WO2021250533A1 - Methods of treatment using omalizumab or ligelizumab - Google Patents

Methods of treatment using omalizumab or ligelizumab Download PDF

Info

Publication number
WO2021250533A1
WO2021250533A1 PCT/IB2021/054956 IB2021054956W WO2021250533A1 WO 2021250533 A1 WO2021250533 A1 WO 2021250533A1 IB 2021054956 W IB2021054956 W IB 2021054956W WO 2021250533 A1 WO2021250533 A1 WO 2021250533A1
Authority
WO
WIPO (PCT)
Prior art keywords
mir
subject
binding fragment
disease
lge antibody
Prior art date
Application number
PCT/IB2021/054956
Other languages
French (fr)
Inventor
Volkan MANGA
William R. HENDERSON Jr.
Theodor K. BAMMLER
James W. MACDONALD
Taha AL-SHAIKHLY
Original Assignee
Novartis Ag
University Of Washington
Genentech, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag, University Of Washington, Genentech, Inc. filed Critical Novartis Ag
Publication of WO2021250533A1 publication Critical patent/WO2021250533A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE

Definitions

  • the present disclosure relates to methods of preventing or treating diseases or disorders in subjects in need of treatment using an anti-lgE antibody, e.g., omalizumab or ligelizumab.
  • an anti-lgE antibody e.g., omalizumab or ligelizumab.
  • Immunoglobulin E is an antibody associated with hypersensitivity and allergic reactions. IgE mainly binds on the high-affinity IgE receptor (FcsRI) on mast cells, basophils and dendritic cells and hence decreases the induction of the regulatory T cells.
  • FcsRI high-affinity IgE receptor
  • Xolair ® (omalizumab) is a recombinant DNA-derived humanized monoclonal antibody that selectively binds to free, circulating human immunoglobulin E (IgE) thus inhibiting IgE binding to IgE receptors on the surface of mast cells and basophils resulting in decreased release of allergic mediators.
  • IgE immunoglobulin E
  • omalizumab By binding to free, circulating IgE, omalizumab also lowers serum free IgE levels and down-regulates the number of IgE receptors on the surface of mast cells and basophils.
  • Omalizumab widely used for the treatment of Allergic asthma, allergic rhinitis and chronic spontaneous urticaria (CSU).
  • Ligelizumab is a humanized monoclonal antibody with higher affinity binding to human immunoglobulin E (IgE) than omalizumab.
  • IgE immunoglobulin E
  • ligelizumab Upon binding, ligelizumab is able to block the interaction of IgE with both the high and low affinity IgE receptors (FceRI and FceRII).
  • FceRI and FceRII high and low affinity IgE receptors
  • IgE is necessary for the enhanced expression of the FceRI seen in atopic subjects, and a decrease in FceRI expression on circulating basophils accompanies ligelizumab treatment.
  • IgE production decreased B cell numbers and reduced cytokine production by T cells.
  • This mechanism confers benefit to subjects with chronic spontaneous urticaria (CSU) by preventing the itchy hives and angioedema that are associated with degranulation (histamine release) of mast cells and basophils.
  • CSU chronic spontaneous urticaria
  • Ligelizumab demonstrated dose- and time-dependent suppression of free IgE, basophil FcsRI, basophil surface IgE, and skin prick test responses to allergen, superior in extent and duration to those observed with omalizumab.
  • Superior affinity and pharmacodynamic (PD) outcomes of ligelizumab compared to omalizumab may translate into superior posology and superior clinical efficacy in subjects with CSU.
  • MicroRNAs small non-coding RNAs, are released by cells into the blood and other biofluids within small ( ⁇ 150 nm in diameter), stable membrane-bound microvesicles(termed exosomes), making them accessible and attractive biomarkers of disease pathogenesis and response to therapeutic intervention.
  • IgE-mediated disease or disorder e.g. Fc RI-mediated pathophysiologies.
  • diseases or disorders are selected from the group consisting of: asthma (e.g. allergic asthma, moderate to severe persistent allergic asthma), Atopic dermatitis, Bullous Pemphigoid, uricaria, (e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria), Chronic Inducible Urticarias (CINDUs), rhinitis (e.g.
  • Seasonal allergic rhinitis such as e.g. Cedar pollinosis), Nasal polyposis, IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
  • an anti-lgE antibody e.g. an anti-lgE antibody that selectively binds to free, circulating human IgE (e.g., omalizumab or ligelizumab), to a subject in need thereof.
  • an anti-lgE antibody e.g. an anti-lgE antibody that selectively binds to free, circulating human IgE (e.g., omalizumab or ligelizumab)
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has low expression of miR-141-3p, in exosomes.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least one miRNA selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least two miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least three miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least four miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least five miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • A7 An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR- 3976, and optionally low expression of miR-141-3p, in exosomes.
  • A8. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a basophil level of from about >19 basophils/pL to about 360 basophils/pL
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a basophil level of about >20 basophils/pL.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a basophil level of about >21 basophils/pL
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
  • An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
  • the anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR-4664-3p in basophils.
  • A14 The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, wherein the subject has a basophil level of from about >19 basophils/pL to about 360 basophils/pL, or of about 320 basophils/pL, or of about 321 basophils/pL.
  • A15 The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, or according to embodiment A14, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
  • A16 The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, or according to embodiment A14, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
  • A17 The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, or according to embodiment A14, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR-4664-3p in basophils.
  • A18 The anti-lgE antibody or antigen binding fragment thereof according to any one of embodiments A1-A17, comprising administering to the subject in need thereof a therapeutically effective amount of an anti-lgE antibody or antigen binding fragment thereof.
  • asthma e.g. allergic asthma, moderate to severe persistent allergic asthma
  • Atopic dermatitis Bullous Pemphigoid
  • uricaria e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria
  • CINDUs Chronic Inducible Urticarias
  • rhinitis e.g. Seasonal allergic rhinitis, such as e.g. Cedar pollinosis
  • Nasal polyposis IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
  • A20 The anti-lgE antibody or antigen binding fragment thereof according to any of the above embodiments, wherein the disease or condition is selected from allergic asthma, chronic spontaneous urticaria, allergic rhinitis, food allergy.
  • A21 The anti-lgE antibody or antigen binding fragment thereof according to any of the above embodiments, wherein the anti-lgE antibody is omalizumab or ligelizumab.
  • A22 The anti-lgE antibody or antigen binding fragment thereof according to embodiment A21 , wherein the anti-lgE antibody is omalizumab.
  • A23 The anti-lgE antibody or antigen binding fragment thereof according to embodiment A22, wherein omalizumab is administered at a dose of about 75 mg to about 600 mg, e g. at a maximum dose of 600 mg.
  • A25 The anti-lgE antibody or antigen binding fragment thereof according to embodiment A24, wherein ligelizumab is administered at a dose of about 24 mg to about 600 mg, e g. at a maximum dose of 600 mg.
  • A26 The anti-lgE antibody or antigen binding fragment thereof according to embodiment A22 or embodiment A23, wherein omalizumab is administered every two to four weeks.
  • A28 The anti-lgE antibody or antigen binding fragment thereof according to any one of embodiments A21 to A27, wherein omalizumab or ligelizumab, is administered during up to 16 weeks, e.g. 12 to 16 weeks.
  • A29 The anti-lgE antibody or antigen binding fragment thereof according to any of the above embodiments, wherein the anti-lgE antibody is co-administered with a corticosteroid and/or an immunusuppressor, or a long-acting beta agonist (LABA).
  • LAA long-acting beta agonist
  • a method of treating or preventing a disease or disorder involving IgE in a subject in need thereof comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least one miRNA selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • a method of treating or preventing a disease or disorder involving IgE in a subject in need thereof comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least two miRNAs selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • a method of treating or preventing a disease or disorder involving IgE in a subject in need thereof comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least three miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • a method of treating or preventing a disease or disorder involving IgE in a subject in need thereof comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least four miRNAs selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • a method of treating or preventing a disease or disorder involving IgE in a subject in need thereof comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least five miRNAs selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • an anti-lgE antibody or antigen binding fragment comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2- 3p, miR-6877-3p, and miR-3976, and optionally low expression of miR-141-3p, in exosomes.
  • a method of identifying subjects responders to a therapy with an anti-lgE antibody comprising (1) measuring the expression level of one or more miR-1200, miR-1236-3p and miR-4664-3p in basophils, in a sample from said subject, and (2) determining the responders based on the expression level of such miRNAs. C2.
  • a method of identifying subjects responders to a therapy with an anti-lgE antibody comprising (1) measuring the expression level of one or more miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, miR-3976, and miR-141-3p in exosomes, in a sample from said subject, and (2) determining the responders based on the expression level of such miRNAs.
  • a method of screening a patient to be treated with a therapy comprising an anti-lgE antibody or antigen binding fragment comprising measuring the expression level of one or more miR-1200, miR-1236-3p and miR-4664-3p in basophils, in a sample from a patient in need of such a therapy, wherein an high level of such miRNA indicates responder to such anti-lgE antibody or antigen binding fragment.
  • a method of screening a patient to be treated with a therapy comprising an anti-lgE antibody or antigen binding fragment comprising measuring the expression level of one or more miR-6499- 5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976, in a sample from a patient in need of such therapy, wherein an high level of such miRNA indicates responder to such anti-lgE antibody or antigen binding fragment.
  • a method of miRNAs in patient screening comprising measuring the expression level of miR- 141 -3p in exosomes, in a sample from a subject in need of an anti-lgE antibody or antigen binding fragment, wherein an low level of such miRNA indicates responder to such anti-lgE antibody or antigen binding fragment.
  • miRNAs to identify subjects in need of therapy with an anti-lgE antibody or antigen binding fragment, comprising (1 ) measuring the expression level of one or more miR-1200, miR- 1236-3p and miR-4664-3p in a sample from said subject, and (2) selecting a subject having a high level expression of said miRNA.
  • miRNAs to identify subjects in need of therapy with an anti-lgE antibody or antigen binding fragment, comprising (1) measuring the expression level of one or more miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976, in a sample from said subject, and (2) selecting a subject having a high level expression of said miRNA.
  • miRNAs to identify subjects in need of therapy with an anti-lgE antibody or antigen binding fragment, comprising (1) measuring the expression level of miR-141-3p in exosomes, in a sample from a subject in need of an anti-lgE antibody or antigen binding fragment, and (2) selecting a subject having a low level expression of said miRNA.
  • miRNAs to select a subject responder to a therapy with an anti-lgE antibody or antigen binding fragment, comprising selecting a subject having a high expression level of one or more miR-1200, miR-1236-3p, miR-4664-3p miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976.
  • miRNAs to select a subject responder to a therapy with an anti-lgE antibody or antigen binding fragment, comprising selecting a subject having a low expression level of miR- 141 -3p in exosomes.
  • miRNAs selected from the list: miR-1200, miR-1236-3p, miR-4664-3p, miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, miR-3976, and miR-141- 3p as a biomarker for treatment response to therapy using anti-lgE antibody or antigen binding fragment.
  • basophil levels as a biomarker for treatment response to therapy using anti-lgE antibody or antigen binding fragment, wherein the basophil level is from about >19 basophils/pL to about 360 basophils/pL, or of about 320 basophils/pL, or of about 321 basophils/pL.
  • a biomarker comprising one or more miRNAs selected from the list: miR-1200, miR-1236- 3p, miR-4664-3p, miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, miR- 3976, and miR-141-3p.
  • kits for treating particular subjects having disease or disorder involving IgE comprising an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, (e.g., in liquid or lyophilized form) or a pharmaceutical composition comprising the anti-lgE antibody as described herein, and a biomarker according to any of the above embodiments.
  • an anti-lgE antibody e.g., omalizumab or ligelizumab
  • antigen-binding fragment thereof e.g., in liquid or lyophilized form
  • a pharmaceutical composition comprising the anti-lgE antibody as described herein, and a biomarker according to any of the above embodiments.
  • high expression miRNA is referring to a difference between expression of such an miRNA between a non-responder and a complete responder of at least 3 logCPM, at least 3 logCPM, at least 4 logCPM, wherein the expression of the miRNA is higher in the responder than in the non-responder.
  • low expression miRNA is referring to a difference between expression of such an miRNA between a complete responder and a non-responder of at least 3 logCPM, at least 3 logCPM, at least 4 logCPM, wherein the expression of the miRNA is lower in the responder than in the non-responder.
  • IgE refers to Immunoglobulin E.
  • composition “comprising” encompasses “including” as well as “consisting,” e.g., a composition “comprising” X may consist exclusively of X or may include something additional, e.g., X + Y.
  • antibody as referred to herein includes naturally-occurring and whole antibodies.
  • a naturally-occurring “antibody” is a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed hypervariable regions or complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
  • antigen-binding fragment of an antibody, as used herein, refers to fragments of an antibody that retain the ability to specifically bind to IgE. It has been shown that the antigenbinding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding portion" of an antibody include a Fab fragment, a monovalent fragment consisting of the V L , V , CL and CH1 domains; a F(ab) 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CH1 domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a dAb fragment (Ward et al. , 1989 Nature 341 :544-546), which consists of a V domain; and an isolated CDR.
  • Fab fragment a monovalent fragment consisting of the V L , V , CL and CH1 domains
  • F(ab) 2 fragment a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region
