US20190233533A1 - Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38 - Google Patents

Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38 Download PDF

Info

Publication number
US20190233533A1
US20190233533A1 US16/312,133 US201716312133A US2019233533A1 US 20190233533 A1 US20190233533 A1 US 20190233533A1 US 201716312133 A US201716312133 A US 201716312133A US 2019233533 A1 US2019233533 A1 US 2019233533A1
Authority
US
United States
Prior art keywords
antibody
seq
ige
specifically binds
mediated disease
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US16/312,133
Inventor
Henny G. Otten
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UMC Utrecht Holding BV
Original Assignee
UMC Utrecht Holding BV
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 UMC Utrecht Holding BV filed Critical UMC Utrecht Holding BV
Priority to US16/312,133 priority Critical patent/US20190233533A1/en
Assigned to UMC UTRECHT HOLDING B.V. reassignment UMC UTRECHT HOLDING B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OTTEN, Henny G.
Publication of US20190233533A1 publication Critical patent/US20190233533A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01035Hyaluronoglucosaminidase (3.2.1.35), i.e. hyaluronidase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Definitions

  • the present invention relates to treatment of IgE-mediated disease with antibodies that specifically bind CD38.
  • IgE-mediated allergies are triggered by binding of IgE to the high affinity IgE receptor (Fc ⁇ RI), which is expressed on effector mast cells, basophils and activated eosinophils.
  • Fc ⁇ RI high affinity IgE receptor
  • effector mast cells basophils
  • activated eosinophils As a result of these high affinity interactions, stable Fc ⁇ RI:IgE complexes are displayed on the surface of effector cells.
  • Exposure to allergens leads to cross-linking and eventually clustering of IgE:Fc ⁇ RI complexes, thus triggering effector cell activation, degranulation and release of stored pro-allergenic mediators that leads to the initiation of an allergic response.
  • Atopic allergy is associated with anaphylactic hypersensitivity and includes the disorders, e.g., asthma, allergic rhinitis and conjunctivitis (hay fever), eczema, urticaria and food allergies. Further, an allergic reaction may lead to a dangerous life-threatening condition such as anaphylactic shock, which may be provoked by insect bites.
  • IgE In addition to allergies, IgE plays a role in autoimmune disorders contributing to their pathogenesis (Ettinger et al., Autoimmunity 50:25-35, 2017; Holgate, World Allergy Organization Journal 7:17, 2014).
  • IgE-mediated diseases such as allergies and autoimmune diseases.
  • the invention provides for a method of treating an IgE-mediated disease, comprising administering to a subject in need thereof an antibody that specifically binds CD38 for a time sufficient to treat the IgE-mediated disease.
  • FIG. 1 shows that total IgE and timothy grass and house dust mite-specific IgE is reduced in a multiple myeloma patient treated with DARZALEXTM (daratumumab) over time.
  • CD38 refers to the human CD38 protein (synonyms: ADP-ribosyl cyclase 1, cADPr hydrolase 1, cyclic ADP-ribose hydrolase 1).
  • Human CD38 has an amino acid sequence shown in GenBank accession number NP_001766 and in SEQ ID NO: 1. It is well known that CD38 is a single pass type II membrane protein with amino acid residues 1-21 representing the cytosolic domain, amino acid residues 22-42 representing the transmembrane domain, and residues 43-300 representing the extracellular domain of CD38.
  • Antibodies as used herein is meant in a broad sense and includes immunoglobulin molecules including monoclonal antibodies including murine, human, humanized and chimeric monoclonal antibodies, antibody fragments, bispecific or multispecific antibodies, dimeric, tetrameric or multimeric antibodies, single chain antibodies, domain antibodies and any other modified configuration of the immunoglobulin molecule that comprises an antigen binding site of the required specificity.
  • Immunoglobulins may be assigned to five major classes, namely IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence.
  • IgA and IgG are further sub-classified as the isotypes IgA1, IgA2, IgG1, IgG2, IgG3 and IgG4.
  • Antibody light chains of any vertebrate species can be assigned to one of two clearly distinct types, namely kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of their constant domains.
  • Antibody fragments refers to a portion of an immunoglobulin molecule that retains the heavy chain and/or the light chain antigen binding site, such as heavy chain complementarity determining regions (HCDR) 1, 2 and 3, light chain complementarity determining regions (LCDR) 1, 2 and 3, a heavy chain variable region (VH), or a light chain variable region (VL).
  • HCDR heavy chain complementarity determining regions
  • LCDR light chain complementarity determining regions
  • VH heavy chain variable region
  • VL light chain variable region
  • Antibody fragments include a Fab fragment, a monovalent fragment consisting of the VL, VH, 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 domain antibody (dAb) fragment (Ward et al., Nature 341:544-6, 1989), which consists of a VH domain or a VL domain.
  • dAb domain antibody
  • VH and VL domains may be engineered and linked together via a synthetic linker to form various types of single chain antibody designs where the VH/VL domains pair intramolecularly, or intermolecularly in those cases when the VH and VL domains are expressed by separate single chain antibody constructs, to form a monovalent antigen binding site, such as single chain Fv (scFv) or diabody; described for example in Intl. Pat. Publ. Nos. WO1998/44001, WO1988/01649, WO1994/13804, and WO1992/01047.
  • scFv single chain Fv
  • WO1998/44001 WO1988/01649
  • WO1994/13804 WO1992/01047.
  • isolated antibody refers to an antibody or antibody fragment that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody specifically binding CD38 is substantially free of antibodies that specifically bind antigens other than human CD38).
  • An isolated antibody that specifically binds CD38 may have cross-reactivity to other antigens, such as orthologs of human CD38, such as Macaca fascicularis (cynomolgus) CD38.
  • the bispecific antibody specifically binds two antigens of interest, and is substantially free of antibodies that specifically bind antigens other that the two antigens of interest.
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • isolated antibody encompasses antibodies that are isolated to a higher purity, such as antibodies that are 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% pure.
  • An antibody variable region consists of a “framework” region interrupted by three “antigen binding sites”.
  • the antigen binding sites are defined using various terms: Complementarity Determining Regions (CDRs), three in the VH (HCDR1, HCDR2, HCDR3) and three in the VL (LCDR1, LCDR2, LCDR3) are based on sequence variability (Wu and Kabat J Exp Med 132:211-50, 1970; Kabat et al Sequences of Proteins of Immunological Interest, 5th Ed.
  • HVR HVR
  • HV antigen-binding variable domains
  • IMGT-CDRs Lefranc et al., Dev Comparat Immunol 27:55-77, 2003
  • SDRU Specificity Determining Residue Usage
  • IMGT International ImMunoGeneTics
  • Chothia residues as used herein are the antibody VL and VH residues numbered according to Al-Lazikani (Al-Lazikani et al., J Mol Biol 273:927-48, 1997).
  • Framework or “framework sequences” are the remaining sequences of a variable region other than those defined to be antigen binding sites. Because the antigen binding sites can be defined by various terms as described above, the exact amino acid sequence of a framework depends on how the antigen-binding site was defined.
  • Humanized antibody refers to an antibody in which the antigen binding sites are derived from non-human species and the variable region frameworks are derived from human immunoglobulin sequences. Humanized antibodies may include substitutions in the framework regions so that the framework may not be an exact copy of expressed human immunoglobulin or germline gene sequences.
  • Human antibody refers to an antibody having heavy and light chain variable regions in which both the framework and the antigen binding sites are derived from sequences of human origin and is optimized to have minimal immune response when administered to a human subject. If the antibody contains a constant region, the constant region also is derived from sequences of human origin.
  • a human antibody comprises heavy or light chain variable regions that are “derived from” sequences of human origin wherein the variable regions of the antibody are obtained from a system that uses human germline immunoglobulin or rearranged immunoglobulin genes.
  • Such systems include human immunoglobulin gene libraries displayed on phage, and transgenic non-human animals such as mice carrying human immunoglobulin loci as described herein.
  • a “human antibody” may contain amino acid differences when compared to the human germline immunoglobulin or rearranged immunoglobulin genes due to differences between the systems used to obtain the antibody and human immunoglobulin loci, introduction of somatic mutations or intentional introduction of substitutions in the framework or antigen binding site, or both.
  • human antibody is at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical in amino acid sequence to an amino acid sequence encoded by human germline immunoglobulin or rearranged immunoglobulin genes.
  • human antibody may contain consensus framework sequences derived from human framework sequence analyses, for example as described in Knappik et al., J Mol Biol 296:57-86, 2000, or synthetic HCDR3 incorporated into human immunoglobulin gene libraries displayed on phage, for example as described in Shi et al., J Mol Biol 397:385-96, 2010 and Intl. Pat. Publ. No. WO2009/085462.
  • Human antibodies derived from human immunoglobulin sequences may be generated using systems such as phage display incorporating synthetic CDRs and/or synthetic frameworks, or can be subjected to in vitro mutagenesis to improve antibody properties, resulting in antibodies that do not naturally exist within the human antibody germline repertoire in vivo.
