating the patient. Typically, the treatment period refers to the period between the first administration of anti-OSMRp antibody and the end of the last administration interval, when the next dose would otherwise be administered.
  • the present invention provides, among other things, methods of treating atopic dermatitis comprising a step of administering to a subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of atopic dermatitis relative to a control. Also provided are methods of treating uremic pruritus, comprising a step of administering to a subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of atopic dermatitis relative to a control.
  • Atopic dermatitis is a chronic inflammatory skin disease, characterized by
  • Th2 cell-mediated immune responses impaired skin barrier function, and bacterial colonization.
  • the pre valence of AD is about 20% in children and 1% to 10% in adults.
  • Approximately 20% of patients with AD have moderate to severe disease involving large body surface areas and suffer from chronic intense pruritus, leading to sleep deprivation and poor quality of life (Boguniewicz et al., 2011; Brandt et al., 201 1 ; Gittler et a , 2012; Silverberg et al., 2013).
  • severity of atopic dermatitis can assessed by various routine tests such as Investigators Global
  • EASI Eczema Area and Severity Index
  • SCORing Atopic Dermatitis In EASI scoring system, the extent of eczema and severity of the four signs (erythema, edema/papulation, excoriation, and lichemfication) in each of a body region (head and neck, trunk, upper extremities, and lower extremities) are accessed.
  • Topical corticosteroid and calc eurin inhibitors are used for the treatment of moderate to severe disease, but these therapies have limited efficacy, and prolonged use is associated with side effects.
  • systemic corticosteroids or cyclosporine though efficacious, are associated with significant toxicities (Ring et al., 2012: Sidbury et al., 2014).
  • IL-31 axis has been consistently shown to be up-regulated. Serum levels of IL-31 were elevated and correlated with AD disease severity in children (Ezzat et al., 201 1) and in adults (Raap et al., 2008). Increased IL-31 mRNA was observed in skin biopsies from AD and PN patients compared to healthy skin (Sonkoly et al., 2006); and IL-31, OSMRJ3, and IL-31 receptor a (IL-31Ra) staining was enhanced in AD skin (Nobbe et al, 2012).
  • IL-31 is produced by activated Th2 cells (Dillon et al., 2004), and its expression is induced by IL-4 (Stott et al., 2013). Accordingly, peripheral blood mononuclear cells (PBMC) from atopic donors produce more IL-31 upon activation compared with PBMCs from non-atopic donors (Stott et al., 2013). Once released, IL-31 participates in a feedback loop that perpetuates the inflammatory response in AD. IL-31 increases the production of IL-4, IL-5, and IL-1.3 in PBMCs from atopic donors and in nasal epithelial cells (Liu et al., 2015).
  • IL-31 synergizes with IL-4 in production of CCL2, VEGF, and, very importantly, in the induction of more IL-4, IL-5, and IL-13 (Ip et al., 2007; Stot et al., 2013; Liu et al., 2015).
  • Staphylococcus that occasionally infect the skin in AD.
  • Staphylococcal Enterotoxin B (SEB) and Staphylococcal a toxin, super antigens produced by Staphylococcus increase the production of IL-31 in PBMCs and skin of AD patients (Sonkoly et al., 2006; Niebuhr et al ., 2011), further reinforcing the vicious cycle of inflammation.
  • SEB Staphylococcal Enterotoxin B
  • Staphylococcal a toxin super antigens produced by Staphylococcus
  • increase the production of IL-31 in PBMCs and skin of AD patients Nonkoly et al., 2006; Niebuhr et al ., 2011
  • the inflammatory response is also reinforced on the cytokine receptor side.
  • Keratinocytes and skin-infiltrating macrophages in AD express IL- 3 IRcc; and SEB, TLR2 agonists (a cellular component of Staphylococcus), lFN-g, OSM, IL-4, and IL-13 upregulate the expression of IL-3 lRa on macrophages and keratinocytes (Bilsborough et al., 2006; Heise et al., 2009; Kasraie et ah, 2011; Edukulla et a , 2015).
  • OSM also plays an important role in AD pathology and echoes many of the functions of IL-31.
  • OSM is produced by skin infiltrating T cells in AD, and OSMR]3 levels are increased in the skin of AD patients (Boniface et ah, 2007).
  • OSMR OSMR
  • OSM is produced by macrophages and neutrophils under inflammatory conditions (Richards, 2013). Once produced, OSM induces the production of multiple cytokines: IL-4, IL- 5, 1L-13, 1L-6, 1L-12, tumor necrosis factor (TNF), and IL-10, and chemokmes (CXCL1,
  • HBD human-beta-defensin
  • HBD2 and HBD3 feed into the vicious cycle of inflammation by inducing the production of more OSM, IL-22, IL-4, IL-13, and TL-31 (Kanda et al., 2012). This cycle is further fueled by OSM upregulation of IL-4Rot (Mozaffarian et al., 2008: Fritz et al., 2009; Fritz et al., 2011).
  • IL-4Rot Mozaffarian et al., 2008: Fritz et al., 2009; Fritz et al., 2011.
  • 1L-31 and OSM reinforce the inflammatory’ response and compromise the skin barrier function in AD through multiple overlapping pathways.
  • an antibody such as the anti-OSMRp antibodies described herein, that antagonizes both IL-31 and OSM provides a therapeutic opportunity in AD through the inhibition of downstream signaling events stimulated by IL-31 and OSM, two cytokines that drive pruritus and inflammation
  • one or more symptoms of atopic dermatiti s are assessed by an
  • one or more symptoms of atopic dermatitis are assessed by an Eczema Area and Severity Index (EAS1). In some embodiments, one or more symptoms of atopic dermatitis are assessed by scoring atopic dermatitis (SCORAD). In some embodiments, one or more symptoms of atopic dermatitis are assessed by atopic dermatitis Area Photographs. In some embodiments, one or more symptoms of atopic dermatitis are assessed by Body Surface Area Invol vement (BSA) of Atopic Dermatitis. In some embodiments, one or more symptoms of atopic dermatiti s are assessed by a
  • DLQI Dermatology Life Quality Index
  • one or more symptoms of atopic dermatitis are assessed by a Hospital Anxiety and Depression Scale (HADS).
  • HADS Hospital Anxiety and Depression Scale
  • one or more symptoms of atopic dermatitis such as sleep quality and sleep quantity, are assessed by aetigraphy.
  • one or more symptoms of atopic dermatitis are assessed by a quantitative numerical pruritus scale, e.g.. Pruritus Numerical Rating Scale (NRS), Visual Analogue Scale (VAS) or Verbal Rating Scale (VRS).
  • NFS Pruritus Numerical Rating Scale
  • VAS Visual Analogue Scale
  • VRS Verbal Rating Scale
  • atopic dermatitis is treated by
  • atopic dermatitis refers to amelioration of one or more symptoms associated with atopic dermatitis, prevention or delay of the onset of one or more symptoms of atopic dermatitis, and/or lessening of the severity or frequency of one or more symptoms of atopic dermatitis.
  • the terms, "treat” or “treatment,” as used in the context of atopic dermatitis herein, refers to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of atopic dermatitis.
  • the terms, “treat” or “treatment,” as used in the context of atopic dermatitis herein refers to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of atopic dermatitis.
  • administration of an anti-OSMRfi antibody results in a statistically-significant drop on a quantitative numerical pruritus scale.
  • the step of administering compri ses subcutaneous administration.
  • subcutaneous administration is through subcutaneous injection.
  • subcutaneous administration is through a subcutaneous pump.
  • subcutaneous injection of the anti-OSMRp antibody can be performed in the upper arm, the anterior surface of the thigh, the lower portion of the abdomen, the upper back or the upper area of the buttock.
  • the site of injection is rotated.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the effect of an anti-OSMRp antibody on atopic dermatitis is measured relative to a control.
  • a control is indicative of the one or more symptoms of atopic dermatitis in the subject before the treatment.
  • one or more symptoms of atopic dermatitis in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 5.
  • one or more symptoms of atopic dermatitis in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 7.
  • a subject in need of treatment has been diagnosed with atopic dermatitis for at least one year.
  • a subject in need of treatment has been diagnosed with moderate to severe atopic dermatitis.
  • moderate to severe atopic dermatitis comprises an IGA score of 3 or 4.
  • moderate to severe atopic dermatitis comprises a BSA involvement of approximately 10% or more.
  • moderate to severe atopic dermatitis comprises an IGA score of 3 or 4 and BSA involvement of approximately 10% or more.
  • a control is indicative of the one or more symptoms of atopic dermatitis in a control subject with the same disease status without treatment.
  • the control is indicative of the one or more symptoms of atopic dermatitis in a control subject with the same disease status that was administered a placebo.
  • subjects naive to topical corticosteroid appeared to respond to the anti-OSMRp antibody treatment more favorably on the pruritus endpoint and were less likely to have an atopic dermatitis flare (e.g., eczema flare) during the treatm nt period than subject who had used TCS prior to the treatment period. Therefore, in some embodiments, a subject who has not received prior treatment with a topical corticosteroid is selected for the treatment of atopic dermatitis. In some embodiments, a subject is selected based on the subject’s serum IgE level.
  • a subject who has a serum TgE level lower than 300 lU/ml is selected for the treatment of atopic dermatitis.
  • the subject who has an IgE level lowur than the Upper Limit of Normal (ULN 158 !U/ml) is selected.
  • the subject is selected based on the eosinophil count in the subject’s blood.
  • the subject is selected when the blood eosinophil count is lower than 0.4x1 Q 9 /L.
  • the subject is selected when the blood eosinophil count is 5% or less of the total leukocyte count m the blood.
  • a subject in need of treatment has elevated levels of one or more cytokines associated with the OSMRp signaling pathway in comparison to a healthy subject. Accordingly, in some embodiments, the subject in need of treatment has elevated levels of one or more of IL-31, OSM, TL-31Ra, and OSMRp in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated levels of one or more of IL-31 in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated levels of one or more of OSM in comparison to a healthy subject. In some
  • the subject in need of treatment has elevated levels of one or more of IL-3 IRa in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated level s of one or more of OSMRp in comparison to a healthy subject. [0155] In some embodiments, treating the subject in need thereof results in a decrease or stabilization of MCP-1/CCL2 levels in the sub j ect. Accordingly, in some embodiments, treating a subject in need thereof results in a decrease of MCP-l levels in comparison to the diseased state. In some embodiments, treating a subject m need thereof results in stabilization of MCP-l levels. By‘stabilization” is meant that the levels of MCP-l remain about the same and do not increase or decrease. In some embodiments, treating a subject results in reduced MCP-l levels in lymphocytes and/or endothelial cells.
  • the subject in need of treatment has WI-NRS scores of about 4, about 5, about 6, about 7, about 8 or above. Accordingly, in some embodiments, the subject in need of treatment has WI-NRS score of about 4. In some embodiments, the subject in need of treatment has WI-NRS score of about 5. In some embodiments, tire subject in need of treatment has WI-NRS score of about 6. In some embodiments, the subject in need of treatment has WI-NRS score of about 7. In some embodiments, the subject in need of treatment has WI- NRS score of about 8. In some embodiments, the subject in need of treatment has WI-NRS score of more than 8.
  • a subject is selected for treatment who has MCP-1/CCL2 levels greater than found in a healthy individual. In some embodiments, the subject selected for treatment does not have elevated levels of MCP-1/CCL2 in comparison to a healthy individual.
  • IL-31 expression level is elevated in the subject relative to a control . In some embodiments, IL-31 expression level is not elevated in the subject relative to a control. In some embodiments, IL-31 expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the IL-31 expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition. In some embodiments, IL-3 IRa expression level is elevated in the subject relative to a control. In some embodiments, OSM expression level is elevated in the subject relative to a control.
  • OSMRp expression level is elevated in the subject relative to a control . In some embodiments, OSMRp expression level is not elevated in the subject relative to a control. In some embodiments, OSMRp expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the OSMRp expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition.
  • a therapeutically effective dose of an anti-QSMRp antibody for treating atopic dermatitis can occur at various dosages.
  • a therapeutically effective dose is equal to or greater than about 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 0 6 mg/kg, 0 8 mg/kg, 0 9 mg/kg, 1 mg/kg, 1.2 mg/kg, 1 5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 8.5 mg/kg, 9 mg/kg, 9.5 mg/kg, 10 mg/kg, 10.5 mg/kg, 11 mg/kg, 11.5 mg/kg, 12 mg/kg, 12.5 mg/kg, 13 mg/kg, 13.5 mg/kg, 14
  • a therapeutically effective dose is equal to or greater than 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg or 20 mg/kg.
  • a therapeutically effective dose is approximately 0.1-20 mg/kg, approximately 0.3-20 mg/kg, approximately 0.5-20 mg/kg, approximately 0.75-20 mg/kg, approximately 1-20 mg/kg, approximately 1.5-20 mg/kg, approximately 2-20 mg/kg, approximately 2.5-20 mg/kg, approximately 3-20 mg/kg, approximately 3.5-20 mg/kg, approximately 4-20 mg/kg, approximately 4.5-20 mg/kg, approximately 5-20 mg/kg, approximately 5 5-20 mg/kg, approximately 6-20 mg/kg, approximately 6.5-20 mg/kg, approximately 7-20 mg/kg, approximately 7.5-20 mg/kg, approximately 8-20 mg/kg, approximately 8.5-20 mg/kg, approximately 9-20 mg/kg, approximately 9.5-20 mgkg, approximately 10-20 mg/kg, approximately 10.5-20 mg/kg.
  • a therapeutically effective dose is approximately 3-20 mg/kg, approximately 4-20 mg/kg, approximately 5-20 mg/kg, approximately 6-20 mg/kg, approximately 7-20 mg/kg, approximately 8-20 mg/kg, approximately 9-20 mg/kg,
  • a pharmaceutically acceptable amount approximately 3-14 mg/kg, approximately 3-13 mg/kg, approximately 3-12 mg/kg, approximately 3-11 mg/kg, approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mg/kg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, or approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • a pharmaceutically acceptable amount approximately 3-14 mg/kg, approximately 3-13 mg/kg, approximately 3-12 mg/kg, approximately 3-11 mg/kg, approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mg/kg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, or approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • therapeutically effective dose is about 5 mg/kg. In some embodiments, a therapeutically effective dose is about 10 mg/kg.
  • the therapeutically effective dose is equal to or greater than 50 mg/kg, 100 mg/kg, 150 mg/kg, 200 mg/kg, or 250 mg/kg, 300 mg/kg, 310 mg/kg, 320 mg/kg, 330 mg/kg, 340 mg/kg, 350 mg/kg, 360 mg/kg, 370 mg/kg, 380 mg/kg, 390 mg/kg, 400 mg/kg, 450 mg/kg, 500 mg/kg, 550 mg/kg, 600 mg/kg, 650 mg/kg, 700 mg/kg, 710 mg/kg, 720 mg/kg, 730 mg/kg, 740 mg/kg, 750 mg/kg, 800 mg/kg, 850 mg/kg, 900 mg/kg, 950 mg/kg, or
  • a therapeutically effective dose is approximately 50-1,000 mg/kg, approximately 100-1,000 mg/kg, approximately 150-1,000 mg/kg, approximately 200- 1,000 mg/kg, approximately 250-1,000 mg/kg, approximately 300-1,000 mg/kg, approximately 350-1,000 mg/kg, approximately 400-1,000 mg/kg, approximately 450-1,000 mg/kg, approximately 500-1,000 mg/kg, approximately 550-1 ,000 mg/kg, approximately 600-1,000 mg/kg, approximately 650-1,000 mg/kg, approximately 700-1,000 rng/kg, approximately 750- 1,000 mg/kg, approximately 800-1,000 mg/kg, approximately 850-1,000 mg/kg, approximately 900-1,000 mg/kg, approximately 950-1,000 mg/kg, approximately 50-950 mg/kg, approximately 50-900 mg/kg, approximately 50-850 mg/kg, approximately 50-800 mg/kg, approximately 50- 750 mg/kg, approximately 50-700 mg/kg, approximately 50-650 mg/kg, approximately 50-600 mg/kg, approximately 50-550 mg/kg, approximately 50-500
  • administering comprises an initial bolus or loading dose, followed by at least one maintenance dose.
  • the initial bolus or loading dose is greater than the at least one maintenance dose.
  • the initial bolus or loading dose is at least one-fold, two-fold, three-fold, four fold or five-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is two-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is 720 mg and the maintenance dose is 360 mg.
  • a maintenance dose is administered after administration of the loading dose.
  • a flat dose is used as an initial bolus or loading dose and/or maintenance dose.
  • a suitable flat dose is provided in a single injection syringe.
  • a suitable flat dose may be administered (e.g., subcutaneously or
  • a suitable flat dose is about between 10 mg and 800 mg. Accordingly, in some embodiments, a suitable flat dose is equal to or greater than about 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75mg, 80 mg, 85mg, 90 mg, 95 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg, 140 mg, 140 mg, 150 mg, 155 mg, 160 mg, 165 mg, 170 mg, 175 mg, 180 mg, 185 mg, 190 mg, 195 mg, 200 mg, 205 mg, 210 mg, 215 mg, 220 mg, 225 mg, 230 mg, 235 mg, 240 mg, 245 mg, 250 mg, 255 mg, 260 mg, 265 mg, 270 mg, 275 mg, 280 mg, 285 mg, 290 g, 295 mg, 300 mg, 305 mg,
  • a suitable flat dose ranges from 50- 800 mg, 50-700 mg, 50-600 mg, 50-500 mg, 100-800 mg, 100-700 mg, 100-600 mg, 100-500 mg, 100-500 mg, 100- 400 mg, 150-400 mg, 200-400 mg, 250-400 mg, 300-350 mg, 320-400 mg, or 350-400 mg.
  • a loading dose is about 700 mg, 705 mg, 710 mg, 715 mg, 720 mg, 725 mg, 730 mg, 735 mg, 740 mg, 745 mg, 750 g, 755 mg, 760 mg, 765 mg, 770 mg, 775 mg, 780 g, 785 mg, 790 mg, 795 mg, or 800.
  • a suitable initial bolus flat dose is 720 mg.
  • a maintenance dose is about 300 mg, 305 mg, 310 mg, 315 mg, 320 mg, 325 mg, 330 mg, 335 mg, 340 mg, 345 mg, 350 mg, 355 mg, 360 mg, 365 mg, 370 mg, 375 mg, 380 mg, 380 mg, 390 mg, 395 mg, or 400 mg.