  • a Fd fragment consisting of the VH and CH1 domains
  • the two domains of the Fv fragment, V L and V H are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al., 1988 Science 242:423-426; and Huston et al., 1988 Proc. Natl. Acad. Sci. 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antibody”.
  • Single chain antibodies and antigen-binding portions are obtained using conventional techniques known to those of skill in the art.
  • K D is intended to refer to the dissociation rate of a particular antibody-antigen interaction.
  • K D is intended to refer to the dissociation constant, which is obtained from the ratio of K d to K a (i.e., K d /K a ) and is expressed as a molar concentration (M).
  • K D values for antibodies can be determined using methods well established in the art. A preferred method for determining the KD of an antibody is by using surface plasmon resonance, or using a biosensor system such as a Biacore® system.
  • the anti-lgE antibody or antigen-binding fragment thereof according to the invention binds human IgE with a K D of about 0.02 to 7.7 nM, e.g. 100-250 pM.
  • affinity refers to the strength of interaction between antibody and antigen at single antigenic sites. Within each antigenic site, the variable region of the antibody “arm” interacts through weak non-covalent forces with antigen at numerous sites; the more interactions, the stronger the affinity.
  • Standard assays to evaluate the binding affinity of the antibodies toward IgE of various species are known in the art, including for example, ELISAs, western blots and RIAs.
  • the binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore analysis.
  • derivative is used to define amino acid sequence variants, and covalent modifications (e.g. pegylation, deamidation, hydroxylation, phosphorylation, methylation, etc.) of an anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab, according to the present disclosure, e.g., of a specified sequence (e.g., a variable domain).
  • a “functional derivative” includes a molecule having a qualitative biological activity in common with the disclosed anti-lgE antibodies.
  • a functional derivative includes fragments and peptide analogs of an anti-lgE antibody as disclosed herein. Fragments comprise regions within the sequence of a polypeptide according to the present disclosure, e.g., of a specified sequence.
  • substantially identical means that the relevant amino acid or nucleotide sequence (e.g., VH or VL domain) will be identical to or have insubstantial differences (e.g., through conserved amino acid substitutions) in comparison to a particular reference sequence. Insubstantial differences include minor amino acid changes, such as 1 or 2 substitutions in a 5 amino acid sequence of a specified region (e.g., V H or V L domain).
  • the second antibody has the same specificity and has at least 50% of the affinity of the same. Sequences substantially identical (e.g., at least about 85% sequence identity) to the sequences disclosed herein are also part of this application.
  • sequence identity of a derivative anti-lgE antibody can be about 90% or greater, e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher relative to the disclosed sequences.
  • Identity with respect to a native polypeptide and its functional derivative is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity, and not considering any conservative substitutions as part of the sequence identity. Neither N- or C-terminal extensions nor insertions shall be construed as reducing identity. Methods and computer programs for the alignment are well known. The percent identity can be determined by standard alignment algorithms, for example, the Basic Local Alignment Search Tool (BLAST) described by Altshul et al. ((1990) J. Mol. Biol., 215: 403410); the algorithm of Needleman et al.
  • BLAST Basic Local Alignment Search Tool
  • a set of parameters may be the Blosum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percent identity between two amino acid or nucleotide sequences can also be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM 120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • amino acid(s) refer to all naturally occurring L-a-amino acids, e.g., and include D-amino acids.
  • amino acid sequence variant refers to molecules with some differences in their amino acid sequences as compared to the sequences according to the present disclosure. Amino acid sequence variants of an antibody according to the present disclosure, e.g., of a specified sequence, still have the ability to bind the IgE.
  • Amino acid sequence variants include substitutional variants (those that have at least one amino acid residue removed and a different amino acid inserted in its place at the same position in a polypeptide according to the present disclosure), insertional variants (those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a polypeptide according to the present disclosure) and deletional variants (those with one or more amino acids removed in a polypeptide according to the present disclosure).
  • pharmaceutically acceptable means a nontoxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s).
  • administering in relation to a compound, e.g., an anti-lgE antibody, is used to refer to delivery of that compound to a subject by any route.
  • a “therapeutically effective amount” refers to an amount of anti-lgE antibody (e.g., omalizumab or an antigen-binding fragment thereof) that is effective, upon single or multiple dose administration to a subject (such as a human) for treating, preventing, preventing the onset of, curing (if applicable), delaying, reducing the severity of, ameliorating at least one symptom of a disorder or recurring disorder, or prolonging the survival of the subject beyond that expected in the absence of such treatment.
  • an individual active ingredient e.g., an anti-lgE antibody, e.g., omalizumab
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • treatment or “treat” is herein defined as the application or administration of an anti-lgE antibody according to the disclosure, for example, omalizumab, or a pharmaceutical composition comprising said anti-lgE antibody, to a subject or to an isolated tissue or cell line from a subject, where the subject has a particular disease, a symptom associated with the disease, or a predisposition towards development of the disease, where the purpose is to cure (if applicable), delay the onset of, reduce the severity of, alleviate, ameliorate one or more symptoms of the disease, improve the disease, reduce or improve any associated symptoms of the disease or the predisposition toward the development of the disease.
  • treatment or “treat” includes treating a subject suspected to have the disease as well as subjects who are ill or who have been diagnosed as suffering from the disease or medical condition, and includes suppression of clinical relapse.
  • treatment or “treat” is herein defined as the application or administration of an IgE antibody according to the disclosure, for example ligelizumab, or a pharmaceutical composition comprising said anti- IgE antibody, to a subject or to an isolated tissue or cell line from a subject, where the subject has a particular disease (e.g., CSU), a symptom associated with the disease (e.g., CSU), or a predisposition towards development of the disease (e.g., CSU) (if applicable), where the purpose is to cure (if applicable), delay the onset of, reduce the severity of, alleviate, ameliorate one or more symptoms of the disease, improve the disease, reduce or improve any associated symptoms of the disease or the predisposition toward the development of the disease.
  • treatment or “treat” includes treating a subject suspected to have the disease as well as subjects who are ill or who have been diagnosed as suffering from the disease or medical condition, and includes suppression of clinical relapse.
  • the phrase “population of subjects” is used to mean a group of subjects.
  • the IgE antagonist e.g., IgE antibody, such as ligelizumab
  • IgE antibody such as ligelizumab
  • basophil level refers to the number of basophils per ml blood, as determined according to the known methods in the art.
  • the term “diseases or disorders involving IgE” refers to IgE-mediated disease or disorder, e.g. FcsRI-mediated pathophysiologies e.g. to disease or disorder selected from the group consisting of: asthma (e.g. allergic asthma, moderate to severe persistent allergic asthma), Atopic dermatitis, Bullous Pemphigoid, uricaria, (e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria), Chronic Inducible Urticarias (CINDUs), rhinitis (e.g. Seasonal allergic rhinitis, such as e.g. Cedar pollinosis), Nasal polyposis, IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
  • asthma e.g. allergic asthma, moderate to severe persistent allergic asthma
  • Atopic dermatitis e.g. chronic spontaneous urticaria (CSU),
  • the term “high expression” of a miRNA refers to an expression level above the geometric mean expression level for that microRNA, e.g. having a log counts/million (logCPM) above zero, e.g. above 1 , e.g. above 2, e.g. around or above 4.
  • logCPM log counts/million
  • the term “low expression” of a miRNA refers to an expression level below the geometric mean expression level for that microRNA, e.g. having a log counts/million (logCPM) below zero, e.g. 1 logCPM below zero, e.g. 2 or more logCPM below zero.
  • logCPM log counts/million
  • log counts/million may be measured by using the voomWithQualityWeights function (Liu R, Holik AZ, Su S, et al., Nucleic Acids Res 2015;43:e97) from the Bioconductor limma package (Ritchie ME, Phipson B, Wu D, et al., Nucleic Acids Res 2015;43:e47).
  • Responders are defined as those subjects with an AUC for the weekly UAS7 over the 12-week treatment period that was equal to or less than the 75th percentile and achieved 90% improvement of baseline UAS7.
  • Non-responders are defined as subjects who a) failed to achieve a 90% improvement of their baseline weekly UAS7, or b) had an AUC for the weekly UAS7 over the 12-week study period that was above the 75th percentile (i.e. , top quartile), or c) had an exacerbation of their symptoms necessitating the addition of an anti-inflammatory or an immunosuppressive agent.
  • Non-responders to therapy using an anti-lgE antibody or antigen-binding fragment thereof are defined as subjects who failed to achieve a 90% improvement of their baseline or had an exacerbation of their symptoms. Responders to therapy using an anti-lgE antibody or antigenbinding fragment thereof are defined as those subjects who achieved 90% improvement of baseline.
  • the anti-lgE antibody or antigen-binding fragment thereof is a monoclonal antibody. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is a human or humanized antibody. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is a humanized antibody. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is a human antibody of the lgGi subtype. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is omalizumab. In other embodiments, the anti-lgE antibody or antigen-binding fragment thereof is ligelizumab.
  • anti-lgE antibodies include, but are not limited to, omalizumab, quilizumab, ligelizumab and etrolizumab.
  • an anti-lgE antibody or antigen-binding fragment thereof used in the disclosed methods may be an amino acid sequence variant of the reference anti-lgE antibodies set forth herein.
  • the disclosure also includes anti-lgE antibodies or antigen-binding fragments thereof (e.g., omalizumab) in which one or more of the amino acid residues of the VH or VL domain of omalizumab, typically only a few (e.g. 1-10), are changed; for instance by mutation, e.g., site directed mutagenesis of the corresponding DNA sequences.
  • omalizumab antigen-binding fragments thereof in which one or more of the amino acid residues of the VH or VL domain of omalizumab, typically only a few (e.g. 1-10), are changed; for instance by mutation, e.g., site directed mutagenesis of the corresponding DNA sequences.
  • the disclosed anti-lgE antibody e.g., omalizumab or ligelizumab
  • antigen-binding fragment thereof may be used in vitro, ex vivo, or incorporated into pharmaceutical compositions and administered in vivo to treat subjects (e.g., human subjects) affected by one or more disease or disorder involving IgE.
  • the anti-lgE antibody e.g., omalizumab or ligelizumab
  • antigen-binding fragment thereof may be used as a pharmaceutical composition when combined with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier may contain, in addition to the anti-lgE antibody or antigen-binding fragment thereof, carriers, various diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art.
  • the characteristics of the carrier will depend on the route of administration.
  • compositions for use in the disclosed methods may be manufactured in conventional manner.
  • the pharmaceutical composition is provided in lyophilized form.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • Other formulations comprise liquid or lyophilized formulation.
  • Antibodies e.g., antibodies to IgE, or antigen-binding fragment thereof, are typically formulated either in aqueous form ready for parenteral administration or as lyophilisates for reconstitution with a suitable diluent prior to administration.
  • the anti-lgE antibody or antigen-binding fragment thereof e.g., omalizumab
  • Suitable lyophilisate formulations can be reconstituted in a small liquid volume (e.g., 2ml or less, e.g., 1 ml) to allow subcutaneous administration and can provide solutions with low levels of antibody aggregation. Techniques for purification of antibodies to a pharmaceutical grade are well known in the art.
  • diseases or disorders involving IgE are selected from the group consisting of.
  • the disease or disorder involving IgE refers to food allergies, e.g. peanuts allergy.
  • the disease or disorder involving IgE refers to diseases or disorders selected from Allergic asthma, allergic rhinitis and chronic spontaneous urticaria (CSU).
  • the disease or disorder involving IgE refers to chronic spontaneous urticaria (CSU).
  • CSU chronic spontaneous urticaria
  • the disease or disorder involving IgE refers to Allergic asthma.
  • anti-lgE antibodies e.g omalizumab or ligelizumab
  • methods of, and anti-lgE antibodies for use in, preventing, treating or modifying the course of a disease or disorder involving IgE in a subject in need thereof, comprising administering the subject a therapeutically effective amount of anti-lgE antibody or antigen-binding fragment thereof.
  • the subject in need thereof may have an insulin allergy.
  • the subject is affected by a disease or disorder selected from allergy, asthma, urticarial and rhinitis, e.g. a disease or condition selected from allergic asthma, CSU and allergic rhinitis.
  • a disease or disorder selected from allergy, asthma, urticarial and rhinitis e.g. a disease or condition selected from allergic asthma, CSU and allergic rhinitis.
  • the subject is affected by asthma, e.g. allergic asthma.
  • the subject is affected by urticaria, e.g. chronic spontaneous urticaria
  • the subject is affected by rhinitis, e.g. allergic rhinitis.
  • the subject is affected by food allergies, e.g. peanuts allergy.
  • Ligelizumab demonstrated dose- and time-dependent suppression of free IgE, basophil FcsRI, basophil surface IgE, and skin prick test responses to allergen, superior in extent and duration to those observed with omalizumab. Superior affinity and pharmacodynamic (PD) outcomes of ligelizumab compared to omalizumab may translate into superior posology and superior clinical efficacy in subjects.
  • Lieglizumab binds to IgE with 150fold higher affinity than omalizumab (A. Eggel, Molecular, Structural And Mechanistic Insight Into Ligelizumab Mediated Suppression Of IgE Dependent Allergic Responses, EAACI 2019).
  • anti-lgE antibodies or antigen-binding fragments thereof for use in, preventing, treating or modifying the course of a disease or disorder involving IgE in a subject in need thereof, comprising administering the subject a therapeutically effective amount of anti-lgE antibody or antigen-binding fragment thereof, wherein the subject is not affected by allergy, asthma, urticarial or rhinitis.
  • the subject in need thereof has allergy.
  • the subject is affected by asthma, e.g. allergic asthma.