  • Antibodies in which antigen binding sites are derived from a non-human species are not included in the definition of human antibody.
  • Recombinant antibody includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse or a rat) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further below), antibodies isolated from a host cell transformed to express the antibody, antibodies isolated from a recombinant, combinatorial antibody library, and antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences, or antibodies that are generated in vitro using Fab arm exchange such as bispecific antibodies.
  • “Monoclonal antibody” refers to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope, or in a case of a bispecific monoclonal antibody, a dual binding specificity to two distinct epitopes.
  • “Monoclonal antibody” therefore refers to an antibody population with single amino acid composition in each heavy and each light chain, except for possible well known alterations such as removal of C-terminal lysine from the antibody heavy chain or alterations due to post-translational modification(s) of amino acids, such as methionine oxidation or asparagine or glutamine deamidation.
  • Monoclonal antibodies may have heterogeneous glycosylation within the antibody population.
  • Monoclonal antibody may be monospecific or multispecific, or monovalent, bivalent or multivalent. A bispecific antibody is included in the term monoclonal antibody.
  • Epitope means a portion of an antigen to which an antibody specifically binds.
  • Epitopes usually consist of chemically active (such as polar, non-polar or hydrophobic) surface groupings of moieties such as amino acids or polysaccharide side chains and may have specific three-dimensional structural characteristics, as well as specific charge characteristics.
  • An epitope may be composed of contiguous and/or noncontiguous amino acids that form a conformational spatial unit. For a noncontiguous epitope, amino acids from differing portions of the linear sequence of the antigen come in close proximity in 3-dimensional space through the folding of the protein molecule.
  • Variant refers to a polypeptide or a polynucleotide that differs from a reference polypeptide or a reference polynucleotide by one or more modifications for example, substitutions, insertions or deletions.
  • “In combination with” means that two or more therapeutics are administered to a subject together in a mixture, concurrently as single agents or sequentially as single agents in any order. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • Treat” or “treatment” refers to therapeutic treatment wherein the object is to slow down (lessen) an undesired physiological change or disease, or to provide a beneficial or desired clinical outcome during treatment.
  • Beneficial or desired clinical outcomes include alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state.
  • Those in need of treatment include those subjects already with the undesired physiological change or disease as well as those subjects prone to have the physiological change or disease.
  • “Therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • a therapeutically effective amount may vary according to factors such as the disease severity, age, sex, and weight of the individual, and the ability of a therapeutic or a combination of therapeutics to elicit a desired response in the individual.
  • Exemplary indicators of an effective therapeutic or combination of therapeutics include, for example, improved well-being of the subject, reduction in symptoms of the disease, such as reduction in sneezing, coughing, sinus congestion, mucus production in the sinuses (rhinitis) or lungs (asthma), itching, swelling, and/or decreased IgE levels in a subject.
  • “Patient” includes any human or nonhuman animal.
  • “Nonhuman animal” includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc.
  • the terms “patient” and “subject” are used interchangeably.
  • Specific binding or “specifically binds” or “binds” refers to an antibody binding to an antigen or an epitope within the antigen with greater affinity than for other antigens.
  • the antibody binds to the antigen or the epitope within the antigen with an equilibrium dissociation constant (K D ) of about 1 ⁇ 10 ⁇ 8 M or less, for example about 1 ⁇ 10 ⁇ 9 M or less, about 1 ⁇ 10 ⁇ 10 M or less, about 1 ⁇ 10 ⁇ 11 M or less, or about 1 ⁇ 10 ⁇ 12 M or less, typically with the K D that is at least one hundred-fold less than its K D for binding to a non-specific antigen (e.g., BSA, casein).
  • K D equilibrium dissociation constant
  • the dissociation constant may be measured using standard procedures.
  • Antibodies that specifically bind to the antigen or the epitope within the antigen may, however, have cross-reactivity to other related antigens, for example to the same antigen from other species (homologs), such as human or monkey, for example Macaca fascicularis (cynomolgus, cyno), Pan troglodytes (chimpanzee, chimp) or Callithrix jacchus (common marmoset, marmoset). While a monospecific antibody specifically binds one antigen or one epitope, a bispecific antibody specifically binds two distinct antigens or two distinct epitopes.
  • homologs such as human or monkey
  • Macaca fascicularis cynomolgus, cyno
  • Pan troglodytes chimpanzee, chimp
  • Callithrix jacchus common marmoset, marmoset
  • An IgE-mediated disease refers to a disease that is mediated, at least in part, by an increase in the level of IgE in a subject. “Increase in the level of IgE” refers to the level of total IgE of >_2 kU/L and/or the level of allergen specific IgE of ⁇ 0.35 kU/L using ImmunoCAP assay (ThermoFisher, Uppsala, Sweden) using methodology described herein and according to the manufacturer's instructions.
  • IgE-mediated diseases include disorders associated with increased IgE levels or activity in which atypical symptoms may manifest due to levels of IgE locally and/or systemically in the body even if the threshold of IgE of >2 kU/L is not achieved systemically.
  • the current invention is based, at least in part, on the identification that treatment of a subject with an antibody that specifically binds CD38 reduces total IgE and allergen-specific IgE in the subject over time.
  • an antibody that specifically binds CD38 mediates killing of the B cells expressing and/or secreting IgE.
  • Allergic diseases are conventionally described as IgE-mediated diseases.
  • Clinical manifestations of allergic diseases include allergic asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and anaphylactic reactions to insect bites or drugs.
  • Allergic diseases are caused by hypersensitivity reactions of the immune system to specific allergens (such as pollen, stings, drugs, or food). The most severe form of an allergic disorder is anaphylactic shock.
  • IgE immunoglobulin type E
  • sensitization After sensitization, exposure to the allergen induces crosslinking of IgE bound to mast cells and basophils causing a wide release of vasoactive mediators, such as histamine. Therefore, therapy directed to IgE has been a research focus for several years.
  • a different method of targeting IgE may be by depleting plasma cells that produce (specific) IgE.
  • the effects of B-cell depletion on IgE levels and clinical symptoms of atopic diseases have been studied before.
  • Rituxan® did not significantly decrease serum IgE levels compared to placebo at three or six months in patients with Idiopathic Thrombocytopenic Purpura (Dasgupta A et al., Allergy Asthma Clin Immunol 2013; 9:39. doi:10.1186/1710-1492-9-39).
  • VELCADE® (bortezomib), targeting (malignant) plasma cells via proteasome inhibition, is also being investigated as a treatment option in (auto) antibody mediated diseases (Rosenberg AS et al., Clin Immunol 2016. doi:10.1016/j.clim.2016.02.009).
  • bortezomib treatment reduced specific IgE levels (Wegmann M et al., Int Arch Allergy Immunol 2012; 158:43-53. doi:10.1159/000330103).
  • the invention provides a method of treating an IgE-mediated disease, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody that specifically binds CD38 for a time sufficient to treat the IgE-mediated disease.
  • the invention also provides for an antibody that specifically binds CD38 for use in treating a subject having an IgE-mediated disease.
  • the invention also provides use of an antibody that specifically binds CD38 in the manufacture of a medicament for the treatment of an IgE-mediated disease.
  • the invention also provides use of an antibody that specifically binds CD38 in the preparation of a pharmaceutical composition for the treatment of an IgE-mediated disease.
  • the IgE-mediated disease is an allergic response to an allergen.
  • allergens include airborne allergens, such as those of house dust mite, pets and pollens, for example house dust mites allergens obtained from Dermatophagoides spp or D. pteronyssinus, D. farinae and D.
  • microceras Euroglyphus maynei or Blomia sp.
  • allergens from insects present in cockroach or Hymenoptera allergens from pollen, such as pollens of tree, grass and weed, allergens from animals, especially in cat, dog, horse and rodent, allergens from fungi, such as from Aspergillus, Altemaria or Cladosporium, and occupational allergens including animal and plant antigens as well as drugs, detergents, metals and immunoenhancers such as isocyanates and allergens present in products such as latex or amylase.
  • allergens also include ingested allergens responsible for food hypersensitivity, such as fruits, vegetables and milk, such as food allergens present in peanuts, fish e.g. codfish, egg white, crustacean e.g. shrimp, milk e.g. cow's milk, wheat, cereals, fruits of the Rosacea family (apple, plum, strawberry), vegetables of the Liliacea, Cruciferae, Solanaceae and Umbelliferae families, tree nuts, sesame, peanut, soybean and other legume family allergens, spices, melon, avocado, mango, fig, banana.
  • Rosacea family apple, plum, strawberry
  • Rosacea family apple, plum, strawberry
  • vegetables of the Liliacea, Cruciferae, Solanaceae and Umbelliferae families tree nuts, sesame, peanut, soybean and other legume family allergens, spices, melon, avocado, mango, fig, banana.