  • a suitable maintenance flat dose is 360 mg.
  • the flat dose is 720 mg initial bolus dose, and is 360 mg maintenance dose.
  • an initial loading or bolus dose of about 720 mg is administered.
  • the therapeutically effective dose comprises an initial bolus or loading dose of about 720 mg, followed by at least one maintenance dose of about 360 mg.
  • a weight-based dose is used as an initial bolus or loading dose and/or maintenance dose.
  • the dose is provided in a single injection syringe.
  • the dose may be administered (e.g., subcutaneously or intravenously) in a single injection or by multiple injections.
  • a loading dose is about 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 21 mg/kg, 22 mg/kg, 23 mg/kg, 24 mg/kg, or 25 mg/kg.
  • a loading dose is about between 5 mg/kg and 25 mg/kg and a maintenance dose is about between 2.5 mg/kg and 7.5 mg/kg.
  • the maintenance dose is about 2.0 mg/kg, 2,5 mg/kg, 3.0 mg/kg, 3.5 mg/kg, 4.0 mg/kg, 4.5 mg/kg, 5.0 mg/kg, 5.5 mg/kg, 6.0 mg/kg, 6.0 mg/kg, 6.5 mg/kg, 7.0 mg/kg, or 7.5 mg/kg.
  • an initial loading or bolus dose of about 10 mg/kg is administered.
  • the therapeutically effective dose comprises an initial bolus dose of about 10 mg/kg, followed by at least one maintenance dose of about 5 mg/kg.
  • An administration interval of an anti-OSMR antibody in the treatment of atopic dermatitis can occur at various durations.
  • the administration interval is daily.
  • the administration interval is ever ⁇ other day.
  • the administration interval is multiple times a week.
  • the administration interval is once ever ⁇ 7 week.
  • the administration interval is once every two weeks.
  • the administration interval is once every three weeks in some embodiments, the administration interval is once every four weeks.
  • the administration interval is once every five weeks.
  • a treatment period of atopic dermatitis with an anti-OSMRp antibody can vary in duration.
  • tire treatment period is at least one month.
  • the treatment period is at least 4 weeks, or at least 5 weeks, or at least 6 weeks, or at least 7 weeks, or at least 8 weeks, or at least 9 weeks, or at least 10 weeks, or at least 11 weeks, or at least 12 weeks, or at least 13 weeks, or at least 15 weeks, or at least 18 weeks, or at least 20 weeks, or at least 22 weeks, or at least 24 weeks.
  • the treatment period is at least two months.
  • the treatment period is at least three months.
  • the treatment period is at least six months.
  • the treatment period is at least nine months. In some embodiments, the treatment period is at least one year. In some embodiments, the treatment period is at least two years. In some embodiments, the treatment period continues throughout the subject’s life. In some embodiments, the treatment period continues until an atopic dermatitis flare (e.g., eczema flare) occurs.
  • an atopic dermatitis flare e.g., eczema flare
  • Evaluation of anti-OSMR antibody concentration-time profiles in serum of subjects with atopic dermatitis may be evaluated directly by measuring systemic serum anti- OSMRB antibody concentration-time profiles.
  • anti-OSMRB antibody typically, anti-OSMRB antibody
  • pharmacokinetic and pharmacodynamic profiles are evaluated by sampling the blood of treated subjects periodically.
  • the following standard abbreviations are used to represent the associated pharmacokinetic parameters.
  • OSMRB antibody administration are used in PK analysis.
  • blood samples are typically collected within 15 or 30 minutes prior to anti-OSMRB antibody administration (pre injection baseline or time 0) and at hours 1, 4, 8 or 12, or days 1 (24 hours), 2, 3, 4, 5, 6, 7, 10,
  • ELISA enzyme-linked immunosorbent assay
  • Pharmacokinetic parameters may be evaluated at any stage during the treatment, for example, at day 1 , day 2, day 3, day 4, day 5, day 6, week 1, week 2, week 3, week 4, week 5, week 6, week 7, week 8, week 9, week 10, week 11, week 12, week 13, week 14, week 15, week 16, week 17, week 18, week 19, week 20, week 21, week 22, week 23, week 24, or later.
  • pharmacokinetic parameters may be evaluated at month 1, month 2, month 3, month 4, month 5, month 6, month 7, month 8, month 9, month 10, month 1 1, month 12, month 13, month 14, month 15, month 16, month 17, month 18, month 19, month 20, month 21, month 22, month 23, month 2/4, or later during the treatment.
  • Adverse effects related to the treatment of atopic dermatitis can include peripheral edema, exacerbation of atopic dermatitis, nasopharyngitis, upper respiratory ' tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, and dry eye.
  • administration of an anti-OSMRp antibody results in no serious adverse effects in the subject.
  • administration of an anti-OSMRp antibody does not result in one or more of peripheral edema, exacerbation of atopic dermatitis, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, and dr ' eye.
  • an anti-OSMRp antibody described herein may be used in combination with one or more other therapeutic agents for the treatment of atopic monatitis (AD).
  • an anti-OSMRp antibody may be administered in combination with one or more of concomitant corticosteroids (e.g , TCS), topical calcineurin inhibitors, antimicrobials and/or antiseptics, antihistamines, and others (e.g., coal tar, phosphodiesterase inhibitors) that are administered systemical!y (e.g., orally) or topically in some embodiments, an anti-OSMRp antibody and one or more other therapeutic agents may be administered simultaneously.
  • concomitant corticosteroids e.g , TCS
  • topical calcineurin inhibitors e.g., antimicrobials and/or antiseptics
  • antihistamines e.g., coal tar, phosphodiesterase inhibitors
  • an anti-OSMRp antibody and one or more other therapeutic agents may be administered
  • an anti-OSMRp antibody and one or more other therapeutic agents may be administered sequentially. In some embodiments, one or more other therapeutic agents may be administered as needed. In some embodiments, TCS is administered after a flare occurs in the subject. In some embodiments, TCS is administered 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks after administration of the anti-OSMRp antibody. In some embodiments,
  • TCS is administered to treat a flare as needed.
  • the dose of anti- OSMRp antibody is decreased and TCS is administered.
  • the administration interval of anti- OSMRp antibody is increased (i.e., longer period between doses) and TCS is administered.
  • Uremic pruritus is a debilitating disease with a significant negative impact on patient quality of life. Roughly more than half of patients with end stage renal disease (ESRD) undergoing dialysis suffer from pruritus ( ⁇ ,000,000 lough. 2010). The prevalence of moderate to severe disease has been estimated at 42% based on results from an international dialysis outcomes and practice study (Pisoni et al., 2006). The underlying etiology of UP is unknown, but !L-31 has been implicated.
  • ESRD end stage renal disease
  • an antibody such as the anti-OSMRp antibodies described herein, that antagonizes both IL-31 and OSM, provides a therapeutic opportunity in UP through the inhibition of downstream signaling events stimulated by IL-31 and OSM, two cytokines that drive pruritus, inflammation, and fibrosis in chronic pruritic diseases.
  • Pruritus may be constant or intermittent.
  • the hack is the most commonly affected area, but arms, head, and abdomen are also commonly affected.
  • Excoriations with no primary lesi ons, and sparing of the butterfly area of the back, are typical.
  • one or more symptoms of uremic pruritus are assessed by a Pruritus Numerical Rating Scale (NRS). In some embodiments, one or more symptoms of uremic pruritus are assessed by a Dermatology Life Quality Index (DLQI). In some embodiments, one or more symptoms of uremic pruritus are assessed by a Hospital Anxiety and Depression Scale (HADS). In some embodiments, one or more symptoms of uremic pruritus, such as sleep quality and sleep quantity, are assessed by actigraphy
  • the methods of die invention are used for treating pruritus in a subject having a kidney disease.
  • the methods of the invention are used for treating pruritus in subjects having chronic kidney disease.
  • the methods of the invention are used in predialysis subjects having chronic kidney disease.
  • the composition and the methods of the invention are useful in the treating pruritus a subgroup of subjects having chronic kidney disease, and who have not undergone dialysis.
  • administering of an anti-OSMR]3 antibody occurs prior to, during, or immediately following dialysis.
  • uremic pruritus is treated by administering to a sub ject in need of treatment an anti-QSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of uremic pruritus relative to a control.
  • the terms, "treat” or "treatment,” as used in the context of uremic pruritus herein, refers to amelioration of one or more symptoms associated with uremic pruritus, prevention or delay of the onset of one or more symptoms of uremic pruritus, and/or lessening of the severity or frequency of one or more symptoms of uremic pruritus.
  • the terms, "treat” or “treatment,” as used in the context of uremic pruritus herein, refers to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or snore symptoms or features of uremic pruritus.
  • the administration of an anti-OSMRB antibody results a statistically-significant drop on a quantitative numerical pruritus scale.
  • the step of administering comprises subcutaneous administration.
  • subcutaneous administration is through subcutaneous injection.
  • subcutaneous administration is through a subcutaneous pump.
  • subcutaneous injection of the anti-OSMRB antibody can be performed in the upper arm, the anterior surface of the thigh, the lower portion of the abdomen, the upper back or the upper area of the buttock.
  • the site of injection is rotated.
  • the step of administering comprises intravenous administration. In some embodiments, the step of administering comprises intravenous administration followed by subcutaneous administration. In some embodiments, the step of administering occurs one day before the subject undergoes hemodialysis. In other embodiments, the step of administering occurs during hemodialysis. In other embodiments, the step of administering occurs within one day after hemodialysis.
  • the subject need of treatment has end stage renal disease.
  • the subject in need of treatment is undergoing a hemodialysis regimen of at least one time-per-week.
  • the subject in need of treatment is undergoing a three-times-per-week hemodialysis regimen.
  • the three- times-per-week hemodialysis regimen has been stable for at least three months.
  • the effect of an anti-QSMRB antibody on uremic pruritus is measured relative to a control.
  • a control is indicative of the one or more symptoms of uremic pruritus in the subject before the treatment.
  • one or more symptoms of uremic pruritus in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 5.
  • one or more symptom s of uremic pruritus in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 7.
  • a control is indicative of the one or more symptoms of uremic pruritus in a control subject with the same disease status without treatment.
  • the control is indicative of the one or more symptoms of uremic pruritus in a control subject with the same disease status that was administered a placebo.
  • a subject in need of treatment of an inflammatory or pruritic skin disease or disorder in accordance with the invention has elevated levels of one or more cytokines associated with the OSMR signaling pathway in comparison to a healthy subject. Accordingly, in some embodiments, the subject in need of treatment has elevated levels of one or more of IL-31, OSM, IL-3 IRa, and OSMRj3 in comparison to a healthy subject. In some embodiments, the subject in need of treatment has ele vated le vels of one or more of IL-31 in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated levels of one or more of OSM in comparison to a heal thy subject. In some
  • the subject in need of treatment has elevated levels of one or more of IL-3 IRa in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated levels of one or more of 08MKb in comparison to a healthy subject.
  • treating the subject in need thereof results in a decrease or stabilization of MCP-1/CCL2 levels in the sub j ect. Accordingly, in some embodiments, treating a subject in need thereof results in a decrease of MCP-l levels in comparison to the diseased state. In some embodiments, treating a subject in need thereof results in stabilization of MCP-l levels.
  • stabilization is meant that the levels of MCP-l remain about the same and do not increase or decrease.
  • treating a subject results in reduced MCP-l levels in lymphocytes and/or endothelial cells.
  • the subject in need of treatment has WI-NRS scores of about 4, about 5, about 6, about 7, about 8 or above. Accordingly, in some embodiments, the subject in need of treatment has WI-NRS score of about 4. In some embodiments, the subject in need of treatment has WI-NRS score of about 5. In some embodiments, the subject in need of treatment has WI-NRS score of about 6. In some embodiments, the subject in need of treatment has WI-NRS score of about 7. In some embodiments, the subject in need of treatment has WI- NRS score of about 8. In some embodiments, the subject in need of treatment has WI-NRS score of more than 8.
  • a subject is selected for treatment who has MCP-1/CCL2 levels greater than found in a healthy individual. In some embodiments, the subject selected for treatment does not have elevated levels of MCP-1/CCL2. in comparison to a healthy individual . In some embodiments, IL-3 i expression level is elevated in the subject relative to a control . In some embodiments, IL-31 expression level is not elevated in the subject relative to a control.
  • IL-31 expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the IL-31 expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition.
  • IL-31 Rot expression level is elevated in the subject relative to a control.
  • QSM expression level is elevated in the subject relative to a control.
  • OSMRp expression level is elevated in the subject relative to a control .
  • OSMRp expression level is not elevated in the subject relative to a control.
  • OSMRp expression level in a portion of the subject’s skm affected by a pruritic disease or condition is approximately the same as the OSMRp expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition.
  • a therapeutically effective dose of an anti-OSMRB antibody for treating uremic pruritus or for treating pruritus in predialysis subjects having kidney disease can occur at various dosages.
  • a therapeutically effective dose is equal to or greater than about 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 0.6 mg/kg, 0.8 mg/kg, 0.9 mg/kg, 1 mg/kg, 1.2 mg/kg, 1.5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 8.5 mg/kg, 9 mg/kg, 9.5 mg/kg, 10 mg/kg, 10.5 mg/kg, 11 mg/kg, 1 1.5 mg/kg, 12 mg/kg, 12.5 mg/kg, 13 mg
  • a therapeutically effective dose is equal to or greater than 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg / kg, 19 mg/kg or 20 mg / kg.
  • a therapeutically effective dose is approximately 0.1-20 mg/kg, approximately 0.3-20 mg/kg, approximately 0.5-20 mg/kg, approximately 0.75-20 mg/kg, approximately 1-20 mg/kg, approximately 1.5-20 mg/kg, approximately 2-20 mg/kg, approximately 2.5-20 mg/kg, approximately 3-20 mg/kg, approximately 3.5-20 mg/kg, approximately 4-20 mg/kg, approximately 4.5-20 mg/kg, approximately 5-20 mg/kg, approximately 5.5-20 mg/kg, approximately 6-20 mg/kg, approximately 6.5-20 mg/kg, approximately 7-20 mg/kg, approximately 7.5-20 mg/kg, approximately 8-20 mg/kg, approximately 8.5-20 mg/kg, approximately 9-20 mg/kg, approximately 9.5-20 mg/kg, approximately 10-20 mg/kg, approximately 10.5-20 mg/kg.
  • a therapeutically effective dose is approximately 3-20 mg/kg, approximately 4-20 mg/kg, approximately 5-20 mg/kg, approximately 6-20 mg/kg, approximately 7-20 mg/kg, approximately 8-20 mg/kg, approximately 9-20 mg/kg,
  • a pharmaceutically acceptable amount approximately 3-11 mg/kg, approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mgkg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, or approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • a pharmaceutically acceptable amount approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mgkg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, or approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • therapeutically effective dose is about 5 mg/kg. In some embodiments, a therapeutically effective dose is about 10 mg/kg.
  • the therapeutically effective dose is equal to or greater than 50 mg/kg, 100 mg/kg, 150 mg/kg, 200 mg/kg, or 250 mg/kg, 300 mg/kg, 310 mg/kg, 320 mg/kg, 330 mg/kg, 340 mg/kg, 350 mg/kg, 360 mg/kg, 370 mg/kg, 380 mg/kg, 390 mg/kg, 400 mg/kg, 450 mg/kg, 500 mg/kg, 550 mg/kg, 600 mg/kg, 650 mg/kg, 700 mg/kg, 710 mg/kg, 720 mg/kg, 730 mg/kg, 740 mg/kg, 750 mg/kg, 800 mg/kg, 850 mg/kg, 900 mg/kg, 950 mg/kg, or
  • a therapeutically effective dose is approximately 50-1,000 mg/kg, approximately 100-1,000 mg/kg, approximately 150-1 ,000 mg/kg, approximately 200- 1 ,000 mg/kg, approximately 250-1,000 mg/kg, approximately 300-1,000 mg/kg, approximately 350-1,000 mg/kg, approximately 400-1,000 mg/kg, approximately 450-1,000 mg/kg, approximately 500-1,000 mg/kg, approximately 550-1,000 mg/kg, approximately 600-1,000 mg/kg, approximately 650-1,000 mg/kg, approximately 700-1,000 mg/kg, approximately 750- 1,000 mg/kg, approximately 800-1,000 mg/kg, approximately 850-1,000 mg/kg, approximately 900-1,000 mg/kg, approximately 950-1,000 mg/kg, approximately 50-950 mg/kg, approximately 50-900 mg/kg, approximately 50-850 mg/kg, approximately 50-800 mg/kg, approximately 50- 750 mg/kg, approximately 50-700 mg/kg, approximately 50-650 mg/kg, approximately 50-600 mg/kg, approximately 50-550 mg/kg, approximately 50-
  • administering comprises an initial bolus or loading dose, followed by at least one maintenance dose.
  • the initial bolus or loading dose is greater than the at least one maintenance dose.
  • the initial bolus or loading dose is at least one-fold, two-fold, three-fold, four fold or five-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is two-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is 720 mg and the maintenance dose is 360 mg.
  • a maintenance dose is administered after administration of the loading dose.
  • a flat dose is used as an initial bolus or loading dose and/or maintenance dose.