  • the subject is affected by urticaria, e.g. chronic spontaneous urticaria
  • the subject is affected by rhinitis, e.g. allergic rhinitis.
  • the appropriate dosage will vary depending upon, for example, the particular anti-lgE antibody to be employed, the host, the mode of administration and the nature and severity of the condition being treated, and on the nature of prior treatments that the subject has undergone. It may also depend on the level of IgE in the subject’s blood before initiating the treatment with the anti-lgE antibody.
  • the attending health care provider will decide the amount of the anti-lgE antibody with which to treat each individual subject.
  • the attending health care provider may administer low doses of the anti-lgE antibody and observe the subject’s response, in particular the blood level of IgE.
  • the usual dose range of omalizumab is between 75 mg and 600 mg in one to four subcutaneously injections, and the maximum recommended dose is 600 mg.
  • the dosing of omalizumab for treating asthma is determined based on the subject’s weight and the subject’s serum total IgE level.
  • the dosage of omalizumab for chronic urticaria indication is 300 mg sc per month.
  • omalizumab is administered subcutaneously at a dose of about 75mg to about 600 mg, e.g. at a dose of about 300 mg, e.g. at a maximum dose of 600 mg.
  • ligelizumab is administered subcutaneously at a dose of about 24 mg to about 600 mg, at a dose of about 24 mg to about 240 mg, e.g. at a dose of about 24 mg, of about 72 mg, of about 120 mg, or of about 240 mg, e.g. at a maximum dose of 600 mg.
  • the level of IgE in the subject’s blood is measured before initiating the administration of the anti-lgE antibody, e.g., omalizumab or ligelizumab, and the dose of the antibody is adjusted based on the weight of the subject and/or his serum total IgE level.
  • the anti-lgE antibody e.g., omalizumab or ligelizumab
  • the duration of therapy using a pharmaceutical composition of the present disclosure will vary, depending on the severity of the disease or disorder to be treated and the condition and personal response of each individual subject.
  • the subject is administered the anti-lgE antibody (e.g., omalizumab or ligelizumab) for long-term, e.g. at least 12 weeks, e.g. up to 16 weeks, e.g. to 12 to 16 weeks.
  • the anti-lgE antibody e.g., omalizumab or ligelizumab
  • the anti-lgE antagonist e.g., omalizumab or ligelizumab
  • the anti-lgE antagonist is administered to the subject every two weeks, e.g. every two or four weeks, e.g. monthly.
  • the anti-lgE antibody or antigen-binding fragment thereof according to the present disclosure is conveniently administered parenterally, e.g., intravenously, intramuscularly, or subcutaneously, e.g. subcutaneously.
  • the anti-lgE antibody or antigen-binding fragment thereof may be administered to the subject subcutaneously (SC), e.g. at about 75 mg to about 600 mg (e.g. about 75 mg, about 600 mg), e.g. at about 300 mg.
  • SC subcutaneously
  • the anti-lgE antibody or antigen-binding fragment thereof may be administered to the subject subcutaneously (SC), e.g. at about 24 mg to about 600 mg (e.g. about 24 mg, about 72 mg, about 120 mg, about 240 mg, or about 600 mg), e.g. at about 120 mg.
  • SC subcutaneously
  • a therapeutically effective amount of an anti-lgE antibody e.g., omalizumab or ligelizumab
  • a subject e.g., a mammal (e.g., a human).
  • anti-lgE antibody e.g., omalizumab or ligelizumab
  • antigen-binding fragment thereof this does not preclude that, if the subject is to be ultimately treated with an anti- lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, therapy is necessarily a monotherapy.
  • the anti-lgE antibody e.g., omalizumab or ligelizumab
  • the anti-lgE antibody may be administered in accordance with the methods of the disclosure either alone or in combination with other agents and therapies for treating the subject affected by the disease or disorder involving IgE, e.g., in combination with at least one additional therapeutic agent, such as e.g., a corticosteroid or an immumosuppressor, e.g., a systemic corticosteroid or an immunosuppressor.
  • the subject to be treated is allergic, or when the subject is also affected by another disease or disorder selected from asthma, urticaria, and rhinitis, e.g. selected from allergic asthma, CSU, and allergic rhinitis.
  • another disease or disorder selected from asthma, urticaria, and rhinitis, e.g. selected from allergic asthma, CSU, and allergic rhinitis.
  • the anti-lgE antibody or antigen-binding fragment thereof may be administered either simultaneously with the other agent, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering the anti-lgE antibody or antigen-binding fragment thereof in combination with other agents and the appropriate dosages for co-delivery.
  • Various therapies may be beneficially combined with the disclosed anti-lgE antibodies, such as omalizumab or ligelizumab, during treatment of the disease or disorder involving IgE, disclosed herein.
  • Such therapies include for example corticosteroids (e.g., systemic corticosteroids) or immunosuppressors.
  • an anti-lgE antibody e.g., omalizumab
  • antibodies as the active ingredient of pharmaceuticals is now widespread, including the products HERCEPTINTM (trastuzumab), RITUXANTM (rituximab), SYNAGISTM (palivizumab), etc.
  • Techniques for purification of antibodies to a pharmaceutical grade are known in the art.
  • an IgE antagonist e.g., IgE binding molecules (e.g., IgE antibody or antigen-binding fragment thereof, e.g., ligelizumab) or IgE receptor binding molecules (e.g., IgE antibody or antigen-binding fragment thereof)
  • the IgE antagonist will be in the form of a pyrogen-free, parenterally acceptable solution.
  • a pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection may contain, in addition to the IgE antagonist, an isotonic vehicle such as sodium chloride, Ringer's solution, dextrose, dextrose and sodium chloride, lactated Ringer's solution, or other vehicle as known in the art.
  • an isotonic vehicle such as sodium chloride, Ringer's solution, dextrose, dextrose and sodium chloride, lactated Ringer's solution, or other vehicle as known in the art.
  • an IgE antagonist e.g., IgE binding molecule (e.g., IgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab) or IgE receptor binding molecule (e.g., IgE antibody or antigen-binding fragment thereof) is administered to a subject, e.g., a mammal (e.g., a human).
  • a mammal e.g., a human
  • an IgE antagonist e.g., omalizumab or ligelizumab
  • the IgE antagonist may be administered in accordance with the methods of the disclosure either alone or in combination with other agents and therapies for treating CSU subjects, e.g., in combination with at least one additional CSU agent.
  • an IgE antagonist When co-administered with one or more additional CSU agent(s), an IgE antagonist may be administered either simultaneously with the other agent, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering the IgE antagonist in combination with other agents and the appropriate dosages for co-delivery.
  • an anti-lgE antibody e.g. ligelizumab
  • the subject has allergy, asthma and/or urticarial, e.g., has a disease or disorder selected from asthma, allergic asthma, rhinitis, allergic rhinitis, urticarial and CSU.
  • the subject has allergic asthma, allergic urticaria and/or CSU.
  • the subject has allergic asthma and CSU.
  • the anti-lgE antibody e.g., omalizumab or ligelizumab
  • antigen-binding fragment thereof can be prescribed as first treatment or added on to any of the standard of care medications.
  • the timing of dosing is generally measured from the day of the first dose of anti-lgE antibody, e.g. ligelizumab or omalizumab, (which is also known as “baseline”).
  • anti-lgE antibody e.g. ligelizumab or omalizumab
  • baseline which is also known as “baseline”.
  • health care providers often use different naming conventions to identify dosing schedules, as shown in
  • Table 1 Common naming conventions for dosing regimens. Bolded items refer to the naming convention used herein.
  • week zero may be referred to as week one by some health care providers, while day zero may be referred to as day one by some health care providers.
  • day zero may be referred to as day one by some health care providers.
  • different physicians will designate, e.g., a dose as being given during week 4 / on day 28, during week 4 / on day 29, during week 4 / on day 28, during week 4 / on day 29, while referring to the same dosing schedule.
  • the first week of dosing will be referred to herein as week 0, while the first day of dosing will be referred to as day 1 .
  • this naming convention is simply used for consistency and should not be construed as limiting, i.e. , weekly dosing is the provision of a weekly dose of the IgE antibody regardless of whether the physician refers to a particular week as “week 0” or “week 1”.
  • the antibody is administered during week 0, 4, 8, 12, 16, 20, etc.
  • Some providers may refer to this regimen as monthly dosing (or dosing every 4 weeks). It will be appreciated by a skilled artisan that administering a patient an injection at weeks 0 followed by once monthly dosing starting at week 4 is the same as: 1) administering the patient an injection at weeks 0 and 4, followed by once monthly dosing starting at week 8; 2) administering the patient an injection at weeks 0 and 4 followed by dosing every 4 weeks; and 3) administering the patient an injection at weeks 0 and 4 followed by monthly administration.
  • kits for treating particular subjects having disease or disorder involving IgE comprise an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, (e.g., in liquid or lyophilized form) or a pharmaceutical composition comprising the anti-lgE antibody (described supra). Additionally, such kits may comprise means for administering the anti-lgE antibody or antigen-binding fragment thereof (e.g., an auto-injector, a syringe and vial, a prefilled syringe, a prefilled pen) and instructions for use.
  • an anti-lgE antibody e.g., omalizumab or ligelizumab
  • antigen-binding fragment thereof e.g., in liquid or lyophilized form
  • a pharmaceutical composition comprising the anti-lgE antibody (described supra).
  • kits may comprise means for administering the anti-lgE antibody or antigen-binding fragment thereof
  • kits may contain additional therapeutic agents (described supra) for treating the disease or disorder involving IgE, e.g., for delivery in combination with the enclosed anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab.
  • additional therapeutic agents for treating the disease or disorder involving IgE, e.g., for delivery in combination with the enclosed anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab.
  • kits may also comprise instructions for administration of the anti-lgE antibody or antigen-binding fragment thereof, (e.g., omalizumab or ligelizumab.) to treat the subject.
  • Such instructions may provide the dose (e.g., 24 mg, 72 mg, 75 mg, 120 mg, 240 mg or 300 mg), route of administration (e.g., IV, SC), and dosing regimen (e.g., every two or four weeks during e.g. 12 to 16 weeks) for use with the enclosed anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab.
  • dose e.g., 24 mg, 72 mg, 75 mg, 120 mg, 240 mg or 300 mg
  • route of administration e.g., IV, SC
  • dosing regimen e.g., every two or four weeks during e.g. 12 to 16 weeks
  • phrases “means for administering” is used to indicate any available implement for systemically administering a drug to a subject, including, but not limited to, a pre-filled syringe, a vial and syringe, an injection pen, an auto-injector, an IV drip and bag, a pump, etc.
  • a subject may self-administer the drug (i.e., administer the drug without the assistance of a physician) or a medical practitioner may administer the drug.
  • kits for use in modifying the disease course in a subject having disease or disorder involving IgE comprising an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof.
  • the kit further comprises means for administering the anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, to the subject.
  • Blood will be collected at the screening visit (Day -14), baseline (Day 1 , prior to omalizumab injection), Day 14, Day 30 (prior to omalizumab injection), Day 60 (prior to omalizumab injection), and Day 90 (study completion) for microsomal miRNA extraction, basophil isolation, and also stored at -70oC for later periostin assays.
  • Plasma exosomal miRNA bioinformatics analyses will be conducted in early (i.e., Day 14) and later (i.e., Day 30, 60, 90) responder groups.
  • the 2 wk time point will capture the early responders and the 4, 8, and 12 wk time points will capture the remaining responder groups based on data of CIU subjects with a starting UAS7 score of 25.3 ⁇ 2.0 (mean ⁇ SEM) treated with Xolair® outside of clinical trials (Metz et al., 2014), 57% attained complete response within one week of their first treatment and a further 29% within 4 weeks. All of the 12 subjects who gained remission during the first treatment week were symptom-free within 24 hr of Xolair® administration (Metz et al., 2014).
  • an urticaria activity score (UAS) during a 7-day period (UAS7) of >16 (on a scale ranging from 0 to 42, with higher scores indicating greater activity) during the 7 days before the first treatment with omalizumab.
  • Exosomes were isolated from the plasma of the study participants using Invitrogen’s Total Exosome Precipitation Reagent (Thermo Fisher Scientific, Inc., Waltham, MA) according to the manufacturer’s protocol. MicroRNAs were then isolated from the exosomes using the QIAGEN miRNeasy Serum/Plasma Kit (QIAGEN Sciences, Germantown, MD).
  • RNA samples were collected in EDTA tubes. Human nucleated cells were separated from red blood cells using HetaSep reagent STEMCELL Technologies, Vancouver, Canada). Basophils were isolated from nucleated cells using the STEMCELL Technologies EasySep Human Basophil Enrichment Kit. Purified basophils were centrifuged at 1 ,500 rpm for 10 minutes. The pellets were resuspended in 200 pL of PBS, 1 mL of Trizol was added, and the purified basophils were stored at -80oC until RNA was isolated.
  • Total RNA from basophils was isolated using the QIAGEN miRNeasy Micro extraction kit. This kit isolates total RNA that includes microRNAs. The integrity of the total RNA samples was evaluated using a 2100 Bioanalyzer instrument (Agilent Technologies, Inc., Palo Alto, CA). A Thermo Fisher Scientific NanoDropTM 1000 Spectrophotometer was used to determine purity of RNA samples by measuring OD260/280 and OD260/230 ratios. Only samples with an RNA Integrity Number of >7 and OD260/280 and OD260/230 ratios >1.8 were used for further analysis. Total RNA concentrations were determined using Thermo Fisher Scientific Qubit RNA HS Assay Kit.
  • MicroRNA profiling was performed at the QIAGEN Service lab in Germantown, MD. MicroRNA samples were evaluated for quality by using the QIAGEN QIAseq microRNA. Total RNA samples that included microRNAs were assessed for quality and quantity by using an Agilent TapeStation 4200 and a Thermo Fisher Scientific NanoDrop 2000c spectrophotometer respectively. Five pL exosomal microRNA and 5 pL of total basophil RNA (100 ng) were reverse transcribed into cDNA in 20 pL reactions using the QIAseq microRNA Library Kit and specially designed 3’ and 5’ adapters were ligated to mature microRNAs.
  • microRNA sequencing libraries prepared with the QIAseq microRNA Library Kit were sequenced using a NextSeq 500 instrument (lllumina, Inc., San Diego, CA). Libraries were quantified, adjusted to 4 nM each and pooled. The pooled libraries were further diluted down to 1.8 pM and loaded into a flow cell on the lllumina NextSeq 500. The sequencing specifications were 1x76 bases and 12 million targeted reads per sample.
  • basophil high subjects defined as those with a baseline basophil level >21 basophils/pL
  • basophil-low ⁇ 21 basophils/pL at baseline
  • High expression defined as those with expression level above the geometric mean expression level for that microRNA having log counts/million (logCPM) above zero, and those with low expression (defined as those with expression level below the geometric mean expression level for that microRNA).