  • Non-antigen specific stimuli that can result in an IgE-mediated reaction include infection, irritants such as smoke, combustion fumes, diesel exhaust particles and sulphur dioxide, exercise, cold and emotional stress.
  • Specific hypersensitivity reactions in atopic and nonatopic individuals with a certain genetic background may result from exposure to proteins in foods (e.g., legumes, peanuts), venom (e.g., insect, snake), vaccines, hormones, antiserum, enzymes, latex, antibiotics, muscle relaxants, vitamins, cytotoxins, opiates, and polysaccharides such as dextrin, iron dextran and polygeline.
  • foods e.g., legumes, peanuts
  • venom e.g., insect, snake
  • vaccines e.g., hormones, antiserum, enzymes, latex, antibiotics, muscle relaxants, vitamins, cytotoxins, opiates, and polysaccharides such as dextrin, iron dextran and polygeline.
  • the allergen is pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
  • IgE has also been associated with pathogenic mechanisms of inflammation and autoimmunity.
  • IgE autoantibodies have been detected in dermatological autoimmune disorders such as bullous pemphigoid (BP) and chronic spontaneous urticaria (CSU), in systemic sclerosis, thyroiditis, multiple sclerosis and atopic dermatitis
  • BP bullous pemphigoid
  • CSU chronic spontaneous urticaria
  • Immune complexes of IgE autoantibodies and autoantigens may trigger mast cell and basophil degranulation and induce IFN ⁇ , TNF and IL-6 production by dendritic cells (DC).
  • IgE can promote antigen cross-presentation triggering both CD4 and CD8 T cell responses, and drive B cell expansion and plasma cell differentiation via DC activation.
  • Xolair® an anti-IgE antibody has been reported to exhibit efficacy or is being investigated in at least non-allergic asthma, Churg-Strauss Syndrome, allergic rhinitis, atopic dermatitis, nasal polyposis, food allergy, chronic urticaria and angioedema, Kimura's disease, mastocytosis, anaphylaxis, systemic lupus erythematosus, Sjogren's Syndrome (Holgate, World Allergy Organization Journal 7:17, 2014; ClinicalTrials registry). Therefore, it can be expected that an anti-CD38 antibody depleting IgE producing cells would be efficacious in these diseases.
  • IgE-mediated diseases include, asthma, atopic dermatitis, allergic rhinitis, fibrosis (e.g., pulmonary fibrosis, such as IPF), allergic asthma, food allergy, anaphylaxis, contact dermatitis, allergic gastroenteropathy, allergic bronchopulmonary aspergillosis, allergic purpura (Henoch-Schonlein), ataxia-telangiectasia, Churg-Strauss Syndrome, eczema, enteritis, gastroenteropathy, graft-versus-host reaction, hyper-IgE (Job's) syndrome, hypersensitivity (e.g., anaphylactic hypersensitivity, candidiasis, vasculitis), IgE myeloma, inflammatory bowel disease (e.g., Crohn's disease, ulcerative colitis, indeterminate colitis and infectious colitis), mucositis (e.g., oral mucositis, gastrointestinal mucositis, nasal mucos
  • lupus may be systemic lupus erythematosus (SLE), discoid lupus erythematosus, subacute cutaneous lupus erythematosus, neonatal lupus or drug-induced lupus.
  • SLE systemic lupus erythematosus
  • discoid lupus erythematosus discoid lupus erythematosus
  • subacute cutaneous lupus erythematosus subacute cutaneous lupus erythematosus
  • neonatal lupus or drug-induced lupus.
  • disorders that may be treatable by lowering IgE levels, regardless of whether the disorders themselves are associated with elevated IgE are within the scope of IgE-mediated disease.
  • the IgE-mediated disease is allergic asthma.
  • the IgE-mediated disease is urticaria.
  • the IgE-mediated disease is angioedema.
  • the IgE-mediated disease is food allergy.
  • the IgE-mediated disease is atopic dermatitis.
  • the IgE-mediated disease is anaphylaxis.
  • the IgE-mediated disease is cutaneous mastocytosis.
  • the IgE-mediated disease is allergic rhinitis.
  • the IgE-mediated disease is nasal polyposis.
  • the IgE-mediated disease is Kimura's disease.
  • the IgE-mediated disease is eosinophilic otitis media.
  • the IgE-mediated disease is eosinophilic gastroenteritis.
  • the IgE-mediated disease is latex allergy.
  • the IgE-mediated disease is bronchopulmonary allergic aspergillosis.
  • the IgE-mediated disease is bullous pemphigoid (BP).
  • the IgE-mediated disease is systemic lupus erythematosus (SLE).
  • the IgE-mediated disease is rheumatoid arthritis (RA).
  • the IgE-mediated disease is acute or chronic.
  • the IgE-mediated disease is an acute disease.
  • the IgE-mediated disease is a chronic disease.
  • the antibody that specifically binds CD38 competes for binding to CD38 with an antibody comprising a heavy chain variable region (VH) of SEQ ID NO: 4 and a light chain variable region (VL) of SEQ ID NO: 5.
  • VH heavy chain variable region
  • VL light chain variable region
  • the antibody that specifically binds CD38 binds at least to the region SKRNIQFSCKNIYR (SEQ ID NO: 2) and the region EKVQTLEAWVIHGG (SEQ ID NO: 3) of CD38 (SEQ ID NO: 1).
  • the antibody that specifically binds CD38 comprises a heavy chain complementarity determining region 1 (HCDR1), a HCDR2 and a HCDR3 amino acid sequences of SEQ ID NOs: 6, 7 and 8, respectively.
  • HCDR1 heavy chain complementarity determining region 1
  • HCDR2 heavy chain complementarity determining region 2
  • HCDR3 amino acid sequences of SEQ ID NOs: 6, 7 and 8, respectively.
  • the antibody that specifically binds CD38 comprises a light chain complementarity determining region 1 (LCDR1), a LCDR2 and a LCDR3 amino acid sequences of SEQ ID NOs: 9, 10 and 11, respectively.
  • LCDR1 light chain complementarity determining region 1
  • LCDR2 LCDR2
  • LCDR3 amino acid sequences of SEQ ID NOs: 9, 10 and 11, respectively.
  • the antibody that specifically binds CD38 comprises the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2, and the LCDR3 amino acid sequences of SEQ ID NOs: 6, 7, 8, 9, 10 and 11, respectively.
  • the antibody that specifically binds CD38 comprises the VH that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 4 and the VL that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 5.
  • the antibody that specifically binds CD38 comprises the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5.
  • the antibody that specifically binds CD38 comprises a heavy chain that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 12 and a light chain that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 13.
  • the antibody that specifically binds CD38 comprises the heavy chain of SEQ ID NO: 12 and the light chain of SEQ ID NO: 13.
  • DARZALEXTM (daratumumab).
  • DARZALEXTM (daratumumab) comprises the heavy chain variable region (VH) and the light chain variable region (VL) amino acid sequences shown in SEQ ID NO: 4 and 5, respectively, heavy chain CDRs HCDR1, HCDR2 and HCDR3 of SEQ ID NOs: 6, 7 and 8, respectively, and light chain CDRs LCDR1, LCDR2 and LCDR3 of SEQ ID NOs: 9, 10 and 11, respectively, and is of IgG1/ ⁇ subtype and described in U.S. Pat. No. 7,829,673.
  • DARZALEXTM (daratumumab) heavy chain amino acid sequence is shown in SEQ ID NO: 12 and light chain amino acid sequence shown in SEQ ID NO: 13.
  • mAb003 comprising the VH and the VL sequences of SEQ ID NOs: 14 and 15, respectively and described in U.S. Pat. No. 7,829,673.
  • the VH and the VL of mAb003 may be expressed as IgG1/ ⁇ .
  • anti-CD38 antibodies that may be used in the methods of the invention include those described in Int. Pat. Publ. No. WO05/103083, Intl. Pat. Publ. No. WO06/125640, Intl. Pat. Publ. No. WO07/042309, Intl. Pat. Publ. No. WO08/047242 or Intl. Pat. Publ. No. WO14/178820.
  • the antibody that specifically binds CD38 is daratumumab or a biosimilar thereof.
  • Biosimilar (of an approved reference product/biological drug) refers to a biological product that is highly similar to the reference product notwithstanding minor differences in clinically inactive components with no clinically meaningful differences between the biosimilar and the reference product in terms of safety, purity and potency, based upon data derived from (a) analytical studies that demonstrate that the biological product is highly similar to the reference product notwithstanding minor differences in clinically inactive components; (b) animal studies (including the assessment of toxicity); and/or (c) a clinical study or studies (including the assessment of immunogenicity and pharmacokinetics or pharmacodynamics) that are sufficient to demonstrate safety, purity, and potency in one or more appropriate conditions of use for which the reference product is licensed and intended to be used and for which licensure is sought for the biosimilar.