  • a suitable flat dose is provided in a single injection syringe.
  • a suitable flat dose may be administered (e.g., subcutaneously or
  • a suitable flat dose is about between 10 mg and 800 mg. Accordingly, in some embodiments, a suitable flat dose is equal to or greater than about 10 rng, 20 mg, 30 mg, 40 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75mg, 80 mg, 85mg, 90 mg, 95 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg, 140 mg, 140 mg, 150 mg, 155 mg, 160 mg, 165 mg, 170 mg, 175 mg, 180 mg,
  • a suitable flat dose ranges from 50- 800 mg, 50-700 mg, 50-600 mg, 50-500 mg, 100-800 mg, 100-700 mg, 100-600 mg, 100-500 mg, 100-500 mg, 100- 400 mg, 150-400 mg, 200-400 mg, 250-400 mg, 300-350 mg, 320-400 mg, or 350-400 mg.
  • a loading dose is about 700 mg, 705 mg, 710 mg, 715 mg, 720 mg, 725 mg, 730 mg, 735 mg, 740 mg, 745 mg, 750 mg, 755 mg, 760 mg, 765 mg, 770 mg, 775 mg, 780 mg, 785 mg, 790 mg, 795 mg, or 800.
  • a suitable initial bolus flat dose is 720 mg.
  • a maintenance dose is about 300 mg, 305 mg, 310 mg, 315 mg, 320 mg, 325 mg, 330 g, 335 mg, 340 mg, 345 mg, 350 mg, 355 mg, 360 g, 365 mg, 370 mg, 375 mg, 380 mg, 380 mg, 390 mg, 395 mg, or 400 mg.
  • a suitable maintenance flat dose is 360 mg.
  • the flat dose is 720 mg initial bolus dose, and is 360 mg maintenance dose.
  • an initial loading or bolus dose of about 720 mg is administered.
  • the therapeutically effective dose comprises an initial bolus or loading dose of about 720 mg, followed by at least one maintenance dose of about 360 mg.
  • a weight-based dose is used as an initial bolus or loading dose and/or maintenance dose.
  • the dose is provided in a single injection syringe.
  • the dose may be administered (e.g., subcutaneously or intravenously) in a single injection or by multiple injections.
  • a loading dose is about 4 mg/leg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 21 mg/kg, 22 mg/kg, 23 mg/kg, 24 mg/kg, or 25 mg/kg. In some embodiments, a loading dose is about between 5 mg/kg and 25 mg/kg and a maintenance dose is about between 2.5 mg/kg and 7.5 mg/kg.
  • the maintenance dose is about 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 3.5 mg/kg, 4.0 mg/kg, 4.5 mg/kg, 5.0 mg/kg, 5.5 mg/kg, 6.0 mg/kg, 6.0 mg/kg, 6.5 mg/kg, 7.0 mg/kg, or 7.5 mg/kg.
  • an initial loading or bolus dose of about 10 mg/kg is administered.
  • the therapeutically effective dose compri ses an initial bolus dose of about 10 mg/kg, followed by at least one maintenance dose of about 5 mg/kg.
  • an administration interval of an anti-OSMRJi antibody in the treatment of uremic pruritus or treatment of pruritus in a chronic kidney disease subject can occur at various durations.
  • the administration interval is daily.
  • the administration interval is every other day.
  • the administration interval is multiple times a week.
  • the administration interval is once every week.
  • the administration interval is once every two weeks.
  • the administration interval is once every' three weeks.
  • the administration interval is once every four weeks.
  • the administration interval is once every five weeks.
  • a treatment period of uremic pruritus with an anti-OSMRB anti body can vary in duration.
  • the treatment period is at least one month.
  • the treatment period is at least 4 weeks, or at least 5 weeks, or at least 6 weeks, or at least 7 weeks, or at least 8 weeks, or at least 9 weeks, or at least 10 weeks, or at least 11 weeks or at least 12 weeks, or at least 13 weeks, or at least 15 weeks, or at least 18 weeks, or at least 2.0 weeks, or at least 22 weeks, or at least 24 weeks.
  • the treatment period is at least two months.
  • the treatment period is at least three months.
  • the treatment period is at least six months.
  • the treatment period is at least nine months.
  • the treatment period is at least one year. In some embodiments, the treatment period is at least two years. In some
  • the treatment period is for as long as the subject is on hemodialysis.
  • Evaluation of anti-OSMRB antibody concentration-time profiles in serum of subjects with uremic pruritus may he evaluated directly by measuring systemic serum anti- OSMRJ3 antibody concentration-time profiles.
  • anti-OSMRJ3 antibody typically, anti-OSMRJ3 antibody
  • pharmacokinetic and pharmacodynamic profiles are evaluated by sampling the blood of treated subjects periodically.
  • the following standard abbreviations are used to represent the associated pharmacokinetic parameters .
  • AUCo-t area under the concentration-time curve (AUC) from time zero to the last measurable concentration calculated using the linear trapezoidal rule for increasing concentrations and the logarithmic rule for decreasing concentrations AUCo-cc AUC from time zero io infinity, calculated using the formula:
  • Vd volume of distribution IV doses only
  • OSMRB antibody administration are used m PK analysis.
  • blood samples are typically collected within 15 or 30 minutes prior to anti-OSMRp antibody administration (pre injection baseline or time 0) and at hours 1, 4, 8 or 12, or days 1 (24 and 28 hours), 2, 3, 4, 5, 6,
  • V arious methods may be used to measure anti-OSMRfi antibody concentration serum.
  • enzyme-linked immunosorbent assay (ELISA) methods are used.
  • Pharmacokinetic parameters may be evaluated at any stage during the treatment, for example, at day 1 , day 2, day 3, day 4, day 5, day 6, week 1, week 2, week 3, week 4, week 5, week 6, week 7, week 8, week 9, week 10, week 1 1, week 12, week 13, week 14, week 15, week 16, week 17, week 18, week 19, week 20, week 21, week 22, week 23, week 24, or later.
  • pharmacokinetic parameters may be evaluated at month 1, month 2, month 3, month 4, month 5, month 6, month 7, month 8, month 9, month 10, month 1 1, month 12, month 13, month 14, month 15, month 16, month 17, month 18, month 19, month 20, month 21, month 22, month 23, month 24, or later during the treatment.
  • Adverse effects related to the treatment of uremic pruritus can include peripheral edema, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, and dry eye.
  • administration of an anti-OSMRB antibody results in no serious adverse effects in the subject. In some embodiments, administration of an anti-OSMRB antibody does not result in one or more of peripheral edema, nasopharyngitis, upper respiratory tract infections, and increased creatine phosphokinase.
  • an anti-OSMRp antibody described herein may be used in combination with one or more other therapeutic agents for the treatment of uremic pruritus (UP).
  • an anti-OSMRp antibody may be administered in combination with one or more of concomitant corticosteroids (e.g., TCS), caicmeurin inhibitors, antimicrobials and/or antiseptics, antihistamines, and others (e.g., coal tar, phosphodiesterase inhibitors) that are administered systemiealiy (e.g., orally) or topically.
  • an anti-OSMRp antibody and one or more other therapeutic agents may be administered simultaneously.
  • an anti-OSMRp antibody and one or more other therapeutic agents may be administered sequentially.
  • one or more other therapeutic agents may be administered as needed.
  • an anti-OSMRp antibody described herein is used in treating prurigo nodularis (PN).
  • the methods of the invention are used for treating pruritus in a subject having PN.
  • PN also known as nodular prurigo is a skin disease
  • nodules characterized by itchy nodules.
  • the nodules usually appear in the arms and legs. Patients often present with multiple excoriating lesions caused by scratching in some embodim nts, the subject presents with pruritic hyperkeratotic nodules.
  • the prurigo nodularis is idiopathic. In some embodiments, the prurigo nodularis is not associated with any other underlying co-morbidities. In some embodiments, the prurigo nodularis is associated with one or more underlying co-morbidities.
  • PN can be a distinct, highly pruritic chronic skin disease that is not defined by its comorbid conditions. 1L-31 could be implicated in the pathogenesis of PN. In some embodiments, the IL-31 pathway could he an attractive target for pharmacological intervention in PN. In some embodiments, IL-31 expression level is elevated in the subject relative to a control.
  • IL-3 IRa expression level is elevated in the subject relative to a control.
  • OSM expression level is elevated in the subject relative to a control.
  • OSMR expression level is elevated in the subject relativ e to a control .
  • the lev els of any one of IL-31, IL-3 1 Rot, OSM and OSMR in the subject is determined via skin biopsy from hyperkeratotic nodules.
  • the control is a healthy subject, who is not diagnosed with a pruritic disease.
  • the method of treating prurigo nodularis comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration in some embodiments, the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • NRS Pruritus Numerical Rating Scale
  • WI-NRS Worst Itch-Numeric Rating Scale
  • one or more symptoms of prurigo nodularis are assessed by the proportion of subjects achieving at least a 4-point reduction from baseline in weekly average WI-NRS. In some embodiments, one or more symptoms of prurigo nodularis are assessed by a change or percent change from baseline m pruritus Visual Analog Scale (VAS) In some embodiments, one or more symptoms of prurigo nodularis are assessed by change from baseline in 3-D Pruritus total score. In some embodiments, one or more symptoms of prurigo nodularis are assessed by change from baseline in Sleep Loss VAS. In some embodiments, one or more symptoms of prurigo nodularis are assessed by change from baseline in weekly average of difficulty failing asleep NRS.
  • VAS Visual Analog Scale
  • one or more symptoms of prurigo nodularis are assessed by change from baseline in 3-D Pruritus total score. In some embodiments, one or more symptoms of prurigo nodularis are assessed by change from baseline in Sleep Loss VAS. In some embodiments, one or more
  • one or more symptoms of prurigo nodularis are assessed by change from baseline in weekly average of sleep quality MRS. In some embodiments, one or more symptoms of prurigo nodularis are assessed by change from baseline in quality of life measures over time. In some embodiments, one or more symptoms of prurigo nodularis are assessed by change from baseline in Prurigo Nodularis Investigor Global Assessment (PN-1GA). In some embodiments, one or more sy mptoms of prurigo nodularis are assessed by change from baseline in Prurigo Nodularis Nodule Assessment Tool (PN-NAT). In some embodiments, one or more symptoms of prurigo nodularis are assessed by a Dermatology Life Quality Index (DLQI).
  • DLQI Dermatology Life Quality Index
  • one or more symptoms of prurigo nodularis are assessed by a Hospital Anxiety and Depression Scale (HADS).
  • HADS Hospital Anxiety and Depression Scale
  • one or more symptom s of prurigo nodularis, such as sleep quality and sleep quantity, are assessed by actigraphy.
  • prurigo nodularis is treated by administering to a subject m need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of prurigo nodularis relative to a control .
  • the tenns, "treat” or “treatment,” as used in the context of prurigo nodularis herein, refers to partially or completely alleviate, ameliorate, relie ve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of prurigo nodularis.
  • the tenns, "treat” or “treatment,” as used in the context of prurigo nodularis herein refers to partially or completely alleviate, ameliorate, relie ve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of prurigo nodularis.
  • the tenns, "treat” or “treatment,” as used in the context of prurigo nodularis herein refers to partially or completely alleviate, ameliorate, relie ve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of prurigo nodularis.
  • administering results in a statistically-significant drop on a quantitati ve numerical pruritus scale.
  • the administration of an anti- OSMRB antibody results in a statistically-significant drop in weekly average Worst Itch- Numerical Rating Scale (WI-NRS).
  • WI-NRS Worst Itch- Numerical Rating Scale
  • the weekly average WI-NRS score has at least a 4-point reduction from baseline.
  • the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change from baseline in pruritus Visual Analog Scale (VAS).
  • VAS pruritus Visual Analog Scale
  • the administration of an anti- OSMRB antibody results in a statistically-significant drop or percent change from baseline in 5- D Pruritus total score.
  • the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change from baseline in Sleep Loss VAS. In some embodiments, the administration of an anti-QSMRB antibody results in a statistically- significant drop or percent change in weekly average of difficulty falling asleep NRS. In some embodiments, the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change in weekly average sleep quality NRS. In some embodiments, the administration of an anti-QSMRB antibody results in a stati stically-significant drop or percent change from baseline in Prurigo Nodularis Investigator Global Assessment (PN-IGA).
  • PN-IGA Prurigo Nodularis Investigator Global Assessment
  • the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change from baseline in Prurigo Nodularis Nodule Assessment Tool (PN-NAT).
  • the administrati on of an anti-OSMRB antibody' results in a statistically- significant decrease or percent change from baseline in Dermatology Life Quality Index (DLQI).
  • the administration of an anti-OSMRB antibody results in a statistically- significant decrease or percent change from baseline in Hospital Anxiety' and Depression Scale (HADS).
  • the administration of an anti-OSMRB antibody results in a statistically-improved or percent change from baseline in actigraphy scores.
  • the administration of an anti-OSMRB antibody results m a statistically-significant increase or percent change in quality of life measures over time. In some embodiments, the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change in UAS7 score. In some embodiments, the step of administering comprises subcutaneous administration. In some embodiments, subcutaneous administration is through subcutaneous injection. In some embodiments, subcutaneous administration is through a subcutaneous pump.
  • subcutaneous injection of the anti-QSMRB antibody can be performed in the upper arm, the anterior surface of the thigh, the lower portion of the abdomen, the upper back or the upper area of the buttock.
  • the site of injection is rotated.
  • the step of adm inistering comprises intravenous administration. In some embodiments, the step of administering comprises intravenous administration followed by subcutaneous administration. In some embodiments, the step of administering occurs one day before the subject undergoes hemodialysis. In other embodiments, the step of administering occurs during hemodialysis. In other embodiments, the step of administering occurs within one day after hemodialysis.
  • the subject in need of treatment has end stage renal disease. In some embodiments, the subject in need of treatment is undergoing a hemodialysis regimen of at least one time-per-week. In some embodiments, the subject in need of treatment is undergoing a three-times-per-week hemodialysis regimen. In some embodiments, the three- times-per-week hemodialysis regimen has been stable for at least three months.
  • the effect of an anti-OSMRB antibody on prurigo nodularis is measured relative to a control.
  • a control is indicative of the one or more symptoms of prurigo nodularis in the subject before the treatment.
  • one or more symptoms of prurigo nodularis in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 5.
  • one or more symptoms of prurigo nodularis in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 7.
  • a control is indicative of tire one or more symptoms of prurigo nodularis in a control subject with the same disease status without treatment.
  • the control is indicative of the one or more symptoms of prurigo nodularis in a control subject with the same disease status that was administered a placebo.
  • a subject in need of treatment has elevated levels of one or more cytokines associated with die OSMR[3 signaling pathway in comparison to a healthy subject. Accordingly, in some embodiments, the subject in need of treatment has elevated levels of one or more of IL-31, OSM, IL-3 IRa, and OSMRJ3 in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated levels of one or more of IL-31 in comparison to a healthy subject. In some embodiments, the subject in need of treatment has elevated levels of one or more of OSM in comparison to a healthy subject. In some
  • the subject in need of treatment has elevated levels of one or more of IL-3 IRa in comparison to a healthy subject. In some embodiments, the subject in need of treatment has ele vated le vels of one or more of QSMRfi in comparison to a healthy subject.
  • treating the subject in need thereof results in a decrease or stabilization of MCP-1/CCL2 levels in the subject. Accordingly, in some embodiments, treating a subject in need diereof results in a decrease of MCP-1 levels in comparison to die diseased state. In some embodiments, treating a subject in need thereof results in stabilization of MCP-1 levels. By“stabilization” is meant that the levels of MCP-1 remain about the same and do not increase or decrease. In some embodiments, treating a subject results in reduced MCP-1 levels in lymphocytes and/or endothelial cells. [0217] In some embodiments, the subject in need of treatment has Wl-NRS scores of about 4, about 5, about 6, about 7, about 8 or above.
  • the subject in need of treatment has WI-NRS score of about 4. in some embodiments, the subject in need of treatment has WI-NRS score of about 5. In some embodiments, the subject in need of treatment has WI-NRS score of about 6. In some embodiments, the subject in need of treatment has WI-NRS score of about 7. In some embodiments, the subject in need of treatment has WI- NRS score of about 8. In some embodiments, the subject in need of treatment has WI-NRS score of more than 8.
  • a subject is selected for treatment who has MCP-1/CCL2 levels greater than found in a healthy individual. In some embodiments, the subject selected for treatment does not have elevated levels of MCP-1/CCL2 in comparison to a healthy individual.
  • IL-31 expression level is elevated the subject relative to a control. In some embodiments, IL-31 expression level is not elevated in the subject relative to a control in some embodiments, IL-31 expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the IL-31 expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition. In some embodiments, IL-3 IRa expression level is elevated in the subject relative to a control. In some embodiments, OSM expression level is elevated in the subject relative to a control.