Landscapes

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

Abstract

The present disclosure relates to methods for modifying the course of a disease or disorder involving IgE, in particular in subjects having an allergic disease or condition.

Description

METHODS OF TREATMENT USING OMALIZUMAB OR LIGELIZUMAB
TECHNICAL FIELD
The present disclosure relates to methods of preventing or treating diseases or disorders in subjects in need of treatment using an anti-lgE antibody, e.g., omalizumab or ligelizumab.
BACKGROUND OF THE DISCLOSURE
Immunoglobulin E (IgE) is an antibody associated with hypersensitivity and allergic reactions. IgE mainly binds on the high-affinity IgE receptor (FcsRI) on mast cells, basophils and dendritic cells and hence decreases the induction of the regulatory T cells.
Despite existing medications, these diseases are still not fully controlled and their treatments (systemic corticosteroids, immumosuppressors) have a known low safety profile. There is a need of identifying better treatments for these diseases, or at least to find alternative therapies that would permit administering lower doses of corticosteroids or immumosuppressors.
Xolair® (omalizumab) is a recombinant DNA-derived humanized monoclonal antibody that selectively binds to free, circulating human immunoglobulin E (IgE) thus inhibiting IgE binding to IgE receptors on the surface of mast cells and basophils resulting in decreased release of allergic mediators. By binding to free, circulating IgE, omalizumab also lowers serum free IgE levels and down-regulates the number of IgE receptors on the surface of mast cells and basophils. Omalizumab widely used for the treatment of Allergic asthma, allergic rhinitis and chronic spontaneous urticaria (CSU).
Ligelizumab is a humanized monoclonal antibody with higher affinity binding to human immunoglobulin E (IgE) than omalizumab. Upon binding, ligelizumab is able to block the interaction of IgE with both the high and low affinity IgE receptors (FceRI and FceRII). When subjects receive ligelizumab, circulating IgE is rapidly bound by the anti-lgE antibody and becomes inaccessible to IgE receptors on mast cells and basophils. IgE is necessary for the enhanced expression of the FceRI seen in atopic subjects, and a decrease in FceRI expression on circulating basophils accompanies ligelizumab treatment. Other potentially beneficial effects from anti-lgE therapy include decreased IgE production, reduced B cell numbers and reduced cytokine production by T cells. This mechanism confers benefit to subjects with chronic spontaneous urticaria (CSU) by preventing the itchy hives and angioedema that are associated with degranulation (histamine release) of mast cells and basophils. Ligelizumab demonstrated dose- and time-dependent suppression of free IgE, basophil FcsRI, basophil surface IgE, and skin prick test responses to allergen, superior in extent and duration to those observed with omalizumab. Superior affinity and pharmacodynamic (PD) outcomes of ligelizumab compared to omalizumab may translate into superior posology and superior clinical efficacy in subjects with CSU.
SUMMARY OF THE DISCLOSURE
MicroRNAs, small non-coding RNAs, are released by cells into the blood and other biofluids within small (<150 nm in diameter), stable membrane-bound microvesicles(termed exosomes), making them accessible and attractive biomarkers of disease pathogenesis and response to therapeutic intervention. We identified subjects receiving anti-lgE therapy, who were most discordant in their clinical response to it, to explore the utility of exosomal and basophil microRNA expression profiles as predictors of clinical response. The identification of those predictors of clinical response to treatment with anti-lgE antibodies, such as omalizumab or ligelizumab, opens the route of new treatment possibilities for many subjects with diseases or disorders involving IgE, in particular IgE-mediated disease or disorder, e g. Fc RI-mediated pathophysiologies. Such diseases or disorders are selected from the group consisting of: asthma (e.g. allergic asthma, moderate to severe persistent allergic asthma), Atopic dermatitis, Bullous Pemphigoid, uricaria, (e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria), Chronic Inducible Urticarias (CINDUs), rhinitis (e.g. Seasonal allergic rhinitis, such as e.g. Cedar pollinosis), Nasal polyposis, IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
Accordingly, disclosed herein are methods of preventing or treating diseases or disorders involving IgE, comprising administering a therapeutically effective amount of an anti-lgE antibody, e.g. an anti-lgE antibody that selectively binds to free, circulating human IgE (e.g., omalizumab or ligelizumab), to a subject in need thereof.
DETAILED DESCRIPTION OF THE DISCLOSURE
Embodiments A:
A1. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has low expression of miR-141-3p, in exosomes.
A2. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least one miRNA selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
A3. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least two miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
A4. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least three miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
A5. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least four miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
A6. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of at least five miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
A7. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has high expression of miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR- 3976, and optionally low expression of miR-141-3p, in exosomes. A8. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a basophil level of from about >19 basophils/pL to about ³60 basophils/pL
A9. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a basophil level of about >20 basophils/pL.
A10. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a basophil level of about >21 basophils/pL
A11 . An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
A12. An anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
A13. The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR-4664-3p in basophils.
A14. The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, wherein the subject has a basophil level of from about >19 basophils/pL to about ³60 basophils/pL, or of about ³20 basophils/pL, or of about ³21 basophils/pL.
A15. The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, or according to embodiment A14, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
A16. The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, or according to embodiment A14, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
A17. The anti-lgE antibody or antigen binding fragment thereof for use in treating or preventing a disease or disorder involving IgE in a subject in need thereof according to any one of embodiments A1-A7, or according to embodiment A14, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR-4664-3p in basophils.
A18. The anti-lgE antibody or antigen binding fragment thereof according to any one of embodiments A1-A17, comprising administering to the subject in need thereof a therapeutically effective amount of an anti-lgE antibody or antigen binding fragment thereof.
A19. The anti-lgE antibody or antigen binding fragment thereof according to any one of the above embodiments, wherein the disease or disorder is selected from the group consisting of: asthma (e.g. allergic asthma, moderate to severe persistent allergic asthma), Atopic dermatitis, Bullous Pemphigoid, uricaria, (e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria), Chronic Inducible Urticarias (CINDUs), rhinitis (e.g. Seasonal allergic rhinitis, such as e.g. Cedar pollinosis), Nasal polyposis, IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
A20. The anti-lgE antibody or antigen binding fragment thereof according to any of the above embodiments, wherein the disease or condition is selected from allergic asthma, chronic spontaneous urticaria, allergic rhinitis, food allergy.
A21. The anti-lgE antibody or antigen binding fragment thereof according to any of the above embodiments, wherein the anti-lgE antibody is omalizumab or ligelizumab.
A22. The anti-lgE antibody or antigen binding fragment thereof according to embodiment A21 , wherein the anti-lgE antibody is omalizumab. A23. The anti-lgE antibody or antigen binding fragment thereof according to embodiment A22, wherein omalizumab is administered at a dose of about 75 mg to about 600 mg, e g. at a maximum dose of 600 mg.
A24. The anti-lgE antibody or antigen binding fragment thereof according to embodiment A21 , wherein the anti-lgE antibody is ligelizumab
A25. The anti-lgE antibody or antigen binding fragment thereof according to embodiment A24, wherein ligelizumab is administered at a dose of about 24 mg to about 600 mg, e g. at a maximum dose of 600 mg.
A26. The anti-lgE antibody or antigen binding fragment thereof according to embodiment A22 or embodiment A23, wherein omalizumab is administered every two to four weeks.
A27. The anti-lgE antibody or antigen binding fragment thereof according to embodiment A24 or embodiment A25, wherein ligelizumab is administered every two to four weeks.
A28. The anti-lgE antibody or antigen binding fragment thereof according to any one of embodiments A21 to A27, wherein omalizumab or ligelizumab, is administered during up to 16 weeks, e.g. 12 to 16 weeks.
A29. The anti-lgE antibody or antigen binding fragment thereof according to any of the above embodiments, wherein the anti-lgE antibody is co-administered with a corticosteroid and/or an immunusuppressor, or a long-acting beta agonist (LABA).
Embodiments B
B1. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has low expression of miR-141-3p, in exosomes.
B2. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least one miRNA selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
B3. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least two miRNAs selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
B4. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least three miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
B5. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least four miRNAs selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
B6. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least five miRNAs selected from miR-6499-5p, miR- 7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
B7. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2- 3p, miR-6877-3p, and miR-3976, and optionally low expression of miR-141-3p, in exosomes. B8. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a basophil level offrom about³19 basophils/pL to about ³60 basophils/pL
B9. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a basophil level of about ³20 basophils/pL.
B10. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a basophil level of about >21 basophils/pL
B11 . A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR- 1236-3p, or miR-4664-3p in basophils.
B12. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR- 1236-3p, or miR-4664-3p in basophils.
B13. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR-4664-3p in basophils.
Embodiments C
C1 . A method of identifying subjects responders to a therapy with an anti-lgE antibody, comprising (1) measuring the expression level of one or more miR-1200, miR-1236-3p and miR-4664-3p in basophils, in a sample from said subject, and (2) determining the responders based on the expression level of such miRNAs. C2. A method of identifying subjects responders to a therapy with an anti-lgE antibody, comprising (1) measuring the expression level of one or more miR-6499-5p, miR-7848-3p, miR-4494, miR- 450a-2-3p, miR-6877-3p, miR-3976, and miR-141-3p in exosomes, in a sample from said subject, and (2) determining the responders based on the expression level of such miRNAs.
C3. A method of screening a patient to be treated with a therapy comprising an anti-lgE antibody or antigen binding fragment comprising measuring the expression level of one or more miR-1200, miR-1236-3p and miR-4664-3p in basophils, in a sample from a patient in need of such a therapy, wherein an high level of such miRNA indicates responder to such anti-lgE antibody or antigen binding fragment.
C4. A method of screening a patient to be treated with a therapy comprising an anti-lgE antibody or antigen binding fragment comprising measuring the expression level of one or more miR-6499- 5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976, in a sample from a patient in need of such therapy, wherein an high level of such miRNA indicates responder to such anti-lgE antibody or antigen binding fragment.
C5. A method of miRNAs in patient screening comprising measuring the expression level of miR- 141 -3p in exosomes, in a sample from a subject in need of an anti-lgE antibody or antigen binding fragment, wherein an low level of such miRNA indicates responder to such anti-lgE antibody or antigen binding fragment.
C6. Use of miRNAs to identify subjects in need of therapy with an anti-lgE antibody or antigen binding fragment, comprising (1 ) measuring the expression level of one or more miR-1200, miR- 1236-3p and miR-4664-3p in a sample from said subject, and (2) selecting a subject having a high level expression of said miRNA.
C7. Use of miRNAs to identify subjects in need of therapy with an anti-lgE antibody or antigen binding fragment, comprising (1) measuring the expression level of one or more miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976, in a sample from said subject, and (2) selecting a subject having a high level expression of said miRNA.
C8. Use of miRNAs to identify subjects in need of therapy with an anti-lgE antibody or antigen binding fragment, comprising (1) measuring the expression level of miR-141-3p in exosomes, in a sample from a subject in need of an anti-lgE antibody or antigen binding fragment, and (2) selecting a subject having a low level expression of said miRNA.