  • the biosimilar may be an interchangeable product that may be substituted for the reference product at the pharmacy without the intervention of the prescribing healthcare professional.
  • the biosimilar is to be expected to produce the same clinical result as the reference product in any given patient and, if the biosimilar is administered more than once to an individual, the risk in terms of safety or diminished efficacy of alternating or switching between the use of the biosimilar and the reference product is not greater than the risk of using the reference product without such alternation or switch.
  • the biosimilar utilizes the same mechanisms of action for the proposed conditions of use to the extend the mechanisms are known for the reference product.
  • the condition or conditions of use prescribed, recommended, or suggested in the labeling proposed for the biosimilar have been previously approved for the reference product.
  • the route of administration, the dosage form, and/or the strength of the biosimilar are the same as those of the reference product and the biosimilar is manufactured, processed, packed or held in a facility that meets standards designed to assure that the biosimilar continues to be safe, pure and potent.
  • the biosimilar may include minor modifications in the amino acid sequence when compared to the reference product, such as N- or C-terminal truncations that are not expected to change the biosimilar performance.
  • the reference product may be approved in at least one of the U.S., Europe, or Japan.
  • the antibody that specifically binds CD38 is a non-agonistic antibody.
  • a non-agonistic antibody that specifically binds CD38 refers to an antibody which upon binding to CD38 does not induce significant proliferation of a sample of peripheral blood mononuclear cells in vitro when compared to the proliferation induced by an isotype control antibody or medium alone.
  • the non-agonistic antibody that specifically binds CD38 induces proliferation of peripheral blood mononuclear cells (PBMCs) in a statistically insignificant manner.
  • PBMC proliferation may be assessed by isolating PBMCs from healthy donors and culturing the cells at 1 ⁇ 10 5 cells/well in flat bottom 96-well plates in the presence or absence of a test antibody in 200 ⁇ l RPMI. After a four-day incubation at 37° C., 30 ⁇ l 3 H-thymidine (16.7 ⁇ Ci/ml) may be added, and culture may be continued overnight.
  • 3 H-thymidine incorporation may be assessed using a Packard Cobra gamma counter (Packard Instruments, Meriden, DT, USA), according to the manufacturer's instructions. Data may be calculated as the mean cpm ( ⁇ SEM) of PBMCs obtained from several donors. Statistical significance or insignificance between samples cultured in the presence or absence of the test antibody is calculated using standard methods.
  • Antibodies that specifically bind CD38 that can be used in the methods of the invention may also be selected de novo from, e.g., a phage display library, where the phage is engineered to express human immunoglobulins or portions thereof such as Fabs, single chain antibodies (scFv), or unpaired or paired antibody variable regions (Knappik et al., J Mol Biol 296:57-86, 2000; Krebs et al., J Immunol Meth 254:67-84, 2001; Vaughan et al., Nature Biotechnology 14:309-314, 1996; Sheets et al., PITAS (USA) 95:6157-6162, 1998; Hoogenboom and Winter, J Mol Biol 227:381, 1991; Marks et al., J Mol Biol 222:581, 1991).
  • a phage display library where the phage is engineered to express human immunoglobulins or portions thereof such as Fabs, single chain antibodies (sc
  • CD38 binding variable domains may be isolated from e.g., phage display libraries expressing antibody heavy and light chain variable regions as fusion proteins with bacteriophage pIX coat protein as described in Shi et al., J. Mol. Biol. 397:385-96, 2010 and Intl. Pat. Publ. No. WO09/085462).
  • the antibody libraries may be screened for binding to CD38 extracellular domain, the obtained positive clones further characterized, Fabs isolated from the clone lysates, and subsequently cloned as full length antibodies.
  • Such phage display methods for isolating human antibodies are established in the art. See for example: U.S. Pat. Nos.
  • Antibodies may be evaluated for their competition with a reference antibody, for example DARZALEXTM (daratumumab) having the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5 for binding to CD38 using well known in vitro methods.
  • a reference antibody for example DARZALEXTM (daratumumab) having the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5 for binding to CD38 using well known in vitro methods.
  • CHO cells recombinantly expressing CD38 may be incubated with an unlabeled reference antibody for 15 min at 4° C., followed by incubation with an excess of a fluorescently labeled test antibody for 45 min at 4° C. After washing in PBS/BSA, fluorescence may be measured by flow cytometry using standard methods.
  • the extracellular domain of CD38 may be coated on the surface of an ELISA plate.
  • HRP horseradish peroxidase
  • test antibody competes with the reference antibody when the reference antibody inhibits binding of the test antibody, or the test antibody inhibits binding of the reference antibody to CD38 by at least 80%, for example 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%.
  • the epitope of the test antibody may further be defined for example by peptide mapping or hydrogen/deuterium protection assays using known methods, or by crystal structure determination.
  • Antibodies binding to the region SKRNIQFSCKNIYR (SEQ ID NO: 2) and the region EKVQTLEAWVIHGG (SEQ ID NO: 3) of CD38 (SEQ ID NO: 1) may be generated for example by immunizing mice with peptides having the amino acid sequences shown in SEQ ID NOs: 2 and 3 using standard methods and those described herein, and characterizing the obtained antibodies for binding to the peptides using for example known ELISA or mutagenesis studies.
  • Antibodies that are substantially identical to the antibody comprising the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5 may be used in the methods of the invention.
  • the term “substantially identical” as used herein means that the antibody VH or VL amino acid sequences being compared are identical or have “insubstantial differences”. Insubstantial differences are substitutions of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in the antibody VL and/or VL that do not adversely affect antibody properties. Percent identity may be determined for example by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9.0.0 (Invitrogen, Carlsbad, Calif.).
  • the protein sequences of the present invention may be used as a query sequence to perform a search against public or patent databases to, for example, identify related sequences.
  • Exemplary programs used to perform such searches are the XBLAST or BLASTP programs (http_//www_ncbi_nlm/nih_gov), or the GenomeQuestTM (GenomeQuest, Westborough, Mass.) suite using the default settings.
  • Exemplary substitutions that may be made to the antibodies that specifically bind CD38 used in the methods of the invention are for example conservative substitutions with an amino acid having similar charge, hydrophobic, or stereochemical characteristics. Conservative substitutions may also be made to improve antibody properties, for example stability or affinity, or to improve antibody effector functions.
  • 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acid substitutions may be made for example to the heavy or the light chain of the antibodies that specifically binds CD38.
  • any native residue in the heavy or light chain may also be substituted with alanine, as has been previously described for alanine scanning mutagenesis (MacLennan et al., Acta Physiol Scand Suppl 643:55-67, 1998; Sasaki et al., Adv Biophys 35:1-24, 1998). Desired amino acid substitutions may be determined by those skilled in the art at the time such substitutions are desired. Amino acid substitutions may be done for example by PCR mutagenesis (U.S. Pat. No. 4,683,195).
  • variants may be generated using well known methods, for example using random (NNK) or non-random codons, for example DVK codons, which encode 11 amino acids (Ala, Cys, Asp, Glu, Gly, Lys, Asn, Arg, Ser, Tyr, Trp) and screening the libraries for variants with desired properties.
  • the generated variants may be tested for their binding to CD38, their ability to induce ADCC, ADCP or apoptosis, or modulate CD38 enzymatic activity in vitro using methods described herein.
  • Constant modifications refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequences.
  • Conservative modifications include amino acid substitutions, additions and deletions.
  • Conservative substitutions are those in which the amino acid is replaced with an amino acid residue having a similar side chain.
  • amino acids with acidic side chains e.g., aspartic acid, glutamic acid
  • basic side chains e.g., lysine, arginine, histidine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine, tryptophan
  • aromatic side chains e.g., phenylalanine, tryptophan, histidine, tyrosine
  • aliphatic side chains e.g., glycine, alanine, valine, leucine, isoleucine, serine, threonine
  • amide e.g., asparagine, glutamine
  • any native residue in the polypeptide may also be substituted with alanine, as has been previously described for alanine scanning mutagenesis (MacLennan et al., (1988) Acta Physiol Scand Suppl 643:55-67; Sasaki et al., (1988) Adv Biophys 35:1-24).
  • Amino acid substitutions to the antibodies of the invention may be made by known methods for example by PCR mutagenesis (U.S. Pat. No. 4,683,195).
  • libraries of variants may be generated for example using random (NNK) or non-random codons, for example DVK codons, which encode 11 amino acids (Ala, Cys, Asp, Glu, Gly, Lys, Asn, Arg, Ser, Tyr, Trp).
  • NNK random
  • DVK codons which encode 11 amino acids (Ala, Cys, Asp, Glu, Gly, Lys, Asn, Arg, Ser, Tyr, Trp).
  • the resulting antibody variants may be tested for their characteristics using assays described herein.