  • OSMRJ3 expression level is elevated in the subject relative to a control. In some embodiments, OSMRp expression level is not elevated in the subject relative to a control. In some embodiments, OSMRJ3 expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the OSMR expression level in (i) a portion of the subject’s sk that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition.
  • a therapeutically effective dose of an anti-OSMRB antibody for treating prurigo nodularis can occur at various dosages.
  • a therape utically effective dose is equal to or greater than about 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 0.6 mg/kg, 0.8 mg/kg, 0.9 mg/kg, 1 mg/kg, i .2 mg/kg, 1 .5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 8.5 mg/kg, 9 mg/kg, 9.5 mg/kg, 10 mg/kg, 10.5 mg/kg, 11 mg/kg, 11.5 mg/kg, 12 mg/kg, 12.5 mg/kg, 13 mg/kg, 1
  • a therapeutically effective dose is equal to or greater than 3 mg/kg, 4 mg/ g, 5 mg/kg, 6 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 nrg/kg, 18 mg/kg, 19 mg/kg or 20 mg/kg.
  • a therapeutically effective dose is approximately 0.1-20 mg/kg, approximately 0.3-20 mg/kg, approximately 0.5-20 mg/kg, approximately 0.75-20 mg/kg, approximately 1-20 nrg/kg, approximately 1.5-20 mg/kg, approximately 2-20 mg/kg, approximately 2.5-20 mg/kg, approximately 3-20 mg/kg, approximately 3.5-20 mg/kg, approximately 4-20 mg/kg, approximately 4.5-20 mg/kg, approximately 5-20 mg/kg, approximately 5.5-20 mg/kg, approximately 6-20 mg/kg, approximately 6.5-20 mg/kg, approximately 7-20 tng/kg, approximately 7.5-20 mg/kg, approximately 8-20 mg/kg, approximately 8 5-20 mg/kg, approximately 9-20 mg/kg, approximately 9.5-20 mg/kg, approximately 10-20 mg/kg, approximately 10.5-20 mg/kg.
  • a therapeutically effective dose is approximately 3-20 mg/kg, approximately 4-20 mg/kg, approximately 5-20 mg/kg, approximately 6-20 mg/kg, approximately 7-20 mg/kg, approximately 8-20 mg/kg, approximately 9-20 mg/kg,
  • a pharmaceutically acceptable amount approximately 3-11 mg/kg, approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mg/kg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, or approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • a pharmaceutically acceptable amount approximately 3-10 mg/kg, approximately 3-9 mg/kg, approximately 3-8 mg/kg, approximately 3-7 mg/kg, approximately 3-6 mg/kg, approximately 3-5 mg/kg, or approximately 3-4 mg/kg, or approximately 5-10 mg/kg.
  • therapeutically effective dose is about 5 mg/kg. In some embodiments, a therapeutically effective dose is about 10 mg/kg.
  • the therapeutically effective dose is equal to or greater than 50 mg/kg, 100 mg/kg, 150 mg/kg, 200 mg/kg, or 250 mgkg, 300 mg/kg, 310 mg/kg, 320 mg/kg, 330 mg/kg, 340 mg/kg, 350 mg/kg, 360 mg/kg, 370 mg/kg, 380 tng/kg, 390 mg/kg, 400 mg/kg, 450 mg/kg, 500 mg/kg, 550 mg/kg, 600 mg/kg, 650 rrig/kg, 700 mg/kg, 710 mg/kg, 720 mg/kg, 730 mg/kg, 740 mg/kg, 750 mg/kg, 800 mg/kg, 850 mg/kg, 900 mg/kg, 950 mg/kg, or 1000 mg/kg.
  • a therapeutically effective dose is approximately 50-1,000 mg/kg, approximately 100-1,000 mg/kg, approximately 150-1,000 mg/kg, approximately 200- 1 ,000 mg/kg, approximately 250-1,000 mg/kg, approximately 300-1,000 mg/kg, approximately 350-1,000 mg/kg, approximately 400-1,000 mg/kg, approximately 450-1,000 mg/kg, approximately 500-1,000 mg/kg, approximately 550-1,000 mg/kg, approximately 600-1,000 mg/kg, approximately 650-1,000 mg/kg, approximately 700-1,000 mg/kg, approximately 750- 1,000 mg/kg, approximately 800-1,000 mg/kg, approximately 850-1 ,000 mg/kg, approximately 900-1,000 mg/kg, approximately 950-1,000 mg/kg, approximately 50-950 mg/kg, approximately 50-900 mg/kg, approximately 50-850 mg/kg, approximately 50-800 mg/kg, approximately 50- 750 mg/kg, approximately 50-700 mg/kg, approximately 50-650 mg/kg, approximately 50-600 mg/kg, approximately 50-550 mg/kg, approximately 50-
  • administering comprises an initial bolus or loading dose, followed by at least one maintenance dose.
  • the initial bolus or loading dose is greater than the at least one maintenance dose.
  • the initial bolus or loading dose is at least one-fold, two-fold, three-fold, four fold or five-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is two-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is 720 mg and the maintenance dose is 360 mg.
  • a maintenance dose is administered after administration of the loading dose.
  • a fiat dose is used as an initial bolus or loading dose and/or maintenance dose.
  • a suitable fiat dose is provided in a single injection syringe.
  • a suitable flat dose may he administered (e.g., subcutaneously or
  • a suitable flat dose is about between 10 mg and 800 mg.
  • a suitable fiat dose is equal to or greater than about 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75mg, 80 mg, 85mg, 90 mg, 95 mg, 100 mg, 105 mg, 1 10 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg, 140 mg, 140 mg, 150 mg, 155 mg, 160 mg, 165 mg, 170 mg, 175 mg, 180 mg,
  • a suitable flat dose ranges from 50- 800 mg, 50-700 mg, 50-600 mg, 50-500 mg, 100-800 mg, 100-700 mg, 100-600 mg, 100-500 mg, 100-500 mg, 100- 400 mg, 150-400 mg, 200-400 mg, 250-400 mg, 300-350 mg, 320-400 mg, or 350-400 mg.
  • a loading dose is about 700 mg, 705 mg, 710 mg, 715 mg, 720 mg, 725 mg, 730 mg, 735 mg, 740 mg, 745 mg, 750 mg, 755 mg, 760 mg, 765 mg, 770 mg, 775 mg, 780 mg, 785 mg, 790 mg, 795 mg, or 800.
  • a suitable initial bolus flat dose is 720 mg.
  • a maintenance dose is about 300 mg, 305 mg, 310 mg, 315 mg, 320 mg, 325 mg, 330 mg, 335 mg, 340 mg, 345 mg, 350 mg, 355 mg, 360 mg, 365 mg, 370 g, 375 mg, 380 mg, 380 mg, 390 mg, 395 mg, or 400 mg.
  • a suitable maintenance flat dose is 360 mg.
  • the flat dose is 720 mg initial bolus dose, and is 360 mg maintenance dose.
  • an initial loading or bolus dose of about 720 mg is administered.
  • the therapeutically effective dose comprises an initial bolus or loading dose of about 720 mg, followed by at least one maintenance dose of about 360 mg.
  • a weight-based dose is used as an initial bolus or loading dose and/or maintenance dose.
  • the dose is provided in a single injection syringe.
  • the dose may be administered (e.g., subcutaneously or intravenously) in a single injection or by multiple injections.
  • a loading dose is about 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 tng/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 21 mg/kg, 22 mg/kg, 23 mg kg, 24 mg/kg, or 25 mg/kg.
  • a loading dose is about between 5 g/kg and 25 mg/kg and a maintenance dose is about between 2.5 mg/kg and 7.5 mg/kg.
  • the maintenance dose is about 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 3.5 mg/kg, 4.0 mg/kg, 4.5 mg/kg, 5.0 mg/kg, 5.5 mg/kg, 6.0 mg/kg, 6.0 mg/kg, 6.5 mg/kg, 7.0 mg/kg, or 7.5 mg/kg.
  • an initial loading or bolus dose of about 10 mg/kg is administered.
  • the therapeutically effecti ve dose comprises an initial bolus dose of about 10 mg/kg, followed by at least one maintenance dose of about 5 mg/kg.
  • an administration interval of an anti-QSMRU antibody in the treatment of prurigo nodularis in a subject can occur at various durations.
  • the administration interval is daily.
  • the administration interval is every other day.
  • the administration interval is multiple times a week.
  • the administration interval is once every week.
  • the administration interval is once ever two weeks.
  • the administration interval is once every three weeks.
  • the administration interval is once every four weeks.
  • the administration interval is once every five weeks.
  • a treatment period of prurigo nodularis with an anti-OSMRB antibody can vary in duration.
  • the treatment period is at least one month.
  • the treatment period is at least 4 weeks, or at least 5 weeks, or at least 6 weeks, or at least 7 weeks, or at least 8 weeks, or at least 9 weeks, or at least 10 weeks, or at least 11 weeks or at least 12 weeks, or at least 13 weeks, or at least 15 weeks, or at least 18 weeks, or at least 20 weeks, or at least 22 weeks, or at least 24 weeks.
  • the treatment period is at least two months. In some embodiments, the treatment period is at least three months. In some embodiments, the treatment period is at least six months. In some embodiments, the treatment period is at least nine months. In some embodiments, the treatment period is at least one year. In some embodiments, the treatment period is at least two years. In some
  • the treatment period is for as long as the subject is on hemodialysis.
  • Evaluation of anti-QSMRU antibody concentration -time profiles in serum of subjects with prurigo nodularis may be evaluated directly by measuring systemic serum anti- OSMRp antibody concentration-time profiles.
  • anti-QSMR[3 antibody pharmacokinetic and pharmacodynamic profiles are evaluated by sampling the blood of treated subjects periodically. The following standard abbreviations are used to represent the associated pharmacokinetic parameters .
  • OSMRB antibody administration are used in PK analysis.
  • blood samples are typically collected within 15 or 30 minutes prior to anti-OSMRp antibody administration (pre injection baseline or time 0) and at hours 1, 4, 8 or 12, or days 1 (24 and 28 hours), 2, 3, 4, 5, 6, 7, 10, 13, 17, 20, 24, 27, 31, 34, 41, 48, 55, 62, 69, 76, 90, following administration.
  • ELISA enzyme-linked immunosorbent assay
  • Pharmacokinetic parameters may be evaluated at any stage during the treatment, for example, at day 1, day 2, day 3, day 4, day 5, day 6, week 1 , week 2, week 3, week 4, week 5, week 6, week 7, week 8, week 9, week 10, week 1 i, week 12, week 13, week 14, week 15, week 16, week 17, week 18, week 19, week 20, week 21, week 22, week 23, week 24, or later.
  • pharmacokinetic parameters may be evaluated at month 1, month 2, month 3, month 4, month 5, month 6, month 7, month 8, month 9, month 10, month 11, month 12, month 13, month 14, month 15, month 16, month 17, month 18, month 19, month 20, month 21, month 22, month 23, month 24, or later during the treatment.
  • Adverse effects related to the treatment of prurigo nodularis can include peripheral edema, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, and dry eye.
  • administration of an anti -OSMRB antibody results in no serious adverse effects in the subject. In some embodiments, administration of an anti-OSMRB antibody does not result in one or more of peripheral edema, nasopharyngitis, upper respiratory tract infections, and increased creatine phosphokinase.
  • an anti-OSMR[3 antibody described herein may be used in combination with one or more other therapeutic agents for the treatment of prurigo nodularis (PN.)
  • an anti-OSMRJ3 antibody may be administered in combination with one or more of concomitant corticosteroids (e.g., TCS), calcineurin inhibitors, antimicrobials and/or antiseptics, antihistamines, and others (e.g., coal tar, phosphodiesterase inhibitors) that are administered systemicaliy (e.g., orally) or topically.
  • an anti-OSMR antibody and one or more other therapeutic agents may be administered simultaneously.
  • an anti-OSMRp antibody and one or more other therapeutic agents may be administered sequentially.
  • one or more other therapeutic agents may be administered as needed. Additional Therapeutic indications
  • the present invention provides methods and compositions for use in treating pruritus associated with Chronic Idiopathic Pruritus (CIP).
  • CIP Chronic Idiopathic Pruritus
  • the method and compositions of the invention are contemplated for use in the treatment of pruritus associated with Chronic Idiopathic Urticaria (CIU).
  • the method and compositions of the invention are contemplated for use in the treatment of pruritus associated with Chronic Spontaneous Urticaria (( SI )
  • the method and compositions of the invention are contemplated for use in the treatmen t of pruritus associated with Cutaneous Amyloidosis (CA).
  • the method and compositions of the invention are contemplated for use in the treatment of pruritus associated with Plaque Psoriasis (PPs). In some embodiments, the method and compositions of the invention are contemplated for use in the treatment of pruritus associated with Lichen Simplex Chromcus (LSC). In some embodiments, the method and compositions of the invention are contemplated for use in the treatment of pruritus associ ated with Lichen Planus (LP). In som e embodiments, the method and compositions of the invention are contemplated for use in the treatment of pruritus associated with Inflammatory Ichthyosis (II).
  • PPs Plaque Psoriasis
  • LSC Lichen Simplex Chromcus
  • LP Lichen Planus
  • II Inflammatory Ichthyosis
  • the method and compositions of the invention are contemplated for use in tire treatment of pruritus associated with Mastocytosis (MA). In some embodiments, the method and compositions of the invention are contemplated for use in the treatment of pruritus associated with Bullous
  • the method of treating CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • one or more symptoms of any of these pruritic conditions are assessed by a change or percent change from baseline m weekly average of Worst Itch-Numeric Rating Scale (WI-NRS).
  • one or more symptom s of any of these pruritic conditions are assessed by the proportion of subjects achieving at least a 4-point reduction from baseline in weekly average WI-NRS. In some embodiments, one or more symptoms of any of these pruritic conditions are assessed by a change or percent change from baseline in pruritus Visual Analog Scale (VAS). In some embodiments, one or more symptoms of any of these pruritic conditions are assessed by change from baseline in 5-D Pruritus total score. In some embodiments, one or more symptoms of any of these pruritic conditions are assessed by change from baseline in Sleep Loss VAS. In some embodiments, one or more symptoms of any of these pruritic conditions are assessed by change from baseline in weekly average of difficulty falling asleep NRS.
  • VAS pruritus Visual Analog Scale
  • one or more symptoms of any of these pruritic conditions are assessed by change from baseline in weekly average of sleep quality NRS. In some embodiments, one or more symptoms of any of these pruritic conditions are assessed by change from baseline in quality' of life measures over time. In some embodiments, one or more symptoms of CIU or CSU are assessed by a change from baseline in weekly itch severity score, a component of Urticaria Activity Score 7 (UAS7). In some embodiments, one or more symptoms of CIU or CSU are assessed by a change from baseline in weekly hive severity score, a component of UAS7. In some embodiments, one or more symptoms of CIU or CSU are assessed by a change from baseline in UAS7.
  • a control is indicative of the one or more symptoms of these pruritic conditions in the subject before the treatment, including, for example, a score on a pruritus NRS greater than or equal to 5.
  • one or more symptoms of these pruritic conditions in a subject before treatment comprises a score on a pruritus NRS greater than or equal to 7.
  • a control is indicative of the one or more symptoms of these pruritic conditions in a control subject with the same disease status without treatment.
  • the control is indicative of the one or more symptoms of these pruritic conditions in a control subject with the same disease status that was administered a placebo.
  • CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP is treated by administering to a subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP relative to a control.
  • treat refers to amelioration of one or more symptoms associated with CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP, prevention or delay of the onset of one or more symptoms of CIP, CIU, CSU, CA, PPs, LSC, LP, NLA or BP, and/or lessening of tire severity or frequency of one or more symptoms of CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP.
  • the terms, "treat” or “treatment,” as used in the context of CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP herein, refers to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP.
  • the administration of an anti-OSMRB antibody results in a statistically-significant drop on a quantitative numerical pruritus scale.
  • the administration of an anti-OSMRB antibody results in a statistically-significant drop in weekly average Worst Itch- Numerical Rating Scale (WI-NRS). In some embodiments, the weekly average WI-NRS score has at least a 4-point reduction from baseline. In some embodiments, the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change from baseline in pruritus Visual Analog Scale (VAS). In some embodiments, the administration of an anti- OSMRB antibody results in a statistically-significant drop or percent change from baseline in 5- D Pruritus total score. In some embodiments, the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change from baseline in Sleep Loss VAS.
  • WI-NRS Worst Itch- Numerical Rating Scale
  • VAS pruritus Visual Analog Scale
  • the administration of an anti- OSMRB antibody results in a statistically-significant drop or percent change from baseline in 5- D Pruritus total score. In some embodiments,
  • the administration of an anti-OSMRB antibody results in a statistically- significant drop or percent change in weekly average of difficulty falling asleep NRS. In some embodiments, the administration of an anti-OSMRB antibody results in a statistically-significant increase or percent change in quality of life measures over time. In some embodiments, the administration of an anti-OSMRB antibody results in a statistically-significant drop or percent change in UAS7 score.
  • the step of administering comprises subcutaneous administration.
  • subcutaneous administration is through subcutaneous injection.
  • subcutaneous administration is through a subcutaneous pump.
  • a therapeutically effective dose is equal to or greater than about 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 0.6 mg/kg, 0.8 mg/kg, 0.9 mg/kg, 1 mg/kg, 1.2 mg/kg, 1.5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 8.5 mg/kg, 9 mg/kg, 9.5 mg/kg, 10 mg/kg, 10.5 mg/kg, 1 1 mg/kg, 11 5 mg/kg, 12 mg/kg, 12.5 mg/kg, 13 mg/kg, 13 5 mg/kg, 14 mg/kg, 14.5 mg//kg,
  • a therapeutically effective dose is equal to or greater than 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 7.5 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 tng/kg, 15 mg/kg, 16 mg/kg, 17 tng/kg, 18 mg/kg, 19 mg/kg or 20 mg/kg.