C9. Use of miRNAs to select a subject responder to a therapy with an anti-lgE antibody or antigen binding fragment, comprising selecting a subject having a high expression level of one or more miR-1200, miR-1236-3p, miR-4664-3p miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976.
C10. Use of miRNAs to select a subject responder to a therapy with an anti-lgE antibody or antigen binding fragment, comprising selecting a subject having a low expression level of miR- 141 -3p in exosomes.
C11. Use of one or more miRNAs selected from the list: miR-1200, miR-1236-3p, miR-4664-3p, miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, miR-3976, and miR-141- 3p as a biomarker for treatment response to therapy using anti-lgE antibody or antigen binding fragment.
C12. Use of basophil levels as a biomarker for treatment response to therapy using anti-lgE antibody or antigen binding fragment, wherein the basophil level is from about >19 basophils/pL to about ³60 basophils/pL, or of about ³20 basophils/pL, or of about ³21 basophils/pL.
C13. A biomarker comprising one or more miRNAs selected from the list: miR-1200, miR-1236- 3p, miR-4664-3p, miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, miR- 3976, and miR-141-3p.
C14. A kit for treating particular subjects having disease or disorder involving IgE comprising an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, (e.g., in liquid or lyophilized form) or a pharmaceutical composition comprising the anti-lgE antibody as described herein, and a biomarker according to any of the above embodiments.
In another embodiment, high expression miRNA is referring to a difference between expression of such an miRNA between a non-responder and a complete responder of at least 3 logCPM, at least 3 logCPM, at least 4 logCPM, wherein the expression of the miRNA is higher in the responder than in the non-responder.
In another embodiment, low expression miRNA is referring to a difference between expression of such an miRNA between a complete responder and a non-responder of at least 3 logCPM, at least 3 logCPM, at least 4 logCPM, wherein the expression of the miRNA is lower in the responder than in the non-responder.
DEFINITIONS
As used herein, IgE refers to Immunoglobulin E.
The term “comprising” encompasses “including” as well as “consisting,” e.g., a composition “comprising” X may consist exclusively of X or may include something additional, e.g., X + Y.
The term “about” in relation to a numerical value x means, for example, +/- 10%. When used in front of a numerical range or list of numbers, the term “about” applies to each number in the series, e.g., the phrase “about 1-5” should be interpreted as “about 1 - about 5”, or, e.g., the phrase “about 1 , 2, 3, 4” should be interpreted as “about 1 , about 2, about 3, about 4, etc.”
The word “substantially” does not exclude “completely,” e.g., a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from the definition of the disclosure.
The term "antibody" as referred to herein includes naturally-occurring and whole antibodies. A naturally-occurring "antibody" is a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed hypervariable regions or complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
The term "antigen-binding fragment" of an antibody, as used herein, refers to fragments of an antibody that retain the ability to specifically bind to IgE. It has been shown that the antigenbinding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody include a Fab fragment, a monovalent fragment consisting of the VL, V , CL and CH1 domains; a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CH1 domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a dAb fragment (Ward et al. , 1989 Nature 341 :544-546), which consists of a V domain; and an isolated CDR. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al., 1988 Science 242:423-426; and Huston et al., 1988 Proc. Natl. Acad. Sci. 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antibody”. Single chain antibodies and antigen-binding portions are obtained using conventional techniques known to those of skill in the art.
The term "KD" is intended to refer to the dissociation rate of a particular antibody-antigen interaction. The term "KD", as used herein, is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e., Kd/Ka) and is expressed as a molar concentration (M). KD values for antibodies can be determined using methods well established in the art. A preferred method for determining the KD of an antibody is by using surface plasmon resonance, or using a biosensor system such as a Biacore® system. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof according to the invention, e.g., omalizumab, binds human IgE with a KD of about 0.02 to 7.7 nM, e.g. 100-250 pM.
The term "affinity" refers to the strength of interaction between antibody and antigen at single antigenic sites. Within each antigenic site, the variable region of the antibody “arm” interacts through weak non-covalent forces with antigen at numerous sites; the more interactions, the stronger the affinity. Standard assays to evaluate the binding affinity of the antibodies toward IgE of various species are known in the art, including for example, ELISAs, western blots and RIAs. The binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore analysis.
The term "derivative", unless otherwise indicated, is used to define amino acid sequence variants, and covalent modifications (e.g. pegylation, deamidation, hydroxylation, phosphorylation, methylation, etc.) of an anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab, according to the present disclosure, e.g., of a specified sequence (e.g., a variable domain). A “functional derivative” includes a molecule having a qualitative biological activity in common with the disclosed anti-lgE antibodies. A functional derivative includes fragments and peptide analogs of an anti-lgE antibody as disclosed herein. Fragments comprise regions within the sequence of a polypeptide according to the present disclosure, e.g., of a specified sequence.
The phrase “substantially identical” means that the relevant amino acid or nucleotide sequence (e.g., VH or VL domain) will be identical to or have insubstantial differences (e.g., through conserved amino acid substitutions) in comparison to a particular reference sequence. Insubstantial differences include minor amino acid changes, such as 1 or 2 substitutions in a 5 amino acid sequence of a specified region (e.g., VH or VL domain). In the case of antibodies, the second antibody has the same specificity and has at least 50% of the affinity of the same. Sequences substantially identical (e.g., at least about 85% sequence identity) to the sequences disclosed herein are also part of this application. In some embodiments, the sequence identity of a derivative anti-lgE antibody (e.g., a derivative of omalizumab, e.g., an omalizumab biosimilar antibody) can be about 90% or greater, e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher relative to the disclosed sequences.
’’Identity” with respect to a native polypeptide and its functional derivative is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity, and not considering any conservative substitutions as part of the sequence identity. Neither N- or C-terminal extensions nor insertions shall be construed as reducing identity. Methods and computer programs for the alignment are well known. The percent identity can be determined by standard alignment algorithms, for example, the Basic Local Alignment Search Tool (BLAST) described by Altshul et al. ((1990) J. Mol. Biol., 215: 403410); the algorithm of Needleman et al. ((1970) J. Mol. Biol., 48: 444 453); or the algorithm of Meyers et al. ((1988) Comput. Appl. Biosci., 4: 11 17). A set of parameters may be the Blosum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5. The percent identity between two amino acid or nucleotide sequences can also be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM 120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
"Amino acid(s)" refer to all naturally occurring L-a-amino acids, e.g., and include D-amino acids. The phrase "amino acid sequence variant" refers to molecules with some differences in their amino acid sequences as compared to the sequences according to the present disclosure. Amino acid sequence variants of an antibody according to the present disclosure, e.g., of a specified sequence, still have the ability to bind the IgE. Amino acid sequence variants include substitutional variants (those that have at least one amino acid residue removed and a different amino acid inserted in its place at the same position in a polypeptide according to the present disclosure), insertional variants (those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a polypeptide according to the present disclosure) and deletional variants (those with one or more amino acids removed in a polypeptide according to the present disclosure).
The term “pharmaceutically acceptable” means a nontoxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s).
The term “administering” in relation to a compound, e.g., an anti-lgE antibody, is used to refer to delivery of that compound to a subject by any route.
As used herein, a “therapeutically effective amount” refers to an amount of anti-lgE antibody (e.g., omalizumab or an antigen-binding fragment thereof) that is effective, upon single or multiple dose administration to a subject (such as a human) for treating, preventing, preventing the onset of, curing (if applicable), delaying, reducing the severity of, ameliorating at least one symptom of a disorder or recurring disorder, or prolonging the survival of the subject beyond that expected in the absence of such treatment. When applied to an individual active ingredient (e.g., an anti-lgE antibody, e.g., omalizumab) administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
The term "treatment" or “treat” is herein defined as the application or administration of an anti-lgE antibody according to the disclosure, for example, omalizumab, or a pharmaceutical composition comprising said anti-lgE antibody, to a subject or to an isolated tissue or cell line from a subject, where the subject has a particular disease, a symptom associated with the disease, or a predisposition towards development of the disease, where the purpose is to cure (if applicable), delay the onset of, reduce the severity of, alleviate, ameliorate one or more symptoms of the disease, improve the disease, reduce or improve any associated symptoms of the disease or the predisposition toward the development of the disease. The term “treatment” or “treat” includes treating a subject suspected to have the disease as well as subjects who are ill or who have been diagnosed as suffering from the disease or medical condition, and includes suppression of clinical relapse.
Furthermore, the term "treatment" or “treat” is herein defined as the application or administration of an IgE antibody according to the disclosure, for example ligelizumab, or a pharmaceutical composition comprising said anti- IgE antibody, to a subject or to an isolated tissue or cell line from a subject, where the subject has a particular disease (e.g., CSU), a symptom associated with the disease (e.g., CSU), or a predisposition towards development of the disease (e.g., CSU) (if applicable), where the purpose is to cure (if applicable), delay the onset of, reduce the severity of, alleviate, ameliorate one or more symptoms of the disease, improve the disease, reduce or improve any associated symptoms of the disease or the predisposition toward the development of the disease. The term “treatment” or “treat” includes treating a subject suspected to have the disease as well as subjects who are ill or who have been diagnosed as suffering from the disease or medical condition, and includes suppression of clinical relapse.
As used herein, the phrase “population of subjects” is used to mean a group of subjects.
In some embodiments of the disclosed methods, the IgE antagonist (e.g., IgE antibody, such as ligelizumab) is used to treat a population of CSU subjects.
As used herein, the term “basophil level” refers to the number of basophils per ml blood, as determined according to the known methods in the art.
As herein defined, the term “diseases or disorders involving IgE” refers to IgE-mediated disease or disorder, e.g. FcsRI-mediated pathophysiologies e.g. to disease or disorder selected from the group consisting of: asthma (e.g. allergic asthma, moderate to severe persistent allergic asthma), Atopic dermatitis, Bullous Pemphigoid, uricaria, (e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria), Chronic Inducible Urticarias (CINDUs), rhinitis (e.g. Seasonal allergic rhinitis, such as e.g. Cedar pollinosis), Nasal polyposis, IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
As herein defined, the term “high expression” of a miRNA refers to an expression level above the geometric mean expression level for that microRNA, e.g. having a log counts/million (logCPM) above zero, e.g. above 1 , e.g. above 2, e.g. around or above 4.
As herein defined, the term “low expression” of a miRNA refers to an expression level below the geometric mean expression level for that microRNA, e.g. having a log counts/million (logCPM) below zero, e.g. 1 logCPM below zero, e.g. 2 or more logCPM below zero.
As herein defined, the log counts/million (logCPM) may be measured by using the voomWithQualityWeights function (Liu R, Holik AZ, Su S, et al., Nucleic Acids Res 2015;43:e97) from the Bioconductor limma package (Ritchie ME, Phipson B, Wu D, et al., Nucleic Acids Res 2015;43:e47).
For example, in the CSU treatment, Responders are defined as those subjects with an AUC for the weekly UAS7 over the 12-week treatment period that was equal to or less than the 75th percentile and achieved 90% improvement of baseline UAS7. Non-responders are defined as subjects who a) failed to achieve a 90% improvement of their baseline weekly UAS7, or b) had an AUC for the weekly UAS7 over the 12-week study period that was above the 75th percentile (i.e. , top quartile), or c) had an exacerbation of their symptoms necessitating the addition of an anti-inflammatory or an immunosuppressive agent.