  • the antibody may bind CD38 with a dissociation constant (K D ) of less than about 1 ⁇ 10 ⁇ 7 M, 1 ⁇ 10 ⁇ 8 M, 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M, 1 ⁇ 10 ⁇ 11 M, 1 ⁇ 10 ⁇ 12 M, 1 ⁇ 10 ⁇ 13 M, 1 ⁇ 10 ⁇ 14 M or 1 ⁇ 10 ⁇ 15 , as determined by surface plasmon resonance or the Kinexa method, as practiced by those of skill in the art.
  • the antibody binds human CD38 with a K D of less than about 1 ⁇ 10 ⁇ 8 M.
  • the antibody binds human CD38 with a K D of less than about 1 ⁇ 10 ⁇ 9 M.
  • K D affinity and other binding parameters
  • K D affinity and other binding parameters
  • a standardized buffer such as the buffer described herein.
  • the internal error for affinity measurements for example using Biacore 3000 or ProteOn may typically be within 5-33% for measurements within the typical limits of detection. Therefore the term “about” in the context of K D reflects the typical standard deviation in the assay. For example, the typical SD for a K D of 1 ⁇ 10 ⁇ 9 M is up to ⁇ 0.33 ⁇ 10 ⁇ 9 M.
  • the antibody that specifically binds CD38 is an IgG1, IgG2, IgG3 or IgG4 isotype.
  • the antibody that specifically binds CD38 is an IgG1 isotype.
  • the antibody that specifically binds CD38 is an IgG2 isotype.
  • the antibody that specifically binds CD38 is an IgG3isotype.
  • the antibody that specifically binds CD38 is an IgG4 isotype.
  • the Fc portion of the antibody may mediate antibody effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) or complement dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • CDC complement dependent cytotoxicity
  • Such function may be mediated by binding of an Fc effector domain(s) to an Fc receptor on an immune cell with phagocytic or lytic activity or by binding of an Fc effector domain(s) to components of the complement system.
  • the effect(s) mediated by the Fc-binding cells or complement components result in inhibition and/or depletion of target cells, for example CD38-expressing cells.
  • Human IgG isotypes IgG1, IgG2, IgG3 and IgG4 exhibit differential capacity for effector functions.
  • ADCC may be mediated by IgG1 and IgG3
  • ADCP may be mediated by
  • Antibody-dependent cellular cytotoxicity is a mechanism for inducing cell death that depends upon the interaction of antibody-coated target cells with effector cells possessing lytic activity, such as natural killer cells, monocytes, macrophages and neutrophils via Fc gamma receptors (Fc ⁇ R) expressed on effector cells.
  • effector cells possessing lytic activity, such as natural killer cells, monocytes, macrophages and neutrophils via Fc gamma receptors (Fc ⁇ R) expressed on effector cells.
  • Fc ⁇ R Fc gamma receptors
  • NK cells express Fc ⁇ RIIIa
  • monocytes express Fc ⁇ RT, Fc ⁇ RII and Fc ⁇ RIIIa.
  • Death of the antibody-coated target cell such as CD38-expressing cells, occurs as a result of effector cell activity through the secretion of membrane pore-forming proteins and proteases.
  • the antibody may be added to CD38-expressing cells in combination with immune effector cells, which may be activated by the antigen antibody complexes resulting in cytolysis of the target cell. Cytolysis is generally detected by the release of label (e.g. radioactive substrates, fluorescent dyes or natural intracellular proteins) from the lysed cells.
  • label e.g. radioactive substrates, fluorescent dyes or natural intracellular proteins
  • exemplary effector cells for such assays include peripheral blood mononuclear cells (PBMC) and NK cells.
  • PBMC peripheral blood mononuclear cells
  • target cells include B cells expressing CD38.
  • target cells are labeled with 20 ⁇ Ci of 51 Cr for 2 hours and washed extensively.
  • Cell concentration of the target cells may be adjusted to 1 ⁇ 10 6 cells/ml, and antibodies specifically binding CD38 at various concentrations are added.
  • Assays are started by adding target cells at an effector:target cell ratio of 40:1. After incubation for 3 hr at 37° C. assays are stopped by centrifugation, and 51 Cr release from lysed cells are measured in a scintillation counter. Percentage of cellular cytotoxicity may be calculated as % maximal lysis which may be induced by adding 3% perchloric acid to target cells.
  • ADCP antibody-dependent cellular phagocytosis
  • CD38-positive cells engineered to express GFP or other labeled molecule as target cells.
  • Effctor:target cell ratio may be for example 4:1.
  • Effector cells may be incubated with target cells for 4 hours with or without antibodies that specifically bind CD38. After incubation, cells may be detached using accutase. Macrophages may be identified with anti-CD11b and anti-CD14 antibodies coupled to a fluorescent label, and percent phagocytosis may be determined based on % GFP fluorescent in the CD11 + CD14 + macrophages using standard methods.
  • “Complement-dependent cytotoxicity”, or “CDC”, refers to a mechanism for inducing cell death in which an Fc effector domain of a target-bound antibody binds and activates complement component C1q which in turn activates the complement cascade leading to target cell death. Activation of complement may also result in deposition of complement components on the target cell surface that facilitate ADCC by binding complement receptors (e.g., CR3) on leukocytes.
  • complement receptors e.g., CR3
  • the ability of monoclonal antibodies to induce ADCC may be enhanced by engineering their oligosaccharide component.
  • Human IgG1 or IgG3 are N-glycosylated at Asn297 with most the glycans in the well-known biantennary G0, G0F, G1, G1F, G2 or G2F forms.
  • Antibodies produced by non-engineered CHO cells typically have a glycan fucose content of about at least 85%. The removal of the core fucose from the biantennary complex-type oligosaccharides attached to the Fc regions enhances the ADCC of antibodies via improved Fc ⁇ RIIIa binding without altering antigen binding or CDC activity.
  • Such mAbs may be achieved using different methods reported to lead to the successful expression of relatively high defucosylated antibodies bearing the biantennary complex-type of Fc oligosaccharides such as control of culture osmolality (Konno et al., Cytotechnology 64:249-65, 2012), application of a variant CHO line Lec13 as the host cell line (Shields et al., J Biol Chem 277:26733-26740, 2002), application of a variant CHO line EB66 as the host cell line (Olivier et al., MAbs; 2(4), 2010; Epub ahead of print; PMID:20562582), application of a rat hybridoma cell line YB2/0 as the host cell line (Shinkawa et al., J Biol Chem 278:3466-3473, 2003), introduction of small interfering RNA specifically against the ⁇ 1,6-fucosyltrasferase (FUT8) gene (Mori
  • ADCC elicited by anti-CD38 antibodies used in the methods of the invention may also be enhanced by certain substitutions in the antibody Fc.
  • Exemplary substitutions are for example substitutions at amino acid positions 256, 290, 298, 312, 356, 330, 333, 334, 360, 378 or 430 (residue numbering according to the EU index) as described in U.S. Pat. No. 6,737,056.
  • the antibody that specifically binds CD38 comprises a substitution in the antibody Fc.
  • the antibody that specifically binds CD38 comprises a substitution in the antibody Fc at amino acid positions 256, 290, 298, 312, 356, 330, 333, 334, 360, 378 or 430 (residue numbering according to the EU index).
  • the antibody that specifically binds CD38 has a biantennary glycan structure with fucose content of about between 0% to about 15%, for example 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or 0%.
  • the antibody that specifically binds CD38 has a biantennary glycan structure with fucose content of about 50%, 40%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or 0%
  • “Fucose content” means the amount of the fucose monosaccharide within the sugar chain at Asn297.
  • the relative amount of fucose is the percentage of fucose-containing structures related to all glycostructures. These may be characterized and quantified by multiple methods, for example: 1) using MALDI-TOF of N-glycosidase F treated sample (e.g. complex, hybrid and oligo- and high-mannose structures) as described in Intl. Pat. Publ. No.
  • WO2008/077546 2) by enzymatic release of the Asn297 glycans with subsequent derivatization and detection/quantitation by HPLC (UPLC) with fluorescence detection and/or HPLC-MS (UPLC-MS); 3) intact protein analysis of the native or reduced mAb, with or without treatment of the Asn297 glycans with Endo S or other enzyme that cleaves between the first and the second G1cNAc monosaccharides, leaving the fucose attached to the first G1cNAc; 4) digestion of the mAb to constituent peptides by enzymatic digestion (e.g., trypsin or endopeptidase Lys-C), and subsequent separation, detection and quantitation by HPLC-MS (UPLC-MS) or 5) separation of the mAb oligosaccharides from the mAb protein by specific enzymatic deglycosylation with PNGase F at Asn 297.
  • UPLC UPLC
  • the oligosaccharides released may be labeled with a fluorophore, separated and identified by various complementary techniques which allow: fine characterization of the glycan structures by matrix-assisted laser desorption ionization (MALDI) mass spectrometry by comparison of the experimental masses with the theoretical masses, determination of the degree of sialylation by ion exchange HPLC (GlycoSep C), separation and quantification of the oligosacharride forms according to hydrophilicity criteria by normal-phase HPLC (GlycoSep N), and separation and quantification of the oligosaccharides by high performance capillary electrophoresis-laser induced fluorescence (HPCE-LIF).