  • a therapeutically effective dose is approximately 0.1-20 mg/kg, approximately 0.3-20 mg/kg, approximately 0.5-20 mg/kg, approximately 0.75-20 mg/kg, approximately 1 -20 mg/kg, approximately 1.5-20 mg/kg, approximately 2-20 mg/kg, approximately 2.5-20 mg/kg, approximately 3-20 mg/kg, approximately 3.5-20 mgkg, approximately 4-20 mg/kg, approximately 4.5-20 mg/kg, approximately 5-20 mg/kg, approximately 5.5-20 mg/kg, approximately 6-20 mg/kg, approximately 6.5-20 mg/kg, approximately 7-20 mg/kg, approximately 7.5-20 mg/kg, approximately 8-20 mg/kg, approximately 8.5-20 mg/kg, approximately 9-20 mg/kg, approximately 9.5-20 mg/kg, approximately 10-20 mg/kg, approximately 10.5-20 mg/kg.
  • a therapeutically effective dose is approximately 3-20 mg/kg, approximately 4-20 mg/kg, approximately 5-20 mg/kg, approximately 6-2.0 mg/kg, approximately 7-20 mg/kg, approximately 8-20 mg/kg, approximately 9-20 mg/kg,
  • therapeutically effective dose is about 5 mg/kg. In some embodiments, a therapeutically effective dose is about 10 mg/kg.
  • the therapeutically effective dose is equal to or greater than 50 mg/kg, 100 mg/kg, 150 mg/kg, 200 mg/kg, or 250 mg/kg, 300 mg/kg, 310 mg/kg, 320 mg/kg, 330 mg/kg, 340 mg/kg, 350 mg/kg, 360 mg/kg, 370 mg/kg, 380 mg/kg, 390 mg/kg, 400 mg/kg, 450 mg kg, 500 mg/kg, 550 mg/kg, 600 mg/kg, 650 mg/kg, 700 mg/kg, 710 mg/kg, 720 mg/kg, 730 mg/kg, 740 mg/kg, 750 mg/kg, 800 mg/kg, 850 rrig/kg, 900 mg/kg, 950 mg/kg, or 1000 mg/kg.
  • a therapeutically effective dose is approximately 50-1 ,000 mg/kg, approximately 100-1,000 mg/kg, approximately 150-1,000 mg/kg, approximately 200- 1 ,000 mg/kg, approximately 250-1,000 mg/kg, approximately 300-1,000 mg/kg, approximately 350-1,000 mg/kg, approximately 400-1,000 mg/kg, approximately 450-1 ,000 mg/kg, approximately 500-1,000 mg/kg, approximately 550-1,000 mg/kg, approximately 600-1,000 mg/kg, approximately 650-1,000 mg/kg, approximately 700-1,000 mg/kg, approximately 750- 1,000 mg/kg, approximately 800-1,000 mg/kg, approximately 850-1 ,000 mg/kg, approximately 900-1 ,000 mg/kg, approximately 950-1,000 mg/kg, approximately 50-950 mg/kg, approximately 50-900 mg/kg, approximately 50-850 mg/kg, approximately 50-800 mg/kg, approximately 50- 750 mg/kg, approximately 50-700 mg/kg, approximately 50-650 mg/kg, approximately 50-600 mg/kg, approximately 50-550 mg
  • administering comprises an initial bolus or loading dose, followed by at least one maintenance dose.
  • the initial bolus or loading dose is greater than the at least one maintenance dose.
  • the initial bolus or loading dose is at least one-fold, two-fold, three-fold, four fold or five-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is two-fold greater in dosage than the dosage of the at least one maintenance dose.
  • the initial bolus or loading dose is 720 mg and the maintenance dose is 360 mg.
  • a maintenance dose is administered after administration of the loading dose.
  • a flat dose is used as an initial bolus or loading dose and/or maintenance dose.
  • a suitable flat dose is provided in a single injection syringe.
  • a suitable flat dose may be administered (e.g., subcutaneously or
  • a suitable flat dose is about between 10 mg and 800 mg. Accordingly, in some embodiments, a suitable flat dose is equal to or greater than about 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75mg, 80 mg, 85mg, 90 mg, 95 mg, 100 mg, 105 mg, 1 10 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg, 140 mg, 140 mg, 150 mg, 155 mg, 160 mg, 165 mg, 170 mg, 175 mg, 180 mg, 185 mg, 190 mg, 195 mg, 200 mg, 205 mg, 210 mg, 215 mg, 220 mg, 225 mg, 230 mg, 235 mg, 240 mg, 245 mg, 250 mg, 255 mg, 260 mg, 265 mg, 270 mg, 275 mg, 280 mg, 285 mg, 290 mg, 295 mg, 300 mg, 305 mg,
  • a suitable flat dose ranges from 50- 800 mg, 50-700 mg, 50-600 mg, 50-500 mg, 100-800 mg, 100-700 mg, 100-600 mg, 100-500 mg, 100-500 mg, 100- 400 mg, 150-400 mg, 200-400 mg, 250-400 mg, 300-350 mg, 320-400 mg, or 350-400 mg.
  • a loading dose is about 700 mg, 705 mg, 710 mg, 715 mg, 720 mg, 725 mg, 730 mg, 735 mg, 740 g, 745 mg, 750 mg, 755 mg, 760 mg, 765 g, 770 mg, 775 mg, 780 mg, 785 mg, 790 mg, 795 mg, or 800.
  • a suitable initial bolus flat dose is 720 mg.
  • a maintenance dose is about 300 mg, 305 mg, 310 mg, 315 mg, 320 mg, 325 mg, 330 mg, 335 mg, 340 mg, 345 mg, 350 mg, 355 mg, 360 mg, 365 mg, 370 mg, 375 mg, 380 mg, 380 mg, 390 mg, 395 mg, or 400 mg.
  • a suitable maintenance flat dose is 360 mg.
  • the flat dose is 720 mg initial bolus dose, and is 360 mg maintenance dose.
  • an initial loading or bolus dose of about 720 mg is administered.
  • the therapeutically effective dose comprises an initial bolus or loading dose of about 720 mg, followed by at least one maintenance dose of about 360 mg.
  • a weight-based dose is used as an initial bolus or loading dose and/or maintenance dose.
  • the dose is provided in a single injection syringe.
  • the dose may be administered (e.g., subcutaneously or intravenously) in a single injection or by multiple injections.
  • a loading dose is about 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, I I mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 21 mg/kg, 22 mg/kg, 23 mg/kg, 24 mg/kg, or 25 mg/kg. In some embodiments, a loading dose is about between 5 mg/kg and 25 mg/kg and a maintenance dose is about between 2.5 mg/kg and 7.5 mg/kg.
  • the maintenance dose is about 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 3.5 mg/kg, 4.0 mg/kg, 4.5 mg/kg, 5.0 mg/kg, 5.5 mg/kg, 6.0 mg/kg, 6.0 mg/kg, 6.5 mg/kg, 7.0 mg/kg, or 7.5 mg/kg.
  • an initial loading or bolus dose of about 10 mg/kg is administered.
  • the therapeutically effective dose comprises an initial bolus dose of about 10 mg/kg, followed by at least one maintenance dose of about 5 mg/kg.
  • an administration interval of an anti-OSMRB antibody in the treatment of CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP in a subject can occur at various durations.
  • the administration interval is daily. In some embodiments, the administration interval is every other day. In some embodiments, the administration interval is multiple times a week. In some embodiments, the administration interval is once even week. In some embodiments, the administration interval is once even ' two weeks. In some embodiments, the administration interval is once every three weeks. In some embodiments, the administration interval is once every four weeks. In some embodiments, the administration interval is once every five weeks.
  • the treatment period is at least one month. In some embodiments the treatment period is at least 4 weeks, or at least 5 weeks, or at least 6 weeks, or at least 7 weeks, or at least 8 weeks, or at least 9 weeks, or at least 10 weeks, or at least 11 weeks or at least 12 weeks, or at least 13 weeks, or at least 15 weeks, or at least 18 weeks, or at least 20 weeks, or at least 22 weeks, or at least 24 weeks. In some embodiments, the treatment period is at least two months. In some embodiments, the treatment period is at least three months. In some embodiments, the treatment period is at least six months. In some embodiments, the treatment period is at least nine months. In some embodiments, the treatment period is at least one year. In some embodiments, the treatment period is at least two years. In some embodiments, the treatment period is for as long as the subject is on hemodialysis.
  • MA or BP can include peripheral edema, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, dry eye, pain, fatigue, arthralgia, fracture, leg pain, arm pa , dizziness, pruritus dermatitis, earache, and anaphalaxis presenting as bronchospasm, hypotension, syncope, urticaria, and/or angioedema of the throat or tongue.
  • administration of an anti-OSMRB antibody results no serious adverse effects in the subject.
  • administration of an anti-OSMRB antibody does not result in one or more of peripheral edema, nasopharyngitis, upper respiratory tract infections, increased creatine phosphokinase, conjunctivitis, blepharitis, oral herpes, keratitis, eye pruritus, other herpes simplex virus infection, dry eye, pain, fatigue, arthralgia, fracture, leg pain, arm pain, dizziness, pruritus dermatitis, earache, and anaphalaxis presenting as bronchospasm, hypotension, syncope, urticaria, and/or angioedema of tire throat or tongue.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Chronic Idiopathic Pruritus (CIP).
  • the methods of the invention are used for treating pruritus in a subject having CIP.
  • the studies presented herein show that QSMRp mRNA levels are increased in subjects who have CIP in comparison to subjects who do not have CIP.
  • the method of treating CIP comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration. In some embodiments, the step of administering comprises intravenous administration. In some embodiments, the step of administering comprises intravenous administration followed by subcutaneous administration. In some embodiments, the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Chronic Spontaneous Urticaria (CSU), also known as Chronic Idiopathic Urticaria (CIU ).
  • CSU Chronic Spontaneous Urticaria
  • the methods of the invention are used for treating pruritus in a subject having CSU.
  • the studies presented herein show that OSMRp mRNA and protein expression levels are increased in subjects who have CSU in comparison to subjects who do not have CSU.
  • the method of treating CSU comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the method of treating CSU comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce urticaria relative to a control.
  • one or more symptoms of CSU are assessed by a change from baseline in UAS7, including, for example, itch or hives severity score.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-QSMRp antibody described herein is used treating pruritus associated with Chronic Idiopathic Urticaria (CIU).
  • the methods of the invention are used for treating pruritus in a subject having CIU.
  • the studies presented herein show that 08MKb mRNA and protein expression levels are increased in subjects who have CIU in comparison to subjects who do not have CIU.
  • the method of treating CIU comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • Tire method of treating CIU comprises administering to tire subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce urticaria relative to a control.
  • one or more symptoms of CIU are assessed by a change from baseline in IJAS7, including, for example, itch or hives se verity score.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Cutaneous Amyloidosis (CA).
  • the methods of the invention are used for treating pruritus in a subject having CA Tire method of CA comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Lichen Simplex Chronicus (LSC).
  • the methods of the invention are used for treating pruritus in a subject having LSC.
  • the studies presented herein show that OSMR mRNA expression levels are increased in subjects who have LSC in comparison to subjects who do not have LSC.
  • the method of treating LSC comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration. In some embodiments, the step of administering comprises intravenous administration. In some embodiments, the step of administering comprises intravenous administration followed by subcutaneous administration. In some embodiments, the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Plaque Psoriasis (PPs).
  • the methods of the invention are used for treating pruritus in a subject having PPs.
  • the method of PPs comprises administering to the sub j ect in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Lichen Planus (LP).
  • the methods of the invention are used for treating pruritus in a subject having LP.
  • the studies presented herein show that OSMRP mRNA expression levels are increased in subjects who have LP in comparison to subjects who do not have LP.
  • the method of treating LP comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Inflammatory Ichthyosis (II).
  • the methods of the invention are used for treating pruritus in a subject having II.
  • the method of treating II comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Mastocytosis (MA).
  • the methods of the invention are used for treating pruritus in a subject having MA.
  • the method of treating MA comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating pruritus associated with Bullous Pemphigoid (BP).
  • tire methods of the invention are used for treating pruritus in a subject having BP.
  • the method of treating BP comprises administering to the subject in need of treatment an anti-OSMRB antibody at a therapeutically effective dose and an administration interval for a treatment period sufficient to improve, stabilize or reduce one or more symptoms of pruritus relative to a control.
  • the step of administering comprises subcutaneous administration.
  • the step of administering comprises intravenous administration.
  • the step of administering comprises intravenous administration followed by subcutaneous administration.
  • the subcutaneous administration is through subcutaneous injection.
  • an anti-OSMRp antibody described herein is used in treating a TH2 -mediated inflammatory' disease Oncostatin M (OSM), a member of the gpl30 cytokine family, is involved in TH2 inflammation, epidermal integrity, and fibrosis.
  • OSM Oncostatin M
  • OSM signaling is independent of 1L-31.
  • the antibody can inhibit OSM-mediated pathways where OSM interacts with other signaling pathways, for example, IL-4 mediated pathway, IL-6 mediated pathway, IL-8 mediated pathway, IL-13 mediated pathway, and others.
  • the anti-OSMRp antibody described herein is used in combination with inhibitors of one or more signaling members of the TH2 mediated inflammatory pathways.
  • a subject who has CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP has el evated l evels of one or more cytokines associated with the OSMRp signaling pathway in comparison to a healthy subject.
  • the subject has elevated levels of one or more of IL-31, OSM, IL-3 IKcc, and QSMRfl in comparison to a healthy subject.
  • the subject has elevated levels of one or more of IL-31 in comparison to a healthy subject.
  • the subject has elevated levels of one or more of OSM in comparison to a healthy subject.
  • the subject has elevated levels of one or more of IL-3 IRcc in comparison to a healthy subject.
  • the subject has elevated levels of one or more of OSMRJ3 in comparison to a healthy subject.
  • LP, MA or BP results in a decrease or stabilization of MCP-1/CCL2 levels in the subject.
  • treating the subject results in a decrease of MCP-1 levels in comparison to the diseased state.
  • treating the subject results in stabilization of MCP-1 levels.
  • stabilization is meant that the levels of MCP-1 remain about the same and do not increase or decrease.
  • treating the subject results in reduced MCP-l levels in lymphocytes and/or endothelial cells.
  • MA or BP has WI-NRS scores of about 4, about 5, about 6, about 7, about 8 or above.
  • the subject in need of treatment has WI-NRS score of about 4. In some embodiments, the subject in need of treatment has WI-NRS score of about 5. In some embodiments, the subject in need of treatment has WI-NRS score of about 6. In some embodiments, the subject in need of treatment has WI-NRS score of about 7. In some embodiments, the subject in need of treatment has WI-NRS score of about 8. In some embodiments, the subject in need of treatment has WI-NRS score of more than 8.
  • a subject who has CIP, CIU, CSU, CA, PPs, LSC, LP, MA or BP is selected for treatment who has MCP-1/CCL2 levels greater than found in a healthy individual. In some embodiments, the subject selected for treatment does not have elevated levels of MCP-1/CCL2 in comparison to a healthy individual .
  • IL-31 expression level is elevated in the subject relative to a control. In some embodiments, IL-31 expression level is not elevated in the subject relative to a control.
  • IL-31 expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the IL-31 expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition.
  • IL- 31Ra expression level is elevated in the subject relative to a control.
  • OSM expression level is elevated in the subject relative to a control .
  • OSMR expression level is elevated in the subject relative to a control.
  • OSMRfi expression level is not elevated in the subject relative to a control.
  • OSMR expression level in a portion of the subject’s skin affected by a pruritic disease or condition is approximately the same as the OSMRp expression level in (i) a portion of the subject’s skin that is unaffected by the pruritic disease or condition, or (ii) a portion of normal skin from a healthy subject, who is not diagnosed with a pruritic disease or condition.
  • inventive compositions and methods provided by the present invention are used to deliver an anti- 08MKb antibody to a subject in need.
  • the anti-OSMRB antibodies are frilly-human monoclonal antibodies that specifically inhibit IL-31 and oncostatin M (OSM)-induced activation of the IL- 31 receptor and type II OSM receptor, respectively, through binding to OSMRft, the subunit common to both receptors.
  • the antibody is comprised of two light chains and two heavy chains.
  • the light chain contains a lambda constant region.
  • the constant regions of the heavy chain contain the CHI, hinge, and CH2 domains of a human immunoglobulin IgG4 antibody fused to the CH3 domain of a human IgGl antibody.
  • the heavy chain of the anti-OSMRB antibody contains a S228P modification to improve stability and a N297Q modification to remove an N -linked glycosylation site.
  • DIVAANTDYYFYYGMDV (SEQ ID NO: 7)
  • NINKRPS SEQ ID NO: 9
  • an anti-OSMRp antibody comprises a light chain complementary-determining region 1 (LCDR1) defined by SEQ ID NO: 8, a light chain complementary-determining region 2 (LCDR2) defined by SEQ ID NO: 9, and a light chain complementary-determining region 3 (LCDR3) defined by SEQ ID NO: 10; and a heavy chain complementary-determining region 1 (HCDR1 ) defined by SEQ ID NO: 5, a heavy chain complementary-determining region 2 (HCDR2) defined by SEQ ID NO: 6, and a heavy chain complementary-determining region 3 (HCDR3) defined by SEQ ID NO: 7.
  • LCDR1 light chain complementary-determining region 1
  • HCDR2 heavy chain complementary-determining region 2
  • HCDR3 heavy chain complementary-determining region 3
  • an anti-OSMRp antibody comprises CDR amino acid sequences with at least /5%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity with one or more of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7.