Non-responders to therapy using an anti-lgE antibody or antigen-binding fragment thereof, are defined as subjects who failed to achieve a 90% improvement of their baseline or had an exacerbation of their symptoms. Responders to therapy using an anti-lgE antibody or antigenbinding fragment thereof are defined as those subjects who achieved 90% improvement of baseline.
Anti-lgE antibodies
In some embodiments of the disclosed uses, methods, and kits, the anti-lgE antibody or antigen-binding fragment thereof is a monoclonal antibody. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is a human or humanized antibody. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is a humanized antibody. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is a human antibody of the lgGi subtype. In some embodiments, the anti-lgE antibody or antigen-binding fragment thereof is omalizumab. In other embodiments, the anti-lgE antibody or antigen-binding fragment thereof is ligelizumab.
Exemplary anti-lgE antibodies include, but are not limited to, omalizumab, quilizumab, ligelizumab and etrolizumab.
Alternatively, an anti-lgE antibody or antigen-binding fragment thereof used in the disclosed methods may be an amino acid sequence variant of the reference anti-lgE antibodies set forth herein.
The disclosure also includes anti-lgE antibodies or antigen-binding fragments thereof (e.g., omalizumab) in which one or more of the amino acid residues of the VH or VL domain of omalizumab, typically only a few (e.g. 1-10), are changed; for instance by mutation, e.g., site directed mutagenesis of the corresponding DNA sequences.
Methods of Treatment and Uses of anti-lgE antibodies
The disclosed anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, may be used in vitro, ex vivo, or incorporated into pharmaceutical compositions and administered in vivo to treat subjects (e.g., human subjects) affected by one or more disease or disorder involving IgE.
The anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof may be used as a pharmaceutical composition when combined with a pharmaceutically acceptable carrier. Such a composition may contain, in addition to the anti-lgE antibody or antigen-binding fragment thereof, carriers, various diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The characteristics of the carrier will depend on the route of administration.
Pharmaceutical compositions for use in the disclosed methods may be manufactured in conventional manner. In one embodiment, the pharmaceutical composition is provided in lyophilized form. For immediate administration it is dissolved in a suitable aqueous carrier, for example sterile water for injection or sterile buffered physiological saline. Other formulations comprise liquid or lyophilized formulation.
Antibodies, e.g., antibodies to IgE, or antigen-binding fragment thereof, are typically formulated either in aqueous form ready for parenteral administration or as lyophilisates for reconstitution with a suitable diluent prior to administration. In some embodiments of the disclosed methods and uses, the anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab, is formulated as a lyophilisate. Suitable lyophilisate formulations can be reconstituted in a small liquid volume (e.g., 2ml or less, e.g., 1 ml) to allow subcutaneous administration and can provide solutions with low levels of antibody aggregation. Techniques for purification of antibodies to a pharmaceutical grade are well known in the art.
Disclosed methods of, and anti-lgE antibodies for use in, preventing, treating or modifying the course of a disease or disorder involving IgE in a subject in need thereof, comprising herein are administering the subject a therapeutically effective amount of an anti-lgE antibody or antigen binding fragment thereof.
For example, diseases or disorders involving IgE are selected from the group consisting of.
In another example, the disease or disorder involving IgE refers to food allergies, e.g. peanuts allergy.
In yet another example, the disease or disorder involving IgE refers to diseases or disorders selected from Allergic asthma, allergic rhinitis and chronic spontaneous urticaria (CSU).
In yet another example, the disease or disorder involving IgE refers to chronic spontaneous urticaria (CSU).
In yet another example, the disease or disorder involving IgE refers to Allergic asthma.
Disclosed herein are methods of, and anti-lgE antibodies (e.g omalizumab or ligelizumab) for use in, preventing, treating or modifying the course of a disease or disorder involving IgE in a subject in need thereof, comprising administering the subject a therapeutically effective amount of anti-lgE antibody or antigen-binding fragment thereof.
The subject in need thereof may have an insulin allergy.
In yet another embodiment, the subject is affected by a disease or disorder selected from allergy, asthma, urticarial and rhinitis, e.g. a disease or condition selected from allergic asthma, CSU and allergic rhinitis.
In another embodiment, the subject is affected by asthma, e.g. allergic asthma.
In another example, the subject is affected by urticaria, e.g. chronic spontaneous urticaria
(CSU).
In another embodiment, the subject is affected by rhinitis, e.g. allergic rhinitis.
In another embodiment, the subject is affected by food allergies, e.g. peanuts allergy.
Ligelizumab demonstrated dose- and time-dependent suppression of free IgE, basophil FcsRI, basophil surface IgE, and skin prick test responses to allergen, superior in extent and duration to those observed with omalizumab. Superior affinity and pharmacodynamic (PD) outcomes of ligelizumab compared to omalizumab may translate into superior posology and superior clinical efficacy in subjects. Lieglizumab binds to IgE with 150fold higher affinity than omalizumab (A. Eggel, Molecular, Structural And Mechanistic Insight Into Ligelizumab Mediated Suppression Of IgE Dependent Allergic Responses, EAACI 2019).
Furthermore, disclosed herein are anti-lgE antibodies or antigen-binding fragments thereof, e.g. omalizumab or ligelizumab, for use in, preventing, treating or modifying the course of a disease or disorder involving IgE in a subject in need thereof, comprising administering the subject a therapeutically effective amount of anti-lgE antibody or antigen-binding fragment thereof, wherein the subject is not affected by allergy, asthma, urticarial or rhinitis.
In one embodiment, the subject in need thereof has allergy.
In another embodiment, the subject is affected by asthma, e.g. allergic asthma.
In another example, the subject is affected by urticaria, e.g. chronic spontaneous urticaria
(CSU).
In another embodiment, the subject is affected by rhinitis, e.g. allergic rhinitis.
The appropriate dosage will vary depending upon, for example, the particular anti-lgE antibody to be employed, the host, the mode of administration and the nature and severity of the condition being treated, and on the nature of prior treatments that the subject has undergone. It may also depend on the level of IgE in the subject’s blood before initiating the treatment with the anti-lgE antibody.
Ultimately, the attending health care provider will decide the amount of the anti-lgE antibody with which to treat each individual subject. In some embodiments, the attending health care provider may administer low doses of the anti-lgE antibody and observe the subject’s response, in particular the blood level of IgE.
For the asthma indication, the usual dose range of omalizumab is between 75 mg and 600 mg in one to four subcutaneously injections, and the maximum recommended dose is 600 mg. The dosing of omalizumab for treating asthma is determined based on the subject’s weight and the subject’s serum total IgE level. The dosage of omalizumab for chronic urticaria indication is 300 mg sc per month.
In one embodiment of the present disclosure, omalizumab is administered subcutaneously at a dose of about 75mg to about 600 mg, e.g. at a dose of about 300 mg, e.g. at a maximum dose of 600 mg.
In one embodiment of the present disclosure, ligelizumab is administered subcutaneously at a dose of about 24 mg to about 600 mg, at a dose of about 24 mg to about 240 mg, e.g. at a dose of about 24 mg, of about 72 mg, of about 120 mg, or of about 240 mg, e.g. at a maximum dose of 600 mg.
In some embodiments, the level of IgE in the subject’s blood is measured before initiating the administration of the anti-lgE antibody, e.g., omalizumab or ligelizumab, and the dose of the antibody is adjusted based on the weight of the subject and/or his serum total IgE level.
The duration of therapy using a pharmaceutical composition of the present disclosure will vary, depending on the severity of the disease or disorder to be treated and the condition and personal response of each individual subject. In some embodiments, the subject is administered the anti-lgE antibody (e.g., omalizumab or ligelizumab) for long-term, e.g. at least 12 weeks, e.g. up to 16 weeks, e.g. to 12 to 16 weeks.
In some embodiments, the anti-lgE antagonist (e.g., omalizumab or ligelizumab) is administered to the subject every two weeks, e.g. every two or four weeks, e.g. monthly.
The anti-lgE antibody or antigen-binding fragment thereof according to the present disclosure, e.g., omalizumab or ligelizumab, is conveniently administered parenterally, e.g., intravenously, intramuscularly, or subcutaneously, e.g. subcutaneously.
The anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab, may be administered to the subject subcutaneously (SC), e.g. at about 75 mg to about 600 mg (e.g. about 75 mg, about 600 mg), e.g. at about 300 mg.
The anti-lgE antibody or antigen-binding fragment thereof, e.g., ligelizumab, may be administered to the subject subcutaneously (SC), e.g. at about 24 mg to about 600 mg (e.g. about 24 mg, about 72 mg, about 120 mg, about 240 mg, or about 600 mg), e.g. at about 120 mg.
In practicing some of the methods of treatment or uses of the present disclosure, a therapeutically effective amount of an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, is administered to a subject, e.g., a mammal (e.g., a human). While it is understood that the disclosed methods provide for treatment of diseases or disorders involving IgE, using anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof this does not preclude that, if the subject is to be ultimately treated with an anti- lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, therapy is necessarily a monotherapy.
Indeed, if a subject is selected for treatment with an anti-lgE antibody or antigen-binding fragment thereof, then the anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, may be administered in accordance with the methods of the disclosure either alone or in combination with other agents and therapies for treating the subject affected by the disease or disorder involving IgE, e.g., in combination with at least one additional therapeutic agent, such as e.g., a corticosteroid or an immumosuppressor, e.g., a systemic corticosteroid or an immunosuppressor.
That can be the case for example, when the subject to be treated is allergic, or when the subject is also affected by another disease or disorder selected from asthma, urticaria, and rhinitis, e.g. selected from allergic asthma, CSU, and allergic rhinitis.
When coadministered with one or more additional psoriasis agent(s), the anti-lgE antibody or antigen-binding fragment thereof may be administered either simultaneously with the other agent, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering the anti-lgE antibody or antigen-binding fragment thereof in combination with other agents and the appropriate dosages for co-delivery.
Various therapies may be beneficially combined with the disclosed anti-lgE antibodies, such as omalizumab or ligelizumab, during treatment of the disease or disorder involving IgE, disclosed herein. Such therapies include for example corticosteroids (e.g., systemic corticosteroids) or immunosuppressors.
Disclosed herein are methods of, and anti-lgE antibodies (e.g., omalizumab, or ligelizumab) or antigen-binding fragment thereof, for use in, modifying the course of a disease or disorder involving IgE, in a subject in need thereof, comprising administering the subject a dose of about 75 mg to about 600 mg of an anti-lgE antibody (e.g., omalizumab) or antigen-binding fragment thereof by subcutaneous injection.
The use of antibodies as the active ingredient of pharmaceuticals is now widespread, including the products HERCEPTIN™ (trastuzumab), RITUXAN™ (rituximab), SYNAGIS™ (palivizumab), etc. Techniques for purification of antibodies to a pharmaceutical grade are known in the art. When a therapeutically effective amount of an IgE antagonist, e.g., IgE binding molecules (e.g., IgE antibody or antigen-binding fragment thereof, e.g., ligelizumab) or IgE receptor binding molecules (e.g., IgE antibody or antigen-binding fragment thereof) is administered by intravenous, cutaneous or subcutaneous injection, the IgE antagonist will be in the form of a pyrogen-free, parenterally acceptable solution. A pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection may contain, in addition to the IgE antagonist, an isotonic vehicle such as sodium chloride, Ringer's solution, dextrose, dextrose and sodium chloride, lactated Ringer's solution, or other vehicle as known in the art.
In practicing some of the methods of treatment or uses of the present disclosure, a therapeutically effective amount of an IgE antagonist, e.g., IgE binding molecule (e.g., IgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab) or IgE receptor binding molecule (e.g., IgE antibody or antigen-binding fragment thereof) is administered to a subject, e.g., a mammal (e.g., a human). While it is understood that the disclosed methods provide for treatment of CSU subjects using an IgE antagonist (e.g., omalizumab or ligelizumab), this does not preclude that, if the subject is to be ultimately treated with an IgE antagonist, such IgE antagonist therapy is necessarily a monotherapy. Indeed, if a subject is selected for treatment with an IgE antagonist, then the IgE antagonist (e.g., omalizumab or ligelizumab) may be administered in accordance with the methods of the disclosure either alone or in combination with other agents and therapies for treating CSU subjects, e.g., in combination with at least one additional CSU agent. When co-administered with one or more additional CSU agent(s), an IgE antagonist may be administered either simultaneously with the other agent, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering the IgE antagonist in combination with other agents and the appropriate dosages for co-delivery.