  • MALDI matrix-assisted laser desorption ionization
  • Low fucose or “low fucose content” as used herein refers to antibodies with fucose content of about 0% -15%.
  • Normal fucose or ‘normal fucose content” as used herein refers to antibodies with fucose content of about over 50%, typically about over 60%, 70%, 80% or over 85%.
  • the antibody that specifically binds CD38 used in the methods of the invention may induce killing of CD38-expressing IgE producing cells by apoptosis.
  • Methods for evaluating apoptosis are well known, and include for example annexin IV staining using standard methods.
  • the antibodies that specifically bind human CD38 may induce apoptosis in about 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% of cells.
  • the antibody that specifically binds human CD38 is a bispecific antibody.
  • the VL and/or the VH regions of the existing antibodies that specifically bind CD38 or the VL and VH regions identified de novo as described herein may be engineered into bispecific full length antibodies.
  • Such bispecific antibodies may be made by modulating the CH3 interactions between the monospecific antibody heavy chains to form bispecific antibodies using technologies such as those described in U.S. Pat. No. 7,695,936; Intl. Pat. Publ. No. WO04/111233; U.S. Pat. Publ. No. US2010/0015133; U.S. Pat. Publ. No. US2007/0287170; Intl. Pat. Publ. No.
  • WO2009/134776 or structures that include various dimerization domains to connect the two antibody arms with different specificity, such as leucine zipper or collagen dimerization domains (Int. Pat. Publ. No. WO2012/022811, U.S. Pat. Nos. 5,932,448; 6,833,441).
  • bispecific antibodies may be generated in vitro in a cell-free environment by introducing asymmetrical mutations in the CH3 regions of two monospecific homodimeric antibodies and forming the bispecific heterodimeric antibody from two parental monospecific homodimeric antibodies in reducing conditions to allow disulfide bond isomerization according to methods described in Intl. Pat. Publ. No. WO2011/131746.
  • the first monospecific bivalent antibody e.g., anti-CD38 antibody
  • the second monospecific bivalent antibody are engineered to have certain substitutions at the CH3 domain that promote heterodimer stability; the antibodies are incubated together under reducing conditions sufficient to allow the cysteines in the hinge region to undergo disulfide bond isomerization; thereby generating the bispecific antibody by Fab arm exchange.
  • the incubation conditions may optimally be restored to non-reducing.
  • Exemplary reducing agents that may be used are 2-mercaptoethylamine (2-MEA), dithiothreitol (DTT), dithioerythritol (DTE), glutathione, tris(2-carboxyethyl) phosphine (TCEP), L-cysteine and beta-mercaptoethanol, preferably a reducing agent selected from the group consisting of: 2-mercaptoethylamine, dithiothreitol and tris(2-carboxyethyl)phosphine.
  • a reducing agent selected from the group consisting of: 2-mercaptoethylamine, dithiothreitol and tris(2-carboxyethyl)phosphine preferably incubation for at least 90 min at a temperature of at least 20° C. in the presence of at least 25 mM 2-MEA or in the presence of at least 0.5 mM dithiothreitol at a pH of from 5-8, for
  • Exemplary CH3 mutations that may be used in a first heavy chain and in a second heavy chain of the bispecific antibody are K409R and/or F405L.
  • the methods of the invention may be used to treat an animal patient belonging to any classification.
  • animals include mammals such as humans, rodents, dogs, cats and farm animals.
  • the antibodies that specifically bind CD38 may be provided in suitable pharmaceutical compositions comprising the antibody and a pharmaceutically acceptable carrier.
  • the carrier may be diluent, adjuvant, excipient, or vehicle with which the antibodies that specifically bind CD38 are administered.
  • vehicles may be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. For example, 0.4% saline and 0.3% glycine may be used. These solutions are sterile and generally free of particulate matter. They may be sterilized by conventional, well-known sterilization techniques (e.g., filtration).
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc.
  • concentration of the molecules or antibodies of the invention in such pharmaceutical formulation may vary widely, i.e., from less than about 0.5%, usually to at least about 1% to as much as 15 or 20%, 25%, 30%, 35%, 40%, 45% or 50% by weight and will be selected primarily based on required dose, fluid volumes, viscosities, etc., according to the particular mode of administration selected.
  • Suitable vehicles and formulations, inclusive of other human proteins, e.g., human serum albumin are described, for example, in e.g. Remington: The Science and Practice of Pharmacy, 21 st Edition, Troy, D. B. ed., Lipincott Williams and Wilkins, Philadelphia, Pa. 2006, Part 5, Pharmaceutical Manufacturing pp 691-1092, see especially pp. 958-989.
  • the mode of administration of the antibodies that specifically bind CD38 may be any suitable route such as parenteral administration, e.g., intradermal, intramuscular, intraperitoneal, intravenous or subcutaneous, pulmonary, transmucosal (oral, intranasal, intravaginal, rectal) or other means appreciated by the skilled artisan, as well known in the art.
  • parenteral administration e.g., intradermal, intramuscular, intraperitoneal, intravenous or subcutaneous, pulmonary, transmucosal (oral, intranasal, intravaginal, rectal) or other means appreciated by the skilled artisan, as well known in the art.
  • the antibodies that specifically bind CD38 may be administered to a subject by any suitable route, for example parentally by intravenous (i.v.) infusion or bolus injection, intramuscularly or subcutaneously or intraperitoneally.
  • i.v. infusion may be given over for example 15, 30, 60, 90, 120, 180, or 240 minutes, or from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 hours.
  • the dose given to a subject is sufficient to alleviate or at least partially arrest the disease being treated (“therapeutically effective amount”) and may be sometimes 0.005 mg to about 100 mg/kg, e.g. about 0.05 mg to about 30 mg/kg or about 5 mg to about 25 mg/kg, or about 4 mg/kg, about 8 mg/kg, about 16 mg/kg or about 24 mg/kg, or for example about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mg/kg, but may even higher, for example about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 40, 50, 60, 70, 80, 90 or 100 mg/kg.
  • therapeutically effective amount e.g. about 0.05 mg to about 30 mg/kg or about 5 mg to about 25 mg/kg, or about 4 mg/kg, about 8 mg/kg, about 16 mg/kg or about 24 mg/kg, or for example about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mg/kg, but may even higher, for example about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 40, 50, 60, 70, 80
  • a fixed unit dose may also be given, for example, 50, 100, 200, 500 or 1000 mg, or the dose may be based on the patient's surface area, e.g., 500, 400, 300, 250, 200, or 100 mg/m 2 .
  • 1 and 8 doses e.g., 1, 2, 3, 4, 5, 6, 7 or 8
  • 9 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more doses may be given.
  • the administration of the antibodies that specifically bind CD38 in the methods of the invention may be repeated after one day, two days, three days, four days, five days, six days, one week, two weeks, three weeks, one month, five weeks, six weeks, seven weeks, two months, three months, four months, five months, six months or longer. Repeated courses of treatment are also possible, as is chronic administration.
  • the repeated administration may be at the same dose or at a different dose.
  • the antibodies that specifically bind CD38 may be administered at 8 mg/kg or at 16 mg/kg at weekly interval for 8 weeks, followed by administration at 8 mg/kg or at 16 mg/kg every two weeks for an additional 16 weeks, followed by administration at 8 mg/kg or at 16 mg/kg every four weeks by intravenous infusion.
  • the antibodies that specifically bind CD38 may be administered as maintenance therapy, such as, e.g., once a week for a period of 6 months or more.
  • the antibodies that specifically bind CD38 may be provided as a daily dosage in an amount of about 0.1-100 mg/kg, such as 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 after initiation of treatment, or any combination thereof, using single or divided doses of every 24, 12, 8, 6, 4, or 2 hours, or any combination thereof.
  • 0.1-100 mg/kg such as 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,
  • the antibodies that specifically bind CD38 may be lyophilized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional protein preparations and well known lyophilization and reconstitution techniques can be employed.
  • the antibodies that specifically bind CD38 may be administered prophylactically to reduce the risk of developing an IgE-mediated disease.
  • the antibodies that specifically bind CD38 may also be administered within seconds, minutes, or hours following a subject's exposure to an allergen, or when at least one symptom of the IgE-mediated disease is present in a subject.
  • the methods used herein are useful in the treatment of both an acute exposure to an allergen and chronic (e.g., seasonal) exposure to an allergen.
  • the antibodies that specifically bind CD38 may be administered in combination with a second therapeutic agent.
  • the second therapeutic agent may be a standard of care treatment for the IgE-mediated disease, such as allergy, allergic asthma, urticarial, angioedema, autoimmune and inflammatory diseases.