  • an anti-OSMRp antibody comprises a light chain variable domain having an amino acid sequence at least 90% identical to SEQ ID NO: 4 and a heavy chain variable domain having an amino acid sequence at least 90% identical to SEQ ID NO: 3
  • an anti-OSMRp antibody has a light chain variable domain amino acid sequence with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO:
  • an anti-OSMRp antibody comprises a light chain variable domain that has the amino acid sequence set forth in SEQ ID NO: 4 and a heavy chain variable domain that has the amino acid sequence set forth in SEQ ID NO: 3.
  • an anti-OSMRp antibody comprises a light chain having an amino acid sequence at least 90% identical to SEQ ID NO: 2 and a heavy chain having an amino acid sequence at least 90% identical to SEQ ID NO: 1.
  • an anti-OSMRp antibody has a Sight chain amino acid sequence with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO: 2 and a heavy chain amino acid sequence with at least 50%. 55%, 60%, 65%. 70%, 75%, 80%. 85%, 90%, 91 %, 92%, 93%, 94%, 95%,
  • an anti-OSMRp antibody comprises a light chain that has the amino acid sequence set forth in SEQ ID NO: 2 and a heavy chain that has the amino acid sequence set forth in SEQ ID NO: 1.
  • OSMRp antibody comprises CHI, hinge and CH2 domains derived from an IgG4 antibody fused to a CH3 domain derived from an IgGl antibody.
  • the CHI, hinge and CH2 domains derived from an IgG4 antibody comprise SEQ ID NO: 13.
  • the CH3 domain derived from an IgGl antibody comprises SEQ ID NO: 14.
  • the heavy chain constant region of an anti-OSMRp antibody according to the present invention comprises an amino acid sequence with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to
  • the heavy chain constant region of an anti-OSMRp antibody according to the present invention comprises an amino acid sequence with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO: 14.
  • the heavy chain constant region of an anti-OSMRp antibody according to the present invention comprises an amino acid sequence with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%,
  • an anti-OSMRp antibody according to the present invention comprises a lambda constant domain derived from an IgG antibody ln some embodiments, the lambda constant domain derived from an IgG comprises SEQ ID NO: 16. In some embodiments, an anti-OSMRp antibody according to the present invention comprises an amino acid sequence with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO: 16.
  • Interleukin-31 induces cytokine and chemokine production from human bronchial epithelial cells through activation of mitogen- activated protein kinase signaling pathways: implications for the allergic response. Immunology. 2007 Dec;122(4):532-41.
  • Kanda N Watanabe S. Increased serum human ⁇ defensin-2 levels in atopic dermatitis:
  • Previously quarantined animals were acclimated to the study room for a minimum of 14 days prior to initiation of dosing. Acclimation phase data was collected from all animals, including spares. During acclimation, each animal was monitored using the Noldus video monitoring system for a duration of at least 30 minutes, and tire number of scratching or grooming events was recorded. Animals that had more than 40 scratching/grooming events during the 30 minute pre-screen duration were replaced with available spares and removed from the study.
  • Clinical observtions were performed twice daily for each animal beginning on the second day of acclimation (Day -13). The first observation occurred in the morning, prior to room cleaning. The second observation was no sooner than four hours after the morning observation (and not during video monitoring). Additional clinical observations were performed as necessary. If clinical observations for an animal demonstrated declining animal condition, a veterinary evaluation was performed.
  • blood was collected via a single draw' and then divided appropriately. If possible, venous blood samples were collected from conscious unscheduled animals prior to anesthesia and necropsy.
  • Figure IB shows raw scratching behavior on the left vertical axis plotted alongside serum concentration of anti-OSMRp antibody shown on the right vertical axis. Data are shown for a single IV administration of anti-OSMRp antibody at 1 mg/kg (left panel), 3 mg/kg (center panel) and 10 mg/kg (right panel). Results from tins PK/PD correlation define a concentration range of 5 pg/ ' ml to 8.5 pg/ml at or above which the anti-OSMRp antibody provides protection from a supra-physiologic concentration of human IL-31 -induced pruritus.
  • the study in this example is designed to evaluate the safety , tolerability , PK and immunogenieity of an anti-OSMRp antibody in subjects with atopic dermatitis.
  • the study also includes exploratory investigations of pharmacogenetics and the effect of tire anti-OSMRB antibody on clinical effect assessments, gene expression, and PD measures.
  • An anti-OSMRB antibody is administered intravenously (IV) to subjects with moderate to severe atopic dermatitis experiencing moderate to severe pruritus. Additionally, the anti-OSMRB antibody is administered subcutaneously (SC) to one group of subjects with moderate to severe atopic dermatitis experiencing moderate to severe pruritus.
  • Subjects are enrolled into one of seven groups as described below. After verification of eligibility, subjects are randomized to receive the anti-OSMRB antibody or placebo. In six of the groups, the anti-OSMRp antibody or placebo is administered IV. In the seventh group, subjects receive either the anti-OSMRp antibody or placebo as a single SC injection.
  • the first group receives 0.3 mg/kg anti-OSMRp antibody or placebo intravenously.
  • the second group receives 1.5 mg/kg anti-OSMRp antibody or placebo intravenously.
  • the third group receives 5 mg/kg anti-OSMRp antibody or placebo intravenously.
  • the fourth group receives 10 mg/kg anti-OSMRp antibody or placebo intravenously.
  • the fifth group receives 20 mg/kg anti- OSMRp antibody or placebo intravenously.
  • the sixth group receives 7.5 mg/kg anti-OSMRp antibody or placebo intravenously.
  • the seventh group receives 1.5 mg/kg anti-OSMRp antibody or placebo subcutaneously. Following dosing, subjects undergo at least 2 days of safety monitoring and intensive PK sampling while confined at the clinical research unit. The PK samples are collected at pre-specified timepoints.
  • the anti-OSMRp antibody drug product is a sterile liquid formulation, supplied as a single use vial for IV or SC injection. 3 mL Schott vials are filled with 2.3 ml, to allow for a delivered volume of 2 mL, for an extractable dose of 200 mg/vial.
  • the anti-OSMRp antibody drug product is diluted to a volume of 100 ml, for IV infusions.
  • Doses administered IV are diluted in saline to a total volume of 100 mL and infused over 1 hour. Subjects are observed closely for any infusion reactions. The infusion is stopped in the event of signs and symptoms suggesting an infusion reaction. The infusion is restarted upon resolution of the signs and symptoms related to the infusion reaction. The duration of infusion can be lengthened to longer than 1 hour during the course of the study.
  • Subjects also have to have a physician-documented diagnosis of atopic dermatitis for at least 1 year and a diagnosis of moderate to severe disease, defined as IGA of 3 or 4, and body surface area (BSA) involvement of 10% or more, for at least 3 months before Screening Visit 1.
  • a physician-documented diagnosis of atopic dermatitis for at least 1 year and a diagnosis of moderate to severe disease, defined as IGA of 3 or 4, and body surface area (BSA) involvement of 10% or more, for at least 3 months before Screening Visit 1.
  • IGA body surface area
  • the Investigator’s Global Assessment is an overall assessment that is performed on each subject.
  • the IGA utilizes a 6-point scale ranging from 0 (clear) to 5 (very- severe disease).
  • An IGA score is assigned based on morphology without referring back to the baseline state.
  • the IGA score is recorded in the electronic Case Report Form (e-CRF).
  • the Eczema Area and Severity Index (EASI) score is used to measure the severity and extent of atopic dermatitis.
  • the 4 body regions (head and neck, trunk, upper limbs, and lower limbs) are assessed separately for erythema, infiltration/papulation, excoriation, and iichenification
  • the average clinical severity' of each sign in each of the 4 body regi ons is assigned a score of 0 to 3, based on severity of disease, and tire score is recorded in the e-CRF.
  • the area of skin involved in each body region is determined and assigned a score of 0 to 6, based on extent of involvement, and the score is recorded in the e-CRF.
  • Total EASI score at each visit is calculated at the end of the study. Qualified dermatologists perform EASI assessment for this study.
  • Scoring Atopic Dermatitis is utilized to assess the severity of atopic dermatitis.
  • the SCORAD is a tool used in clinical research and practice that was developed to standardize the evaluation of the extent and severity of atopic dermatitis.
  • the SCORAD incorporates both objective physician estimates of extent and severity of disease as well as subjective subject assessment of itch and sleep loss. The percentage of each body area affected by atopic dermatitis is determined and the sum of all areas are reported.
  • the severity of 6 symptoms of AD is rated as none (0), mild (1), moderate (2), or severe (3).
  • SCORAD Calculates itch and sleeplessness. The SCORAD is calculated based on a pre defined formula.
  • BSA Body Surface Area
  • the Dermatology Life Quality Index is a 10-question questionnaire that takes into account symptoms and feelings, daily activities, leisure, school, personal relationships, and treatment. Each question is answered on a scale of 0 to 3 (0 for not at all, 1 for a little, 2 for a lot, and 3 for very much), talcing into account the previous week. The scores are added with minimum of 0, meaning no effect on quality of life, and 30, meaning extremely large effect.
  • the Hospital Anxiety and Depression Scale is a general Likert scale used to detect states of anxiety and depression.
  • the 14 items on the questionnaire include 7 that are related to anxiety and 7 that are related to depression.
  • Each item on tire questionnaire is scored on a scale of 0 to 3 with a possible total score between 0 and 21 for each parameter
  • Actigraphy utilizes a portable device (actigraphy watch) that records movement over extended periods of time. Subjects wear a wrist actigraphy watch at night on the non dominant wrist to monitor sleep quality and quantity.
  • AE adverse event
  • Any clinically significant worsening from baseline in subjects’ signs and symptoms of atopic dermatitis is considered an adverse event (AE) (e.g., atopic dermatitis worsening/flare) and triggers consultation with the study site dermatologist, determination of the IGA (for inclusion in the e-CRF), preparation of a detailed clinical summar ' and reporting within 24 hours. Any changes or additions to the subject’s concomitant medications are entered into the e-CRF with appropriate start and stop dates. During the study, all adverse events and severe adverse events are followed until resolution.
  • AE adverse event
  • the study in this example is designed to evaluate the safety, tolerability, PK and immunogenicity of an anti-OSMRp antibody in subjects on hemodialysis with uremic pruritus.
  • the study also includes exploratory investigations of pharmacogenetics and the effect of the anti- OSMRp antibody on clinical effect assessments, gene expression, and PD measures.
  • An anti-OSMR antibody is administered intravenously (IV) to subjects on hemodialysis with uremic pruritus.
  • Subjects are enrolled in one treatment group. After verification of eligibility, subjects are randomized to receive 5 mg/kg or 10 mg/kg of the anti-OSMRp antibody or placebo on Day 0, the day before a regularly scheduled hemodialysis session.
  • PK samples are collected at pre-specified timepoints. Intensive PK sampling is performed at the time of certain hemodialysis sessions. Pre- and post-dialysis blood samples as well as pre- and post-dialyzer samples and dialysate samples are collected at specified timepoints for anti-OSMR
  • Doses administered IV are diluted in saline to a total volume of 100 mL and infused over 1 hour. Subjects are observed closely for any infusion reactions. The infusion is stopped in die event of signs and symptoms suggesting an infusion reaction. The infusion is restarted upon resolution of the signs and symptoms related to the infusion reaction . The duration of infusion can be lengthened to longer than 1 hour during the course of the study.
  • Subjects also have to have end stage renal disease (ESRD) at Screening Visit 1 and be undergoing a three-times-per-week hemodialysis regimen that has been stable for at least 3 months before Screening Visit 1 .
  • ESRD end stage renal disease
  • PK parameters are calculated for each subject, whenever possible, based on the serum concentrations of the anti-OSMRp antibody:
  • the following samples are collected for each subject on hemodialysis days designated for intensive PK sampling: blood immediately before and after the hemodialysis run; a dialysate sample; and samples from upstream and downstream of the dialyzer, urine samples before and after hemodialysis (for subjects capable of producing urine), and a 24-hour urine sample (for subjects capable of producing urine) sometime between Day 0 to Day 2 while confined at the clinical research unit. Weight and standing and supine blood pressure before and after hemodialysis are also recorded. In addition, the hemodialysis flow rate, volume of dialysate, and other hemodialysis parameters are also collected and recorded in the e-CRF.
  • Medications given during hemodialysis are also recorded m the e-CRF.
  • the following additional parameters are also calculated for each subject, whenever possible, based on serum and dialysate concentrations of the anti-OSMRp antibody: dialysate clearance and dialysate extraction ratio calculated as the percentage of administered dose extracted during hemodialysis. The hemodialysis flow' rate and volume of dialysate are recorded.
  • Descriptive statistics (arithmetic mean, standard deviation, minimum, median, maximum, geometric mean, and geometric coefficient of variation, as appropriate) are listed and summarized for serum concentrations of anti-OSMRp antibody and PK parameters.
  • the Dermatology Life Quality Index is a 10-question questionnaire that takes into account symptoms and feelings, daily activities, leisure, school, personal relationships, and treatment. Each question is answered on a scale of 0 to 3 (0 for not at all, 1 for a little, 2 for a lot, and 3 for very much), taking into account the previous week. Hie scores are added with minimum of 0, meaning no effect on quality of life, and 30, meaning extremely large effect.
  • the Hospital Anxiety and Depression Scale is a general Likert scale used to detect states of anxiety and depression.
  • Tire 14 items on the questionnaire include 7 that are related to anxiety and 7 that are related to depression.
  • Each item on the questionnaire is scored on a scale of 0 to 3 with a possible total score between 0 and 2 ! for each parameter
  • Actigraphy utilizes a portable device (actigraphy watch) that records movement over extended periods of time. Subjects wear a wrist actigraphy watch at night on the non dominant wrist to monitor sleep quality and quantity.
  • the study in this example is designed to evaluate the safety , tolerability, PK and immunogenicity of an anti-OSMRp antibody in healthy subjects and in adult subjects with atopic dermatitis (AD) in a randomized, double-blind, placebo (PBO)-controlled, single-ascending dose study of the anti-OSMRp antibody.
  • AD was used as a proxy for IL-31 -driven pruritic diseases to assess target engagement and Early Signal of Efficacy.
  • An anti-OSMRB antibody was administered intravenously (IV) to four groups of adult healthy volunteer (HV) subjects. Additionally, the anti-OSMRp antibody was administered subcutaneously (SC) to two groups of HV subjects. Three groups of AD subjects with moderate to severe atopic dermatitis experiencing moderate to se vere pruritus were administered anti-OSMRp antibody intravenously. Additionally, one group of AD subjects with moderate to severe atopic dermatitis experiencing moderate to severe pruritus was administered anti-OSMRp antibody subcutaneously. Die study design is outlined in Figure 2.
  • HV subjects were enrolled into one of six groups as described below. After verification of eligibility, HV subjects were randomized to receive the anti-OSMRp antibody or placebo. In four of the groups, the anti-OSMRB antibody or placebo was administered IV. In the fifth and sixth groups, HV subjects received either the anti-OSMRB antibody or placebo as a single SC injection.
  • the first group received 1.5 mg/kg anti-OSMRp antibody or placebo intravenously; six HV subjects received the anti-OSMRp antibody, and two HV subjects received placebo.
  • the second group received 5 mg/kg anti-OSMRB antibody or placebo intravenously; six HV subjects received the anti-OSMRB antibody, and two HV subjects received placebo.
  • the third group received 10 mg/kg anti-OSMRp antibody or placebo intravenously; six HV subjects received the anti-OSMRB antibody, and two HV subjects received placebo.
  • the fourth group received 20 mg/kg anti-OSMRp antibody or placebo intravenously; six HV subjects received the anti-OSMRp antibody, and two HV subjects received placebo.
  • the fifth group received 1 .5 mg/kg anti-OSMRB antibody or placebo subcutaneously; six HV subjects received the anti-OSMRp antibody, and two HV subjects received placebo.
  • the sixth group received 360 mg of anti-OSMRB antibody or placebo subcutaneously; six HV subjects received the anti-OSMRp antibody, and five HV subjects received placebo.
  • the study design is represented graphically in Figure 2, left panel.
  • AD subjects were enrolled into one of four groups as described below. After verification of eligibility, AD subjects were randomized to receive the anti-OSMRp antibody or placebo. In three of the groups, the anti-OSMRp antibody or placebo was administered IV. In the fourth group, AD subjects received either the anti-OSMRp antibody or placebo as a single SC injection.
  • the first group received 0.3 mg/kg anti-OSMRp antibody or placebo intravenously; three AD subjects received the anti-OSMRp antibody, and two AD subjects received placebo.
  • the second group received 1.5 mg/kg anti-OSMRp antibody or placebo intravenously; three AD subjects received the anti-OSMRp antibody, and two AD subjects received placebo.