Disclosed herein are methods of, and anti-lgE antibodies (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, for use in, modifying the course of a disease or disorder involving IgE, in a subject in need thereof, comprising administering the subject a dose of about 24 mg to about 600 mg of an anti-lgE antibody (e.g. ligelizumab) or antigen-binding fragment thereof by subcutaneous injection.
In some embodiments of the disclosed uses, methods, and kits, the subject has allergy, asthma and/or urticarial, e.g., has a disease or disorder selected from asthma, allergic asthma, rhinitis, allergic rhinitis, urticarial and CSU. In some embodiments of the disclosed uses, methods, and kits, the subject has allergic asthma, allergic urticaria and/or CSU. In other embodiments of the disclosed uses, methods, and kits, the subject has allergic asthma and CSU. In some embodiments of the disclosed uses, methods, and kits, the anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, can be prescribed as first treatment or added on to any of the standard of care medications.
The timing of dosing is generally measured from the day of the first dose of anti-lgE antibody, e.g. ligelizumab or omalizumab, (which is also known as “baseline”). However, health care providers often use different naming conventions to identify dosing schedules, as shown in
Table 1.
Figure imgf000023_0001
Table 1 : Common naming conventions for dosing regimens. Bolded items refer to the naming convention used herein.
Notably, week zero may be referred to as week one by some health care providers, while day zero may be referred to as day one by some health care providers. Thus, it is possible that different physicians will designate, e.g., a dose as being given during week 4 / on day 28, during week 4 / on day 29, during week 4 / on day 28, during week 4 / on day 29, while referring to the same dosing schedule. For consistency, the first week of dosing will be referred to herein as week 0, while the first day of dosing will be referred to as day 1 . However, it will be understood by a skilled artisan that this naming convention is simply used for consistency and should not be construed as limiting, i.e. , weekly dosing is the provision of a weekly dose of the IgE antibody regardless of whether the physician refers to a particular week as “week 0” or “week 1”.
In a one dosing regimen, the antibody is administered during week 0, 4, 8, 12, 16, 20, etc. Some providers may refer to this regimen as monthly dosing (or dosing every 4 weeks). It will be appreciated by a skilled artisan that administering a patient an injection at weeks 0 followed by once monthly dosing starting at week 4 is the same as: 1) administering the patient an injection at weeks 0 and 4, followed by once monthly dosing starting at week 8; 2) administering the patient an injection at weeks 0 and 4 followed by dosing every 4 weeks; and 3) administering the patient an injection at weeks 0 and 4 followed by monthly administration.
Kits
The disclosure also encompasses kits for treating particular subjects having disease or disorder involving IgE. Such kits comprise an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, (e.g., in liquid or lyophilized form) or a pharmaceutical composition comprising the anti-lgE antibody (described supra). Additionally, such kits may comprise means for administering the anti-lgE antibody or antigen-binding fragment thereof (e.g., an auto-injector, a syringe and vial, a prefilled syringe, a prefilled pen) and instructions for use. These kits may contain additional therapeutic agents (described supra) for treating the disease or disorder involving IgE, e.g., for delivery in combination with the enclosed anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab. Such kits may also comprise instructions for administration of the anti-lgE antibody or antigen-binding fragment thereof, (e.g., omalizumab or ligelizumab.) to treat the subject. Such instructions may provide the dose (e.g., 24 mg, 72 mg, 75 mg, 120 mg, 240 mg or 300 mg), route of administration (e.g., IV, SC), and dosing regimen (e.g., every two or four weeks during e.g. 12 to 16 weeks) for use with the enclosed anti-lgE antibody or antigen-binding fragment thereof, e.g., omalizumab or ligelizumab.
The phrase “means for administering” is used to indicate any available implement for systemically administering a drug to a subject, including, but not limited to, a pre-filled syringe, a vial and syringe, an injection pen, an auto-injector, an IV drip and bag, a pump, etc. With such items, a subject may self-administer the drug (i.e., administer the drug without the assistance of a physician) or a medical practitioner may administer the drug.
Disclosed herein are kits for use in modifying the disease course in a subject having disease or disorder involving IgE, comprising an anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof. In some embodiments, the kit further comprises means for administering the anti-lgE antibody (e.g., omalizumab or ligelizumab) or antigen-binding fragment thereof, to the subject.
General
The details of one or more embodiments of the disclosure are set forth in the accompanying description above. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, the preferred methods and materials are now described. Other features, objects, and advantages of the disclosure will be apparent from the description and from the embodiments. In the specification and the appended embodiments, the singular forms include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents and publications cited in this specification are incorporated by reference. The following Examples are presented in order to more fully illustrate the preferred embodiments of the disclosure.
These examples should in no way be construed as limiting the scope of the disclosed subject matter, as defined by the appended embodiments.
EXAMPLES
1. Clinical Study
This is a single center, non-comparative exploratory study, to investigate the effect of omalizumab over a 3-month treatment period in adult (>18 years) subjects with chronic idiopathic urticaria who had remained symptomatic despite the use of high dose H1-antihistamines. After an initial screening visit within two weeks of the Day 1 baseline visit, subjects receive one subcutaneous injection of omalizumab at a dose of 300 mg on Days 1 , 30, and 60. Blood will be collected at the screening visit (Day -14), baseline (Day 1 , prior to omalizumab injection), Day 14, Day 30 (prior to omalizumab injection), Day 60 (prior to omalizumab injection), and Day 90 (study completion) for microsomal miRNA extraction, basophil isolation, and also stored at -70oC for later periostin assays. Plasma exosomal miRNA bioinformatics analyses will be conducted in early (i.e., Day 14) and later (i.e., Day 30, 60, 90) responder groups. The 2 wk time point will capture the early responders and the 4, 8, and 12 wk time points will capture the remaining responder groups based on data of CIU subjects with a starting UAS7 score of 25.3 ± 2.0 (mean ± SEM) treated with Xolair® outside of clinical trials (Metz et al., 2014), 57% attained complete response within one week of their first treatment and a further 29% within 4 weeks. All of the 12 subjects who gained remission during the first treatment week were symptom-free within 24 hr of Xolair® administration (Metz et al., 2014).
INCLUSION CRITERIA
1) adult subjects (>18 years), 2) at least 6 weeks of CSU with itching despite current use of up to x4 H1 -antihistamines, an urticaria activity score (UAS) during a 7-day period (UAS7) of >16 (on a scale ranging from 0 to 42, with higher scores indicating greater activity) during the 7 days before the first treatment with omalizumab.
EXCLUSION CRITERIA
1) a clearly defined underlying cause for chronic urticaria, 2) routine administration (i.e., daily or every other day for ³5 consecutive days) of systemic glucocorticoids, antiinflammatory, immunomodulatory, or immunosuppressive agents within the previous 30 days, 3) the use of any H2-antihistamine or leukotriene-receptor antagonist within 7 days preceding the screening visit, 4) treatment with omalizumab within the previous year, or a known hypersensitivity to omalizumab, 5) a history of cancer, or 6) women who were pregnant or nursing or unable to use an effective method of contraception during dosing with omalizumab.
OMALIZUMAB TREATMENT
Twenty-seven subjects were screened (Day -14 visit). After initial screening within two weeks of study start, subjects received one subcutaneous injection of omalizumab at a dose of 300 mg on Days 0, 30, and 60.
EXOSOMAL MICRORNA ISOLATION
Exosomes were isolated from the plasma of the study participants using Invitrogen’s Total Exosome Precipitation Reagent (Thermo Fisher Scientific, Inc., Waltham, MA) according to the manufacturer’s protocol. MicroRNAs were then isolated from the exosomes using the QIAGEN miRNeasy Serum/Plasma Kit (QIAGEN Sciences, Germantown, MD).
BASOPHIL ISOLATION
Peripheral blood samples were collected in EDTA tubes. Human nucleated cells were separated from red blood cells using HetaSep reagent STEMCELL Technologies, Vancouver, Canada). Basophils were isolated from nucleated cells using the STEMCELL Technologies EasySep Human Basophil Enrichment Kit. Purified basophils were centrifuged at 1 ,500 rpm for 10 minutes. The pellets were resuspended in 200 pL of PBS, 1 mL of Trizol was added, and the purified basophils were stored at -80oC until RNA was isolated.
BASOPHIL MICRORNA EXTRACTION
Total RNA from basophils was isolated using the QIAGEN miRNeasy Micro extraction kit. This kit isolates total RNA that includes microRNAs. The integrity of the total RNA samples was evaluated using a 2100 Bioanalyzer instrument (Agilent Technologies, Inc., Palo Alto, CA). A Thermo Fisher Scientific NanoDrop™ 1000 Spectrophotometer was used to determine purity of RNA samples by measuring OD260/280 and OD260/230 ratios. Only samples with an RNA Integrity Number of >7 and OD260/280 and OD260/230 ratios >1.8 were used for further analysis. Total RNA concentrations were determined using Thermo Fisher Scientific Qubit RNA HS Assay Kit.
MICRORNA-SEQ PROFILING
MicroRNA profiling was performed at the QIAGEN Service lab in Germantown, MD. MicroRNA samples were evaluated for quality by using the QIAGEN QIAseq microRNA. Total RNA samples that included microRNAs were assessed for quality and quantity by using an Agilent TapeStation 4200 and a Thermo Fisher Scientific NanoDrop 2000c spectrophotometer respectively. Five pL exosomal microRNA and 5 pL of total basophil RNA (100 ng) were reverse transcribed into cDNA in 20 pL reactions using the QIAseq microRNA Library Kit and specially designed 3’ and 5’ adapters were ligated to mature microRNAs. The ligated microRNAs were then reverse transcribed to cDNAs using a reverse transcription primer that contains an integrated Unique Molecular Index (UMI). Following cDNA cleanup, PCR library amplification (19-22 cycles) was performed with a universal forward primer and indexing reverse primers. Following a final library cleanup, the quality of the microRNA libraries was assessed using the Agilent TapeStation 4200. MicroRNA sequencing libraries prepared with the QIAseq microRNA Library Kit were sequenced using a NextSeq 500 instrument (lllumina, Inc., San Diego, CA). Libraries were quantified, adjusted to 4 nM each and pooled. The pooled libraries were further diluted down to 1.8 pM and loaded into a flow cell on the lllumina NextSeq 500. The sequencing specifications were 1x76 bases and 12 million targeted reads per sample.
Results
Baseline basophil level correlated significantly with response to omalizumab a measured by the AUC UAS over the 12-week study period (R2 = 0.33, P = 0.007).
When subjects were classified based on their baseline basophil level, basophil high subjects (defined as those with a baseline basophil level >21 basophils/pL) had a significantly better response to omalizumab with lower mean UAS7 over time compared to basophil-low (<21 basophils/pL at baseline) subjects.
High expression (defined as those with expression level above the geometric mean expression level for that microRNA) having log counts/million (logCPM) above zero, and those with low expression (defined as those with expression level below the geometric mean expression level for that microRNA).
We found seven differentially expressed exosomal microRNAs at baseline that were associated with a complete response to omalizumab (FDR <0.05).
Complete responders had higherexpression of six of these microRNAs: miR-6499-5p , miR-7848- 3p, miR-4494, miR-450a-2-3p, miR-6877 -3p and miR-3976.
Complete responders had lower expression of miR-141-3p.
Higher expression of miR-6499-5p, miR-7848-3p-3p, miR-4494, miR-450a-2-3p, miR- 6877-3p and miR-3976 was associated with favorable response to omalizumab. In contrast, there was a trend for higher expression of miR-141-3p to be associated with poor response to omalizumab.
We found three differentially expressed basophil microRNAs at baseline associated with complete response to omalizumab: Complete responders had higher expression of miR-1200, miR-1236-3p and miR-4664-3p.
Using the QIAGEN® IPA microRNA Target Filter feature, we identified 1019 mRNA targets for the seven differentially expressed exosomal microRNAs between the complete responders and non-responders on Day 0.
Subjects with ³21 basophils/pL had a lower mean UAS7 over the 12-week study (P<0.001). Complete responders had significantly (FDR<0.05) higher expression of miR-6499- 5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, and miR-3976 and lower expression of miR-141-3p in exosomes and higher expression of miR-1200, miR-1236-3p, and miR-4664-3p in basophils. The differentially expressed exosomal microRNAs, acting in concert, modulated Tec Kinase Signaling pathway activity (FDR=0.003).