  • compositions comprising an Antibody that Specifically Binds CD38 and a Hyaluronidase
  • the antibody that specifically binds CD38 may be administered as a pharmaceutical composition comprising the antibody that specifically binds CD38 and a hyaluronidase subcutaneously.
  • the concentration of the antibody that specifically binds CD38 in the pharmaceutical composition administered subcutaneously may be about 20 mg/ml.
  • the pharmaceutical composition administered subcutaneously may comprise between about 1,200 mg-1,800 mg of the antibody that specifically binds CD38.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,200 mg of the antibody that specifically binds CD38.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,600 mg of the antibody that specifically binds CD38.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,800 mg of the antibody that specifically binds CD38.
  • the pharmaceutical composition administered subcutaneously may comprise between about 30,000 U-45,000 U of the hyaluronidase.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,200 mg of the antibody that specifically binds CD38and about 30,000 U of the hyaluronidase.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,800 mg of the antibody that specifically binds CD38 and about 45,000 U of the hyaluronidase.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 30,000 U of the hyaluronidase.
  • the pharmaceutical composition administered subcutaneously may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 45,000 U of the hyaluronidase.
  • the pharmaceutical composition administered subcutaneously may comprise the hyaluronidase rHuPH20 having the amino acid sequence of SEQ ID NO: 22.
  • rHuPH20 is a recombinant hyaluronidase (HYLENEX® recombinant) and is described in Int. Pat. Publ. No. WO2004/078140.
  • Hyaluronidase is an enzyme that degrades hyaluronic acid (EC 3.2.1.35) and lowers the viscosity of hyaluronan in the extracellular matrix, thereby increasing tissue permeability.
  • the administration of the pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be repeated after one day, two days, three days, four days, five days, six days, one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, two months, three months, four months, five months, six months or longer. Repeated courses of treatment are also possible, as is chronic administration. The repeated administration may be at the same dose or at a different dose.
  • the pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be administered once weekly for eight weeks, followed by once in two weeks for 16 weeks, followed by once in four weeks.
  • the pharmaceutical compositions to be administered may comprise about 1,200 mg of the antibody that specifically binds CD38 and about 30,000 U of hyaluronidase, wherein the concentration of the antibody that specifically binds CD38 in the pharmaceutical composition is about 20 mg/ml.
  • the pharmaceutical compositions to be administered may comprise about 1,800 mg of the antibody that specifically binds CD38 and about 45,000 U of hyaluronidase.
  • the pharmaceutical compositions to be administered may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 30,000 U of hyaluronidase.
  • the pharmaceutical compositions to be administered may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 45,000 U of hyaluronidase.
  • the pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be administered subcutaneously to the abdominal region.
  • the pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be administered in a total volume of about 80 ml, 90 ml, 100 ml, 110 ml or 120 ml.
  • 20 mg/ml of the antibody that specifically binds CD38 antibody in 25 mM sodium acetate, 60 mM sodium chloride, 140 mM D-mannitol, 0.04% polysorbate 20, pH 5.5 may be mixed with rHuPH20, 1.0 mg/mL (75-150 kU/mL) in 10 mM L-Histidine, 130 mM NaCl, 10 mM L-Methionine, 0.02% Polysorbate 80, pH 6.5 prior to administration of the mixture to a subject.
  • DARZALEXTM farnesoidumab
  • This pilot study could demonstrate the potential value of daratumumab in management of patients with severe IgE-mediated disease.
  • Residual blood samples were collected from patients with multiple myeloma (MM) treated with daratumumab monotherapy or daratumumab plus lenalidomide-dexamethsone. Routine blood analysis was performed at baseline and after every four weeks of daratumumab treatment.
  • sIgE Determination of sIgE was performed by the ImmunoCAP technique for both the Phadiatop as the specific/total IgE according to the manufacturer's instructions (Thermo Fisher Scientific, Uppsala Sweden). This technique uses allergens covalently coupled to a solid phase matrix consisting of a cellulose derivative enclosed in a capsule. Analysis of binding of sIgE to the coated allergens is quantified by enzyme-labelled anti-IgE using fluorescence as read-out. sIgE levels ⁇ 0.35 kU/L were defined as positive test results.
  • IgE depletion by omalizumab treatment resulted in clinical improvement of disease, as well as increased quality of life and reduced socio-economic burden of disease, both in clinical trials as well as daily practice studies (Abraham I et al., Allergy 2015:n/a-n/a. doi:10.1111/a11.12815).
  • the effect of plasma cell depletion by daratumumab on clinical parameters of allergy has yet to be investigated.
  • grass timothy grass pollen, HDM: house dust mite Reference values: Total IgE: ⁇ 100 kU/L; ImmunoCAP sIgE birch/tim. grass/hdm: ⁇ 0.35 kU/L *Patient was enrolled in the GEN503 study and received daratumumab 16 mg/kg and lenalidomide 25 mg. **Patient was in the GEN501 phase II study and received daratumumab 16 mg/kg.
  • Tim. grass timothy grass pollen
  • HDM house dust mite
  • ImmunoCAP sIgE tim. grass/hdm ⁇ 0.35 kU/L

Abstract

The present invention relates to treatment of Ig E-mediated disease with antibodies that specifically bind CD38.

Description

    SEQUENCE LISTING
  • This application contains a Sequence Listing submitted via EFS-Web, the entire content of which incorporated herein by reference in its entirety. The ASCII text file, created on 20 Jun. 2017, is named JBI5091WOPCT_ST25.txt and is 26 kilobytes in size.
  • FIELD OF THE INVENTION
  • The present invention relates to treatment of IgE-mediated disease with antibodies that specifically bind CD38.
  • BACKGROUND OF THE INVENTION
  • A large and increasing proportion of the population in industrialized countries suffer from allergies. The current estimate for this debilitating condition is one in three people and a large proportion of this population is notably children. The pathogenesis of allergy is mediated by dysregulated triggering of IgE-mediated immune responses following repeated encounters with environmental antigens. IgE-mediated allergies are triggered by binding of IgE to the high affinity IgE receptor (FcϵRI), which is expressed on effector mast cells, basophils and activated eosinophils. As a result of these high affinity interactions, stable FcϵRI:IgE complexes are displayed on the surface of effector cells. Exposure to allergens leads to cross-linking and eventually clustering of IgE:FcϵRI complexes, thus triggering effector cell activation, degranulation and release of stored pro-allergenic mediators that leads to the initiation of an allergic response.
  • Common environmental allergens which induce anaphylactic hypersensitivity are found in pollen, foods, house dust mites, animal danders, fungal spores and insect venoms. Atopic allergy is associated with anaphylactic hypersensitivity and includes the disorders, e.g., asthma, allergic rhinitis and conjunctivitis (hay fever), eczema, urticaria and food allergies. Further, an allergic reaction may lead to a dangerous life-threatening condition such as anaphylactic shock, which may be provoked by insect bites.
  • For example, food allergy affects millions of people and is responsible for substantial morbidity, impaired quality of life and costs to the individual, family and society (Mills et al., Allergy 2007; 62:717-22. doi:10.1111/j.1398-9995.2007.01425.x.). Recent studies estimate that the prevalence of food allergy in the general population is around 5% for adults and 8% for children (Sicherer et al., J Allergy Clin Immunol 2014; 133:291-307.e5. doi:10.1016/j.jaci.2013.11.020.). The economic burden of food allergy in allergic children in the US are estimated $4184 per year per child (Gupta R et al., JAMA Pediatr 2013; 167:1026. doi:10.1001/jamapediatrics.2013.2376). Allergic reactions can vary from mild symptoms limited to the oral cavity and skin, to severe respiratory and cardiovascular symptoms that can be potentially fatal. Emergency management of food allergic reactions includes administration of epinephrine, corticosteroids and antihistamines. With no curative treatment available, strict avoidance of the eliciting allergens is often necessary. Still, accidental ingestion occurs, often leading to hospital admission and treatment in an intensive care unit.
  • In addition to allergies, IgE plays a role in autoimmune disorders contributing to their pathogenesis (Ettinger et al., Autoimmunity 50:25-35, 2017; Holgate, World Allergy Organization Journal 7:17, 2014).
  • A need exists for treatment options for IgE-mediated diseases such as allergies and autoimmune diseases.
  • SUMMARY OF THE INVENTION
  • The invention provides for a method of treating an IgE-mediated disease, comprising administering to a subject in need thereof an antibody that specifically binds CD38 for a time sufficient to treat the IgE-mediated disease.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows that total IgE and timothy grass and house dust mite-specific IgE is reduced in a multiple myeloma patient treated with DARZALEX™ (daratumumab) over time.