  • the third group received 7 5 mg/kg anti-OSMRp antibody or placebo intravenously; ten AD subjects received the anti-OSMRp antibody, and six AD subjects received placebo.
  • the study design is represented graphically in Figure 2, lower right panel.
  • Intravenous (IV) or subcutaneous (SC) anti-OSMRp antibody was administered in escalating dose cohorts: HV IV : 1 .5, 5, 10, and 20 mg/kg; HV SC: 1.5 mg/kg and 360 mg; AD IV: 0.3, 1.5 and 7.5 mg/kg; AD SC: 1.5 mg/kg ( Figure 2).
  • TCS topical corticosteroids
  • Safety and tolerability data included vital signs, physical examination, ECG, laboratory measures, and adverse events (AEs).
  • Anti-OSMRp antibody target engagement and clinical pharmacodynamic (PD) data included daily e-diary WI-NRS and periodic Sleep-Loss Visual Analogue Scale (VAS) until Day 60. Weekly average of daily WI-NRS -was calculated.
  • Drug-related treatment-emergent AEs were infrequent and showed no dose response correlation and all resolved without sequalae: in HVs, 1 mild headache (5 mg/kg IV), 1 mild flushing (1.5 mg/kg SC), and 1 mild anemia (360 mg SC); in AD subjects: 1 mild headache/mild decreased appetite (1.5 mg/kg IV), 1 moderate dizziness (7.5 mg/kg IV), 1 mild dizziness (1.5 mg/kg SC), and 1 mild somnolence (PBO IV). None of the following was observed in any patients treated with anti-OSMRp antibody: deaths, Serious Adverse Events; discontinuations due to AEs; infusion reactions; injection site reactions;
  • FIG. 3A Mean change from baseline in weekly average Pruritus Visual Analog Scale (VAS) are shown in Figure 3A.
  • Figure 3B show's mean percent change in VAS pruritus score from baseline.
  • WI-NRS Worst Itch Numerical Rating Scale
  • Figure 3C Mean change in weekly average Wl-NRS from baseline is shown in Figure 3D.
  • Mean percentage change in weekly average pruritus VAS (a component of SCORAD) was greater in anti-OSMRB antibody recipients vs. PBO: -55.4% active vs. -10 4% PBO on Day 28 (Figure 3B).
  • Mean percentage change in weekly average Wl-NRS was greater in anti-OSMRfi antibody recipients vs. PBO: - 40.7% active vs.
  • Figures 4 and 5A-5D show the percentage of subjects with a >4-point reduction in average weekly WI-NRS from baseline.
  • a >4 point reduction in NRS from baseline is generally considered a clinically meaningful change.
  • a higher percentage of anti-OSMRp antibody recipients demonstrated a >4 -point decrease in weekly average WI-NRS vs. PBO consistently throughout the duration of the study as shown in Figure 4.
  • 50% of the active group demonstrated a >4-point decrease in weekly average WI-NRS vs. 10% in the PBO group.
  • Figure 5A-5D shows the percentage of subjects who responded with a particular magnitude of NRS reduction from baseline (>4 points).
  • Figure 5A and 5C show the respective percentages of anti-OSMRp antibody recipients and Figure SB and SB show the respective percentages of placebo recipients.
  • responder rates were calculated using a denominator that includes subjects with non-missing values.
  • responder rates were calculated using a denominator that includes all subjects. Rescued subjects were considered non-responders in tins assessment.
  • the anti- OSMRp antibody recipients demonstrated a greater magnitude decrease in weekly average WI- NRS vs. PBO consistently throughout the duration of the study. The maximum decrease in WI- NRS at day 28 was greater in anti-OSMRp antibody recipients vs PBO: >8 points active vs 4 points PBO.
  • anti-OSMRp antibody recipients demonstrated a persistent effect on weekly-average WI-NRS through Day 56 in combination with the use of concomitant TCS during the adjunctive therapy period (Figure 3C-D, Figure 4, and Figures 5A-D). Concordant with the effect on pruritus, anti-OSMRp antibody recipients reported improved sleep vs. PBO ( Figures 6A-B), as evidenced by a greater decrease in sleep- loss VAS (a component of SCORAD): -59.5% active vs. -2.3% PBO on Day 28 (Figure 6B).
  • Figure 7A-B shows the change in Eczema Area and Severity Index (EASI) from baseline in antibody and placebo recipients.
  • EASI Eczema Area and Severity Index
  • EASI Eczema Area and Seventy Index
  • OSMRp antibody A single dose of OSMRp antibody at 7.5 mg/kg resulted in serum levels above 5 pg/mL (5 8-28 2 pg/mL) in 80% of recipients 44 to 47 days post-dose.
  • WI-NRS, pruritus YAS, and sleep-loss YAS were compared between 10 anti- OSMRj3 antibody (7.5 mg/kg IV) recipients and 10 PBO IV recipients between days 29-60.
  • Anti- OSMRp antibody recipients experienced a greater WI-NRS improvement that continued into the adjunctive therapy period during which they received concomitant TCS and reached a maximum level at 6 weeks: -51% vs -26.3%.
  • FIG. 10 A The simulated plot in Figure 10 A was derived from plasma concentrations of anti-OSMRp antibody in non-human primates, HV and AD patients.
  • Figure 10A shows a simulated median plot of antibody concentration in plasma over the indicated time in weeks following subcutaneous (SC) or intravenous (IV) administration to heathy volunteers (HV) or Atopic Dermatitis (AD) patients.
  • the upper dotted line indicates the ECvo of the anti-OSMRp antibody in providing protection from supra-physiologic human IL-31 challenge-induced pruritus in non-human primates.
  • the lower dotted line indicates tire EC?. ⁇ of the anti-OSMRp antibody in providing protection from supra-physiologic human IL-31 challenge-induced pruritus in non-human primates.
  • FIG. EC ?j and EC 90 were determined from the study described in Example 1.
  • Figure JOB shows plasma anti-OSMRp antibody concentration profiles for the indicated doses in Atopic Dermatitis (AD) patients.
  • anti-OSMRp antibody exposure (as measured by AUCO-oo) was similar in healthy volunteers and Atopic Dermatitis patients and approached linearity with increasing dose levels. Bioavailability between healthy volunteers and AD subjects at the evaluated SC dose levels was generally comparable (42% vs. 65%, respectively).
  • Anti-OSMRp antibody showed dose-dependent elimination consistent with a target-mediated drug disposition (TMDD) profile.
  • TMDD target-mediated drug disposition
  • At the 7.5 mg/kg TV dose level, anti-OSMRp antibody was detectable through at least 8 weeks.
  • the modeled PK parameters predict viability of subcutaneous administration, and predict that a fixed subcutaneous dose of 360 mg of anti-OSMRp antibody can achieve exposures similar to a 7.5 mg/kg IV dose of the antibody.
  • OSMRp antibody following IV and SC administration in adult healthy volunteers (HV) and subjects with AD, and investigate various SC dosing regimens to optimize practical chronic dosing in a target population.
  • Single dose data from a Phase lb clinical study in 57 HV and subjects with AD were analyzed.
  • Hie PK of the anti- OSMRp antibody in HV and AD subjects following single-dose IV or SC administration was described using a target-mediated drug disposition (TMDD) model to account for its non-linear clearance.
  • TMDD target-mediated drug disposition
  • Association and dissociation rate constants were determined experimentally at 0.734 nM-hr ’! and 0.268 nM-hr ] , respectively, and fixed during model development.
  • Relative bioavailability of SC administration in AD was estimated for the model at 65% (based on the comparison of PK of 1.5 mg/kg IV and SC in HV and AD subjects and then revised for dose- dependency based on PK of 360 mg SC in HVs).
  • Body weight was included as a covariate on the central volume of distribution based on allometrie theory?
  • Figure 10C depicts simulations of various dosing regimens using the final population PK model.
  • a range of simulations performed to evaluate various SC dosing regimens using an exemplary dose of anti-OSMRp antibody (360 rng in 2mL SC injection). Exposure metrics and time to steady-state were derived for each simulated SC dosing regimen.
  • the model (including TMDD) was used to simulate future dosing scenarios for chronic SC dose administration in patients with chronic pruritic diseases in which the target receptor may be upregulated. This model also supports determination of practical chronic dose(s)/dosing intervals using a Ceff derived from clinical trials with anti-OSMRp antibodies of the invention.
  • Prurigo nodularis is a chronic skin disease of unknown etiology characterized by symmetrically-distributed, intensely-pruritic hyperkeratotic nodules. Comorbidities featuring chrome pruritus are implicated in PN pathogenesis by initiating the itch-scratch cycle that leads to nodule formation.
  • OSMRp the shared receptor subunit for IL-31 and oncostatin M (QSM) signaling, involved in pruritus, inflammation and fibrosis, in PN pathogenesis is unknown.
  • QSM oncostatin M
  • IL-31 -expressing mononuclear cells were present in 89% of lesional biopsies (immunohistochemistry) whether or not an underlying condition was identified.
  • IL-31, IL-3 IRa, OSM, and 08MKb expression in mononuclear cells were upregulated m lesional biopsies versus non-lesional biopsies (p ⁇ 0.001).
  • IL-31 mRNA was expressed in 44% of lesional PN, 16% of non-lesional PN, 12.5% of healthy volunteer, and 100% of AD biopsies (lesional [LS] and non-lesional [NL]).
  • IL-31 mRNA was expressed in 64% of LS biopsies from PN patients with WI-NRS >7.
  • IL-31 protein (IHC) was expressed in mononuclear cells in the majority of LS PN biopsies (89%) vs 44% of NL PN biopsies.
  • Polymorphonuclear cells when present, and endothelial cells were other common sources of IL-31 in LS PN skin.
  • 3 mRNA was ubiquitous (74-100%) in LS or NL PN, AD or healthy volunteer biopsies.
  • a higher proportion of LS PN biopsies contained mononuclear cells expressing IL-3 IRa (1.7-fold), OSM (3.6-fold), and OSMRp (1.8-fold) protein than NL PN biopsies.
  • Epidermal cells, and when present, PMN, dermal nerves, and adnexal structures were other common sources of IL-3 IRa and OSMRp in LS PN sk . See Example 7 for further details of this study.
  • PN is a distinct, highly pruritic chronic skin disease that is not defined by its comorbid conditions.
  • IL-31 is implicated in the pathogenesis of PN given its prevalent expression in PN nodules. Tims, the role of IL-31 in the disease mechanism of PN is hereby elucidated.
  • the study in this example further demonstrated that the anti-OSMRp antibody of the invention can effectively treat inflammation.
  • the objectives of these studies were to characterize the in vitro responses of human epidermal keratinocytes (HEK) and human dermal fibroblasts (HDF) to OSM in comparison to LIF and IL-31, using chemokine monocyte chemoattractant protein 1 (MCP-l/CCL-2), which has roles in inflammatory' responses.
  • MCP-l/CCL-2 chemokine monocyte chemoattractant protein 1
  • FIG 11 shows the receptor structure for IL-31 signaling and that of OSM signaling.
  • OSM interacts with two receptors in humans, a type I receptor and a type II receptor.
  • the type I receptor complex comprises a receptor heterodimer of LIFRa and gp!30.
  • the type II receptor complex comprises a receptor heterodimer of 08MKb and gpl30.
  • the data presented in this example show that administering an anti-OSMRfl antibody targets and attenuates OSM-mediated TH2 inflammatory signaling pathway m human epidermal keratinocytes (HEK) and human dermal fibroblasts (HDF) cells.
  • the data also indicate that the antibody can inhibit OSM-mediated inflammatory' pathways independent of IL-31 involvement.
  • Oneostatin M (OSM), a member of the gpl30 cytokine family, is involved in TH2 inflammation, epidermal integrity, and fibrosis.
  • OSM monocyte chemoattractant protein 1
  • IL-4 interleukin-4
  • IL-13 interleukin-13
  • OSMR[3 anti-OSM receptor b
  • MCP-1 levels in supernatants were determined by ELISA. MCP-1 and receptor chain RNAs were measured.
  • OSM (50 ng/mL) strongly- induced MCP-1 protein (in HEK; p ⁇ 0.000l and HDF; p ⁇ 0.0l, Figure 12, panel A) and mRNA (in HEK; p ⁇ 0.0001 and HDF; p ⁇ 0.05, Figure 12, panel B) at 24 hours
  • OSM (but not LIF or IL-31) induced phosphorylation of STAT3 or STAT1 and synergized with either IL-13 or EL-4 in elevating MCP-1 (p ⁇ 0.01). Results were similar for OSM in HDF; LIF or IL-31 minimally activated STAT3 but not MCP- 1.
  • OSM significantly induced mRNA for the receptor chains of type II IL-4 receptor (IL-4Ra/IL!3Ra) and type II OSM receptor OSMRJ3/gpl30 (HEK, p ⁇ 0.05; HDF, p ⁇ 0.01;), but not for chains of LIF receptor or IL-31Ra.
  • Die data in Figure 14 were obtained from HEK cells and show an increase in IL13Ra and IL-4Ra mRNA at 6 hours and 24 hours after treatment with OSM. These data indicate that OSM stimulates mRNA for the receptor chains of type II IL-4 receptor and type II OSM receptor complexes in HDF cells.
  • Anti-IL-31 Ra or isotype control antibody had no significant effect on the OSM- and OSM+IL-4-induced responses ( Figure 15, panels B and C, and Fignre 16, panel B).
  • Figure 15, panels B and C, and Fignre 16, panel B Collectively, the data presented in this example show that OSM regulates expression of pro- inflammatory chemokine MCP-l/CCL-2 in HEK and HDF cells.
  • These data also show that OSM synergizes with TH2 cytokines (IL-4 and IL-13) to induce MCP-l/CCL-2 in the cells, while LIE or IL-31 do not in this system.
  • TH2 cytokines IL-4 and IL-13
  • the anti-OSMRjl monoclonal antibody reduced both the OSM induction and the synergistic OSM+IL-4 induction of MCP-l/CCL-2 protein production. Potent inhibition of OSM activity suggests therapeutic potential of the anti-OSMRp monoclonal antibody in TH2 -mediated diseases distinct from the anti-OSMRp antibody’s inhibition of IL-31.
  • OSMRp axis molecules 1L-31, QSM, IL-3 IRcc, and QSMRfi are present in PN and in atopic dermatitis (AD) skin samples.
  • IL-31 was detected more frequently in lesional (LS) biopsies than in non-les!onai (NL) biopsies.
  • Furthennore the intensity or upregulation of IL-31 expression increased with itch severity in PN patients.
  • lympho-monocytes and endothelial cells are common sources of IL-31 and OSM in both NL and LS tissues.
  • lympho-monocytes from LS biopsies showed significantly higher expression of all target proteins, compared to NL biopsies, ( Figure 20, panels E-H).
  • IL-3 IRa and OSMRp protein levels in lympho- monocytes correlated with itch severity ( Figure 21, panels A-D).
  • Skin biopsy samples were obtained from subjects who have chronic idiopathic urticaria (( ' i t ) and from control subjects who do not have an inflammatory or pruritic skin disease or disorder in order to assess OSMRp mRNA and protein expression levels in the samples.
  • OSMRp mRNA levels w ' ere assessed using RNAscope® in situ hybridization (ISH) and nanoString ® technologies ( Figure 22, panels A and B, respectively).
  • ISH RNAscope® in situ hybridization
  • Figure 22, panels A and B nanoString ® technologies.
  • ISH RNAscope® in situ hybridization
  • 12 human CIU skin samples and 4 human normal skin samples were evaluated in in accordance with standard methods. The same patient samples w'ere used to evaluate OSMRp mRNA expression by RNAscope® and nanoString® technologies.
  • IHC immunohistochemistry
  • OSMRp mRNA is Increased in Lichen Simplex Chronicus (LSC), Lichen Planus (LP) Skin Biopsies and Chronic Idiopathic Pruritus (CIP)
  • LSC Chronicus
  • LP Lichen Planus
  • Example 10 Anti-OSMRp antibody placebo-controlled repeated-single-dose Phase lb study in patents with moderate to severe atopic dermatitis
  • the study in this example is designed to evaluate the efficacy, safety and tolerability of an anti-OSMRp antibody in subjects with moderate to severe atopic dermatitis (AD). Study Design on Subjects with Moderate to Severe Atopic Dermatitis
  • An anti-OSMRb antibody was administered subcutaneously (SC) to patients with moderate to severe atopic dermatitis experiencing moderate to severe pruritus, in a randomized, placebo-controlled study.
  • SC subcutaneously
  • the patients received weekly doses of either placebo or 360 mg of anti-OSMRp antibody for 12 weeks, and were monitored for 21 weeks.
  • TCS was provided as a rescue medication for AD flares as needed.
  • WI-NRS Worst-Itch Numerical Rating Score
  • VAS Visual Analogue Scale
  • periodic Sleep-Loss VAS was recorded in a daily e-diary.
  • atopic dermatitis disease seventy, IGA, EASI, SCORAD were performed at specified study visit.
  • Safety and tolerability data included vital signs, physical examination, ECG, laboratory measures, and adverse events (AEs).
  • the anti-QSMR antibody administered according to the present invention was effective in decreasing the severity of pruritus.
  • FIG 27A there was a rapid and sustained reduction in WI-NRS from baseline in the group treated with anti-OSMR antibody compared to placebo throughout the 12-w'eek study.
  • Mean percentage change in weekly-average WI-NRS decreased by 55.8% in anti-OSMR antibody recipients compared to a 32.5% decrease in placebo recipients at Week 12 (p ⁇ 0.0l).
  • significantly higher percent of patents injected with the anti-OSMR antibody had > 4-point reduction in an average weekly WI-NRS from baseline as shown in Figure 27B.
  • Subjects who flared during the trial tended to have higher serum IgE levels (Figure 29) and a higher baseline eosinophil count and a higher percent of eosinophils of total leukocytes in the blood on average ( Figure 30). Additionally, in the group of patients that flared in the study, 75% had IgE levels above 300 IU/ml, while in the group of patients who did not flare, 89% had IgE levels below 300 Ul/m!. Furthermore, in the group of patients with greater than 300 IU/ml, 90% flared, while in the group of patients with less than 300 IU/ml only 27% flared.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dermatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne, entre autres, des procédés de traitement de maladies ou de troubles prurigineux ou inflammatoires de la peau, ou d'un prurit associé à une maladie ou un trouble, avec un anticorps anti-OSMRβ. En particulier, la présente invention concerne un procédé de traitement de la dermatite atopique comprenant la sélection d'un sujet qui n'a pas reçu de traitement préalable avec un corticoïde topique ou la sélection d'un sujet sur la base d'un taux d'IgE sérique ou d'un comptage d'éosinophiles sériques et l'administration au sujet d'un anticorps anti-OSMRβ à une dose thérapeutiquement efficace et un intervalle d'administration pour un traitement visant à améliorer, stabiliser ou réduire un symptôme associé à une dermatite atopique par rapport à un témoin.
PCT/US2019/046065 2018-08-13 2019-08-09 TRAITEMENT DE MALADIES OU DE TROUBLES DE LA PEAU PAR ADMINISTRATION D'UN ANTICORPS ANTI-OSMRβ WO2020036833A1 (fr)