Claims

WHAT IS EMBODIMENTED IS:
1. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has low expression of miR-141-3p, in exosomes.
2. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least one miRNA selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
3. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least two miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
4. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least three miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
5. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least four miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
6. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of at least five miRNAs selected from miR-6499-5p, miR-7848-3p, miR-4494, miR-450a-2-3p, miR-6877-3p, or miR-3976, and optionally low expression of miR-141-3p, in exosomes.
7. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has high expression of miR-6499-5p, miR-7848-3p, miR- 4494, miR-450a-2-3p, miR-6877-3p, and miR-3976, and optionally low expression of miR- 141 -3p, in exosomes.
8. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a basophil level of from about >19 basophils/pL to about >60 basophils/pL
9. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a basophil level of about >20 basophils/pL.
10. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a basophil level of about >21 basophils/pL
11. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
12. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR-1236-3p, or miR-4664-3p in basophils.
13. A method of treating or preventing a disease or disorder involving IgE in a subject in need thereof, comprising administering an anti-lgE antibody or antigen binding fragment to said subject, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR- 4664-3p in basophils.
14. The method according to any one of embodiments 1-7, wherein the subject has a basophil level of from about >19 basophils/pL to about >60 basophils/pL, or of about >20 basophils/pL, or of about >21 basophils/pL.
15. The method according to any one of claims 1-7, or according to claim 14, wherein the subject has a high expression of at least one miRNA selected from miR-1200, miR-1236- 3p, or miR-4664-3p in basophils.
16. The method according to any one of claim 1-7, or according to claim 14, wherein the subject has a high expression of at least two miRNA selected from miR-1200, miR-1236- 3p, or miR-4664-3p in basophils.
17. The method according to any one of claim 1-7, or according to claim 14, wherein the subject has a high expression of miR-1200, miR-1236-3p and miR-4664-3p in basophils.
18. The method according to any one of claims 1-17, comprising administering to the subject in need thereof a therapeutically effective amount of an anti-lgE antibody or antigen binding fragment thereof.
19. The method according to any one of the above claims, wherein the disease or disorder is selected from the group consisting of: asthma (e.g. allergic asthma, moderate to severe persistent allergic asthma), Atopic dermatitis, Bullous Pemphigoid, uricaria, (e.g. chronic spontaneous urticaria (CSU), also known as chronic idiopathic urticaria), Chronic Inducible Urticarias (CINDUs), rhinitis (e.g. Seasonal allergic rhinitis, such as e.g. Cedar pollinosis), Nasal polyposis, IgE driven food allergy to one or more allergens (e.g. food allergy, peanuts allergy), idiopathic angioedema.
20. The method according to any of the above claims, wherein the disease or condition is selected from allergic asthma, chronic spontaneous urticaria and allergic rhinitis.
21. The method according to any of the above claims, wherein the anti-lgE antibody is omalizumab or ligelizumab.
22. The method according to claim 21 , wherein the anti-lgE antibody is omalizumab.
23. The method according to claim 22, wherein omalizumab is administered at a dose of about 75 mg to about 600 mg, e.g. at a maximum dose of 600 mg.
24. The according to claim 21 , wherein the anti-lgE antibody is ligelizumab
25. The method according to claim 24, wherein ligelizumab is administered at a dose of about 24 mg to about 600 mg, e.g. at a maximum dose of 600 mg.
26. The method according to claim 22 or claim 23, wherein omalizumab is administered every two to four weeks.
27. The method according to claim 24 or claim 25, wherein ligelizumab is administered every two to four weeks.
28. The method according to any one of claims 21 to 27, wherein omalizumab or ligelizumab, is administered during up to 16 weeks, e.g. 12 to 16 weeks.
29. The method according to any of the above claims, wherein the anti-lgE antibody is co administered with a corticosteroid and/or an immunusuppressor, or a long-acting beta agonist (LABA).
PCT/IB2021/054956 2020-06-09 2021-06-07 Methods of treatment using omalizumab or ligelizumab WO2021250533A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202063036663P 2020-06-09 2020-06-09
US63/036,663 2020-06-09
US202063039267P 2020-06-15 2020-06-15
US63/039,267 2020-06-15
US202063128515P 2020-12-21 2020-12-21
US63/128,515 2020-12-21

Publications (1)

Publication Number Publication Date
WO2021250533A1 true WO2021250533A1 (en) 2021-12-16

Family

ID=76422024

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/054956 WO2021250533A1 (en) 2020-06-09 2021-06-07 Methods of treatment using omalizumab or ligelizumab

Country Status (1)

Country Link
WO (1) WO2021250533A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116790615A (en) * 2023-07-11 2023-09-22 康霖生物科技(杭州)有限公司 Gene therapy vector nucleic acid construct for allergic diseases and application method thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019186369A1 (en) * 2018-03-26 2019-10-03 Novartis Ag Methods of treating chronic spontaneous urticaria using ligelizumab

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019186369A1 (en) * 2018-03-26 2019-10-03 Novartis Ag Methods of treating chronic spontaneous urticaria using ligelizumab

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
AL-SHAIKHLY TAHA ET AL: "Extracellular vesicle microRNAs as predictors of response to omalizumab in chronic spontaneous urticaria", ALLERGY, vol. 76, no. 4, 31 December 2020 (2020-12-31), United Kingdom, pages 1274 - 1277, XP055831569, ISSN: 0105-4538, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1111/all.14702> DOI: 10.1111/all.14702 *
ALTSHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 410
ANONYMOUS: "Three-month, Single-center Study to Identify Biomarkers/Response to Xolair Therapy in Chronic Idiopathic Urticaria -NCT02814630", CLINICALTRIALS.GOV, 24 January 2019 (2019-01-24), pages 1 - 6, XP055838611, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/NCT02814630> [retrieved on 20210907] *
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
E. MEYERSW. MILLER, CABIOS, vol. 4, 1989, pages 11 - 17
HUSTON ET AL., PROC. NATL. ACAD. SCI., vol. 85, 1988, pages 5879 - 5883
LIN CHING-KOW E ET AL: "Differential Expression of MicroRNAs and their Possible Roles in Patients with Chronic Idiopathic Urticaria and Active Hives", ALLERGY & RHINOLOGY (PROVIDENCE, R.I.), 1 June 2017 (2017-06-01), Los Angeles, CA, pages 67 - 80, XP055838543, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5468759/pdf/arhe67.pdf> [retrieved on 20210907], DOI: 10.2500/ar.2017.8.0199 *
MEYERS ET AL., COMPUT. APPL. BIOSCI., vol. 4, 1988, pages 11 17
NEEDLEMAN ET AL., J. MOL. BIOL., vol. 48, 1970, pages 444 453
PINTO GOUVEIA M. ET AL: "Long-term management of chronic spontaneous urticaria with omalizumab", CLINICAL AND EXPERIMENTAL DERMATOLOGY, vol. 42, no. 7, 25 June 2017 (2017-06-25), GB, pages 735 - 742, XP055838844, ISSN: 0307-6938, Retrieved from the Internet <URL:https://api.wiley.com/onlinelibrary/tdm/v1/articles/10.1111%2Fced.13173> DOI: 10.1111/ced.13173 *
PUXEDDU ILARIA ET AL: "Biomarkers In Chronic Spontaneous Urticaria: Current Targets And Clinical Implications", JOURNAL OF ASTHMA AND ALLERGY, vol. Volume 12, 20 September 2019 (2019-09-20), pages 285 - 295, XP055838524, Retrieved from the Internet <URL:https://www.dovepress.com/getfile.php?fileID=52912> DOI: 10.2147/JAA.S184986 *
RITCHIE MEPHIPSON BWU D ET AL., NUCLEIC ACIDS RES, vol. 43, 2015, pages e47
SPECJALSKI KRZYSZTOF ET AL: "MicroRNAs: Potential Biomarkers and Targets of Therapy in Allergic Diseases?", ARCHIVUM IMMUNOLOGIAE ET THERAPIAE EXPERIMENTALIS, BIRKHAEUSER VERLAG AG, CH, vol. 67, no. 4, 28 May 2019 (2019-05-28), pages 213 - 223, XP036820214, ISSN: 0004-069X, [retrieved on 20190528], DOI: 10.1007/S00005-019-00547-4 *
SUSSMAN GORDON ET AL: "Real-life experiences with omalizumab for the treatment of chronic urticaria", ANNALS OF ALLERGY, ASTHMA & IMMUNOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 112, no. 2, 28 December 2013 (2013-12-28), pages 170 - 174, XP028815544, ISSN: 1081-1206, DOI: 10.1016/J.ANAI.2013.12.005 *
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
YU CHENJIE ET AL: "Clinical Efficacy and Safety of Omalizumab in the Treatment of Allergic Rhinitis: A Systematic Review and Meta-analysis of Randomized Clinical Trials", AMERICAN JOURNAL OF RHINOLOGY & ALLERGY, vol. 34, no. 2, 1 March 2020 (2020-03-01), US, pages 196 - 208, XP055838971, ISSN: 1945-8924, Retrieved from the Internet <URL:https://journals.sagepub.com/doi/pdf/10.1177/1945892419884774> DOI: 10.1177/1945892419884774 *
ZHU YONG-MING ET AL: "Analysis the effect of miR-141-3p/HMGB1 in LPS-induced mucus production and the apoptosis in nasal epithelial cells", KAOHSIUNG JOURNAL OF MEDICAL SCIENCES, vol. 36, no. 8, 13 April 2020 (2020-04-13), TW, pages 622 - 629, XP055838785, ISSN: 1607-551X, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1002/kjm2.12215> DOI: 10.1002/kjm2.12215 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116790615A (en) * 2023-07-11 2023-09-22 康霖生物科技(杭州)有限公司 Gene therapy vector nucleic acid construct for allergic diseases and application method thereof

Similar Documents

Publication Publication Date Title
AU2019279946B2 (en) Methods for treating atopic dermatitis by administering an IL-4R antagonist
JP6306588B2 (en) Method for treating or preventing asthma by administering an IL-4R antagonist
JP2023139071A (en) Methods for treating or preventing asthma by administering il-4r antagonists
US20210196822A1 (en) Treatment of triple negative breast cancer with targeted tgf-b inhibition
JP2021523881A (en) How to treat chronic idiopathic urticaria with rigerizumab
WO2021250533A1 (en) Methods of treatment using omalizumab or ligelizumab
US20230009657A1 (en) Methods of treating lupus nephritis using interleukin-17 (il-17) antagonists
JP2023527054A (en) Treatment of cardiometabolic disease with inhibitors of type I interferon signaling
WO2018158741A1 (en) Psoriasis disease modification following long-term treatment with an il-17 antagonist
CN113993543B (en) Combination therapy using anti-CD 38 antibodies
JP6923594B2 (en) Methods for treating or preventing asthma by administering IL-4R antagonists
US20220340687A1 (en) Methods of treatment using omalizumab
US20210230265A1 (en) Methods for treating copd by administering an il-33 antagonist

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: 21732124

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: 21732124

Country of ref document: EP

Kind code of ref document: A1