Applications Claiming Priority (16)

Application Number Priority Date Filing Date Title
US201862718324P 2018-08-13 2018-08-13
US62/718,324 2018-08-13
US201862765033P 2018-08-16 2018-08-16
US62/765,033 2018-08-16
US201862731618P 2018-09-14 2018-09-14
US62/731,618 2018-09-14
US201862757047P 2018-11-07 2018-11-07
US62/757,047 2018-11-07
US201862775350P 2018-12-04 2018-12-04
US62/775,350 2018-12-04
US201962789434P 2019-01-07 2019-01-07
US62/789,434 2019-01-07
US201962794356P 2019-01-18 2019-01-18
US62/794,356 2019-01-18
USPCT/US2019/044532 2019-07-31
US2019044532 2019-07-31

Publications (1)

Publication Number Publication Date
WO2020036833A1 true WO2020036833A1 (fr) 2020-02-20

Family

ID=67777414

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/046065 WO2020036833A1 (fr) 2018-08-13 2019-08-09 TRAITEMENT DE MALADIES OU DE TROUBLES DE LA PEAU PAR ADMINISTRATION D'UN ANTICORPS ANTI-OSMRβ

Country Status (1)

Country Link
WO (1) WO2020036833A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013168829A1 (fr) * 2012-05-11 2013-11-14 Wakayama Medical University Anticorps anti-récepteur bêta de l'oncostatine m
WO2014194274A2 (fr) * 2013-05-30 2014-12-04 Biogen Idec Ma Inc. Protéines de liaison à l'antigène du récepteur de l'oncostatine m

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013168829A1 (fr) * 2012-05-11 2013-11-14 Wakayama Medical University Anticorps anti-récepteur bêta de l'oncostatine m
WO2014194274A2 (fr) * 2013-05-30 2014-12-04 Biogen Idec Ma Inc. Protéines de liaison à l'antigène du récepteur de l'oncostatine m

Non-Patent Citations (43)

* Cited by examiner, † Cited by third party
Title
"Bioinformatics Methods and Protocols (Methods in Molecular Biology", vol. 132, 1999, HUMANA PRESS
ALTSCHUL ET AL., METHODS IN ENZYMOLOGY
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
ALTSCHUL ET AL.: "Basic local alignment search tool", J MAL. BIOL., vol. 215, no. 3, 1990, pages 403 - 410, XP002949123, doi:10.1006/jmbi.1990.9999
BAXEVANIS ET AL.: "Bioinformatics: A Practical Guide to the Analysis of Genes and Proteins", 1998, WILEY
BILSBOROUGH JLEUNG DYMAURER MHOWELL MBOGUNIEWICZ MYAO LSTOREY HLECIELHARDER BGROSS JA: "IL-31 is associated with cutaneous lymphocyte antigen-positive skin homing T cells in patients with atopic dermatitis", J ALLERGY CLIN IMMUNOL., vol. 117, no. 2, February 2006 (2006-02-01), pages 418 - 25, XP005275428, doi:10.1016/j.jaci.2005.10.046
BOGUNIEWICZ MLEUNG DY: "Atopic dermatitis: a disease of altered skin barrier and immune dysregulation", IMMUNOL REV., vol. 242, 2011, pages 233 - 46, XP055581377, doi:10.1111/j.1600-065X.2011.01027.x
BONIFACE KATIA ET AL: "Oncostatin M secreted by skin infiltrating T lymphocytes is a potent keratinocyte activator involved in skin inflammation", JOURNAL OF IMMUNOLOGY, vol. 178, no. 7, 1 April 2007 (2007-04-01), pages 4615 - 4622, XP002794921 *
BONIFACE KDIVEU CMOREL FPEDRETTI NFROGER JRAVON E ET AL.: "Oncostatin M secreted by skin infiltrating T lymphocytes is a potent keratinocyte activator involved in skin inflammation", J IMMUNOL., vol. 178, no. 7, 2007, pages 4615 - 22
BOTELHO FMRANGEL-MORENO JFRITZ DRANDALL TDXING ZRICHARDS CD: "Pulmonary expression of oncostatin M (OSM) promotes inducible BALT formation independently of IL-6, despite a role for IL-6 in OSM-driven pulmonary inflammation", J IMMUNOL., vol. 191, no. 3, 2013, pages 1453 - 64
BRANDT EBSIVAPRASAD U: "Th2 cytokines and atopic dermatitis", J CLIN CELL IMMUNOL, vol. 2, 2011, pages 110
DILLON SRSPRECHER CHAMMOND ABILSBOROUGH JROSENFELD-FRANKLIN MPRESNELL SR ET AL.: "Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice", NAT IMMUNOL., vol. 5, no. 7, 2004, pages 752 - 60, XP002407998, doi:10.1038/ni1084
EDUKULLA RSINGH BJEGGA AGSONTAKE VDILLON SRMADALA SK: "Th2 Cytokines Augment IL-31/IL-3I RA Interactions via STAT6-dependent IL-31 RA Expression", J BIOL CHEM., vol. 290, no. 21, 2015, pages 13510 - 20
EZZAT MHHASAN ZESHAHEEN KY: "Serum measurement of interleukin-31 (IL-31) in paediatric atopic dermatitis: elevated levels correlate with severity scoring", J EUR ACAD DERMATOL VENEREOL., vol. 25, no. 3, March 2011 (2011-03-01), pages 334 - 9
FRITZ DKKERR CBOTELHO FSTAMPFLI MRICHARDS CDONCOSTATIN M: "OSM) primes IL-13-and IL-4-induced eotaxin responses in fibroblasts: regulation of the type-II IL-4 receptor chains IL-4Ralpha and IL-13Ralphal", EXP CELL RES., vol. 315, no. 20, 2009, pages 3486 - 99, XP026766758, doi:10.1016/j.yexcr.2009.09.024
FRITZ DKKERR CFATTOUH RLLOP-GUEVARA AKHAN WIJORDANA M ET AL.: "A mouse model of airway disease: oncostatin M-induced pulmonary eosinophilia, goblet cell hyperplasia, and airway hyperrcsponsiveness are STAT6 dependent, and interstitial pulmonary fibrosis is STAT6 independent", J IMMUNOL., vol. 186, no. 2, 2011, pages 1107 - 18
FRITZ DKKERR CTONG LSMYTH DRICHARDS CD: "Oncostatin-M up-regulates VCAM-1 and synergizes with IL-4 in eotaxin expression: involvement of STAT6", J IMMUNOL., vol. 176, no. 7, 2006, pages 4352 - 60
GITTLER JKSHEMER ASUAREZ-FARINAS M ET AL.: "Progressive activation of T(H)2/T(H)22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis", J ALLERGY CLIN IMMUNOL., vol. 30, 2012, pages 1344 - 54
HEISE RNEIS MMMARQUARDT YJOUSSEN SHEINRICH PCMERK HF ET AL.: "IL-31 receptor alpha expression in epidermal keratinocytes is modulated by cell differentiation and interferon gamma", J INVEST DENNATOL, vol. 129, no. l, 2009, pages 240 - 3
IP WKWONG CKLI MLLI PWCHEUNG PFLAM CW: "Interleukin-31 induces cytokine and chemokine production from human bronchial epithelial cells through activation of mitogen-activated protein kinase signaling pathways: implications for the allergic response", IMMUNOLOGY, vol. 122, no. 4, December 2007 (2007-12-01), pages 532 - 4 1
KABASHIMA KENJI ET AL: "Nemolizumab in patients with moderate-to-severe atopic dermatitis: Randomized, phase II, long-term extension study", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 142, no. 4, 10 May 2018 (2018-05-10), pages 1121, XP085496774, ISSN: 0091-6749, DOI: 10.1016/J.JACI.2018.03.018 *
KANDA NWATANABE S: "Increased serum human (i-defensin-2 levels in atopic dermatitis: relationship to IL-22 and oncostatin M", IMMUNOBIOLOGY, vol. 217, no. 4, April 2012 (2012-04-01), pages 436 - 45, XP028905419, doi:10.1016/j.imbio.2011.10.010
KASRAIE SNIEBUHR MBAUMERT KWERFEL T: "Functional effects of interleukin 31 in human primary keratinocytes", ALLERGY, vol. 66, no. 7, July 2011 (2011-07-01), pages 845 - 52
KO MJPENG YSCHEN HYHSU SPPAI MFYANG JY ET AL.: "Interleukin-31 is associated with uremic pruritus in patients receiving hemodialysis", J AM ACAD DERMATOL., vol. 71, no. 6, December 2014 (2014-12-01), pages 1151 - 1159
LIU WLUO RCHEN YSUN CWANG JZHOU L ET AL.: "Interleukin-31 promotes helper T cell type-2 inflammation in children with allergic rhinitis", PEDIATR RES., vol. 77, no. 1-1, January 2015 (2015-01-01), pages 20 - 8
MAKHLOUGH A: "Topical capsaicin therapy for uremic pruritus in patients on hemodialysis", IRAN J KIDNEY DIS., vol. 4, no. 2, April 2010 (2010-04-01), pages 137 - 40
MOZAFFARIAN ABREWER AWTRUEBLOOD ESLUZINA IGTODD NWATAMAS SP ET AL.: "Mechanisms of oncostatin M-induced pulmonary inflammation and fibrosis", J IMMUNOL., vol. 181, no. 10, 15 November 2008 (2008-11-15), pages 7243 - 53
NEMOTO OFURUE MNAKAGAWA HSHIRAMOTO MHANADA RMATSUKI S ET AL.: "The first trial of CIM331, a humanized antihuman interleukin-31 receptor A antibody, in healthy volunteers and patients with atopic dermatitis to evaluate safety, tolerability and pharmacokinetics of a single dose in a randomized, double-blind, placebo-controlled study", BR J DERMATOL., vol. 174, no. 2, February 2016 (2016-02-01), pages 296 - 304
NIEBUHR MMAMEROW DHERATIZADEH ASATZGER IWERFEL T: "Staphylococcal alpha-toxin induces a higher T cell proliferation and interleukin-31 in atopic dermatitis", INT ARCH ALLERGY IMMUNOL., vol. 156, no. 4, 2011, pages 412 - 5
NOBBE SDZIUNYCZ PMUHLEISEN BBILSBOROUGH JDILLON SRFRENCH LE ET AL.: "IL-31 expression by inflammatory cells is preferentially elevated in atopic dermatitis", ACTA DERM VENEREOL., vol. 92, no. 1, 2012, pages 24 - 8, XP055142730, doi:10.2340/00015555-1191
PISONI RLWIKSTROM BELDER SJAKIZAWA TASANO YKEEN ML ET AL.: "Pruritus in haemodialysis patients: International results from the Dialysis Outcomes and Practice Patterns Study (DOPPS", NEPHROL DIAL TRANSPLANT., vol. 21, no. 12, December 2006 (2006-12-01), pages 3495 - 505
RAAP UWICHMANN KBINDER MSTANDER SWEDI BKAPP A ET AL.: "Correlation of IL-31 serum levels with severity of atopic dermatitis", J ALLERGY CLIN IMMUNOL., vol. 122, no. 2, August 2008 (2008-08-01), pages 421 - 3, XP023518360, doi:10.1016/j.jaci.2008.05.047
RABEONY HPETIT-PARIS IGAMIER JBARRAULT CPEDRETTI NGUILLOTEAU K ET AL.: "Inhibition of keratinocyte differentiation by the synergistic effect of IL-17A, IL-22, IL-1 alpha, TNFalpha and oncostatin M", PLOS ONE, vol. 9, no. 7, 2014, pages e101937
RICHARDS CD: "The enigmatic cytokine oncostatin m and roles in disease", ISRN INFLAMM., vol. 2013, 2013, pages 512103
RING JALOMAR ABIEBER TDCLCURAN MFINK-WAGNER AGELMCTTI C ET AL.: "Guidelines for treatment of atopic eczema (atopic dermatitis) part I", J EUR ACAD DERMATOL VENEREOL., vol. 26, 2012, pages 1045 - 60
RUZICKA THANIFIN JFURUE MPULKA GMLYNARCZYK IWOLLENBERG AGAINS RETOH TMIHARA RYOSHIDA H: "Anti-Interleukin-31 Receptor A Antibody for Atopic Dermatitis", N ENGL J MED, vol. 376, no. 9, 2017, pages 826 - 835, XP055588908, doi:10.1056/NEJMoa1606490
SIDBURY RDAVIS DMCOHEN DECORDORO KMBERGER TGBERGMAN JN ET AL.: "Guidelines of care for the management of atopic dermatitis: section 3. Management and treatment with phototherapy and systemic agents", J AM ACAD DERMATOL, vol. 71, 2014, pages 327 - 49
SILVERBERG JIHANIFIN JM: "Adult eczema prevalence and associations with asthma and other health and demographic factors: a US population-based study", J ALLERGY CLIN IMMUNOL, vol. 132, 2013, pages 1132 - 38
SONKOLV EMULLER ALAUERMA AIPIVARCSI ASOTO HKEMENY L ET AL.: "IL-31: a new link between T cells and pruritus in atopic skin inflammation", J ALLERGY CLIN IMMUNOL, vol. 117, no. 2, February 2006 (2006-02-01), pages 411 - 7, XP005275427, doi:10.1016/j.jaci.2005.10.033
STOTT BLAVENDER PLEHMANN SPENNINO DDURHAM SSCHMIDT-WEBER CB: "Human IL-31 is induced by IL-4 and promotes TH2-driven inflammation", J ALLERGY CLIN IMMUNOL, vol. 132, no. 2, 2013, pages 446 - 54 e5
THOMAS RUZICKA ET AL: "Anti-Interleukin-31 Receptor A Antibody for Atopic Dermatitis", THE NEW ENGLAND JOURNAL OF MEDICINE, - NEJM -, MASSACHUSETTS MEDICAL SOCIETY, US, vol. 376, 1 March 2017 (2017-03-01), pages 826 - 835, XP009195193, ISSN: 0028-4793, DOI: 10.1056/NEJMOA1606490 *
WEIDINGER STEPHAN ET AL: "Atopic dermatitis", NATURE REVIEWS DISEASE PRIMERS, NATURE PUBLISHING GROUP UK, LONDON, vol. 4, no. 1, 21 June 2018 (2018-06-21), pages 1 - 20, XP036530441, DOI: 10.1038/S41572-018-0001-Z *
Z. MIKHAK ET AL: "First-In-Human Study of KPL-716, Anti-Oncostatin M Receptor Beta Monoclonal Antibody, in Healthy Volonteers and Subjects With Atopic Dermatitis", 16 September 2018 (2018-09-16), pages 1 - 18, XP002794922, Retrieved from the Internet <URL:https://investors.kiniksa.com/static-files/ccd0f786-dd59-4cd2-8621-5819c180880a> [retrieved on 20191009] *

Similar Documents

Publication Publication Date Title
Chandran et al. Interleukin-6 blockade with tocilizumab increases Tregs and reduces T effector cytokines in renal graft inflammation: a randomized controlled trial
Hoffman et al. Long-term efficacy and safety profile of rilonacept in the treatment of cryopryin-associated periodic syndromes: results of a 72-week open-label extension study
AU2015222951B2 (en) Methods for treating skin infection by administering an IL-4R antagonist
US11795216B2 (en) Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
WO2020234834A1 (fr) Méthode de traitement de maladie intestinale inflammatoire au moyen d&#39;une polythérapie d&#39;anticorps dirigés contre il -23 et tnf alpha
JP2016527324A (ja) ベンラリツマブを使用して喘息の増悪率を低減する方法
TW202037604A (zh) 治療掌蹠膿疱症之抗il-36r抗體
JP2020100653A (ja) ベンラリツマブによる喘息症状の改善方法
JP2022126791A (ja) Il-17アンタゴニストを用いて初発プラーク型乾癬を治療する方法
US20240101690A1 (en) TREATMENT OF SKIN DISEASES OR DISORDERS BY DELIVERY OF ANTI-OSMRBeta ANTIBODY
JP2021147383A (ja) アトピー性皮膚炎及び関連障害を処置するための方法
WO2020036833A1 (fr) TRAITEMENT DE MALADIES OU DE TROUBLES DE LA PEAU PAR ADMINISTRATION D&#39;UN ANTICORPS ANTI-OSMRβ
AU2018361975A1 (en) Method of treating tendinopathy using interleukin-17 (IL-17) antagonists
JP7495403B2 (ja) 巨細胞性動脈炎の治療
JP2024522620A (ja) アトピー性皮膚炎及び関連する障害の治療方法
KR20240049348A (ko) 항-인터루킨-33 항체를 사용한 만성 폐쇄성 폐질환의 치료
WO2020096664A1 (fr) Traitement de l&#39;artérite à cellules géantes
TW202214692A (zh) 治療患有鼻瘜肉的患者的重度氣喘之方法
KR20240090993A (ko) 류마티스성 관절염의 치료에서 또는 진통제로서 사용하기 위한 gm-csf 중화 항체

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: 19759797

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19759797

Country of ref document: EP

Kind code of ref document: A1