  • DETAILED DESCRIPTION OF THE INVENTION
  • “CD38” refers to the human CD38 protein (synonyms: ADP-ribosyl cyclase 1, cADPr hydrolase 1, cyclic ADP-ribose hydrolase 1). Human CD38 has an amino acid sequence shown in GenBank accession number NP_001766 and in SEQ ID NO: 1. It is well known that CD38 is a single pass type II membrane protein with amino acid residues 1-21 representing the cytosolic domain, amino acid residues 22-42 representing the transmembrane domain, and residues 43-300 representing the extracellular domain of CD38.
  • SEQ ID NO: 1
    MANCEFSPVSGDKPCCRLSRRAQLCLGVSILVLILVVVLAVVVPRWR
    QQWSGPGTTKRFPETVLARCVKYTEIHPEMRHVDCQSVWDAFKGAFI
    SKHPCNITEEDYQPLMKLGTQTVPCNKILLWSRIKDLAHQFTQVQRD
    MFTLEDTLLGYLADDLTWCGEFNTSKINYQSCPDWRKDCSNNPVSVF
    WKTVSRRFAEAACDVVHVMLNGSRSKIFDKNSTFGSVEVHNLQPEKV
    QTLEAWVIHGGREDSRDLCQDPTIKELESIISKRNIQFSCKNIYRPD
    KFLQCVKNPEDSSCTSEI
  • “Antibodies” as used herein is meant in a broad sense and includes immunoglobulin molecules including monoclonal antibodies including murine, human, humanized and chimeric monoclonal antibodies, antibody fragments, bispecific or multispecific antibodies, dimeric, tetrameric or multimeric antibodies, single chain antibodies, domain antibodies and any other modified configuration of the immunoglobulin molecule that comprises an antigen binding site of the required specificity.
  • Immunoglobulins may be assigned to five major classes, namely IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence. IgA and IgG are further sub-classified as the isotypes IgA1, IgA2, IgG1, IgG2, IgG3 and IgG4. Antibody light chains of any vertebrate species can be assigned to one of two clearly distinct types, namely kappa (κ) and lambda (λ), based on the amino acid sequences of their constant domains.
  • “Antibody fragments” refers to a portion of an immunoglobulin molecule that retains the heavy chain and/or the light chain antigen binding site, such as heavy chain complementarity determining regions (HCDR) 1, 2 and 3, light chain complementarity determining regions (LCDR) 1, 2 and 3, a heavy chain variable region (VH), or a light chain variable region (VL). Antibody fragments include a Fab fragment, a monovalent fragment consisting of the VL, VH, 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 domain antibody (dAb) fragment (Ward et al., Nature 341:544-6, 1989), which consists of a VH domain or a VL domain. VH and VL domains may be engineered and linked together via a synthetic linker to form various types of single chain antibody designs where the VH/VL domains pair intramolecularly, or intermolecularly in those cases when the VH and VL domains are expressed by separate single chain antibody constructs, to form a monovalent antigen binding site, such as single chain Fv (scFv) or diabody; described for example in Intl. Pat. Publ. Nos. WO1998/44001, WO1988/01649, WO1994/13804, and WO1992/01047. These antibody fragments are obtained using well known techniques known to those of skill in the art, and the fragments are screened for utility in the same manner as are full length antibodies.
  • “Isolated antibody” refers to an antibody or antibody fragment that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody specifically binding CD38 is substantially free of antibodies that specifically bind antigens other than human CD38). An isolated antibody that specifically binds CD38, however, may have cross-reactivity to other antigens, such as orthologs of human CD38, such as Macaca fascicularis (cynomolgus) CD38. In case of a bispecific antibody, the bispecific antibody specifically binds two antigens of interest, and is substantially free of antibodies that specifically bind antigens other that the two antigens of interest. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals. “Isolated antibody” encompasses antibodies that are isolated to a higher purity, such as antibodies that are 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% pure.
  • An antibody variable region consists of a “framework” region interrupted by three “antigen binding sites”. The antigen binding sites are defined using various terms: Complementarity Determining Regions (CDRs), three in the VH (HCDR1, HCDR2, HCDR3) and three in the VL (LCDR1, LCDR2, LCDR3) are based on sequence variability (Wu and Kabat J Exp Med 132:211-50, 1970; Kabat et al Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991); “Hypervariable regions”, “HVR”, or “HV”, three in the VH (H1, H2, H3) and three in the VL (L1, L2, L3) refer to the regions of antibody variable domains which are hypervariable in structure as defined by Chothia and Lesk (Chothia and Lesk Mol Biol 196:901-17, 1987). Other terms include “IMGT-CDRs” (Lefranc et al., Dev Comparat Immunol 27:55-77, 2003) and “Specificity Determining Residue Usage” (SDRU) (Almagro, Mol Recognit 17:132-43, 2004). The International ImMunoGeneTics (IMGT) database (http://www_imgt_org) provides a standardized numbering and definition of antigen-binding sites. The correspondence between CDRs, HVs and IMGT delineations is described in Lefranc et al., Dev Comparat Immunol 27:55-77, 2003.
  • “Chothia residues” as used herein are the antibody VL and VH residues numbered according to Al-Lazikani (Al-Lazikani et al., J Mol Biol 273:927-48, 1997).
  • “Framework” or “framework sequences” are the remaining sequences of a variable region other than those defined to be antigen binding sites. Because the antigen binding sites can be defined by various terms as described above, the exact amino acid sequence of a framework depends on how the antigen-binding site was defined.
  • “Humanized antibody” refers to an antibody in which the antigen binding sites are derived from non-human species and the variable region frameworks are derived from human immunoglobulin sequences. Humanized antibodies may include substitutions in the framework regions so that the framework may not be an exact copy of expressed human immunoglobulin or germline gene sequences.
  • “Human antibody” refers to an antibody having heavy and light chain variable regions in which both the framework and the antigen binding sites are derived from sequences of human origin and is optimized to have minimal immune response when administered to a human subject. If the antibody contains a constant region, the constant region also is derived from sequences of human origin.
  • A human antibody comprises heavy or light chain variable regions that are “derived from” sequences of human origin wherein the variable regions of the antibody are obtained from a system that uses human germline immunoglobulin or rearranged immunoglobulin genes. Such systems include human immunoglobulin gene libraries displayed on phage, and transgenic non-human animals such as mice carrying human immunoglobulin loci as described herein. A “human antibody” may contain amino acid differences when compared to the human germline immunoglobulin or rearranged immunoglobulin genes due to differences between the systems used to obtain the antibody and human immunoglobulin loci, introduction of somatic mutations or intentional introduction of substitutions in the framework or antigen binding site, or both. Typically, “human antibody” is at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical in amino acid sequence to an amino acid sequence encoded by human germline immunoglobulin or rearranged immunoglobulin genes. In some cases, “human antibody” may contain consensus framework sequences derived from human framework sequence analyses, for example as described in Knappik et al., J Mol Biol 296:57-86, 2000, or synthetic HCDR3 incorporated into human immunoglobulin gene libraries displayed on phage, for example as described in Shi et al., J Mol Biol 397:385-96, 2010 and Intl. Pat. Publ. No. WO2009/085462.
  • Human antibodies derived from human immunoglobulin sequences may be generated using systems such as phage display incorporating synthetic CDRs and/or synthetic frameworks, or can be subjected to in vitro mutagenesis to improve antibody properties, resulting in antibodies that do not naturally exist within the human antibody germline repertoire in vivo.
  • Antibodies in which antigen binding sites are derived from a non-human species are not included in the definition of human antibody.
  • “Recombinant antibody” includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse or a rat) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further below), antibodies isolated from a host cell transformed to express the antibody, antibodies isolated from a recombinant, combinatorial antibody library, and antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences, or antibodies that are generated in vitro using Fab arm exchange such as bispecific antibodies.
  • “Monoclonal antibody” refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope, or in a case of a bispecific monoclonal antibody, a dual binding specificity to two distinct epitopes. “Monoclonal antibody” therefore refers to an antibody population with single amino acid composition in each heavy and each light chain, except for possible well known alterations such as removal of C-terminal lysine from the antibody heavy chain or alterations due to post-translational modification(s) of amino acids, such as methionine oxidation or asparagine or glutamine deamidation. Monoclonal antibodies may have heterogeneous glycosylation within the antibody population. Monoclonal antibody may be monospecific or multispecific, or monovalent, bivalent or multivalent. A bispecific antibody is included in the term monoclonal antibody.
  • “Epitope” means a portion of an antigen to which an antibody specifically binds. Epitopes usually consist of chemically active (such as polar, non-polar or hydrophobic) surface groupings of moieties such as amino acids or polysaccharide side chains and may have specific three-dimensional structural characteristics, as well as specific charge characteristics. An epitope may be composed of contiguous and/or noncontiguous amino acids that form a conformational spatial unit. For a noncontiguous epitope, amino acids from differing portions of the linear sequence of the antigen come in close proximity in 3-dimensional space through the folding of the protein molecule.
  • “Variant” refers to a polypeptide or a polynucleotide that differs from a reference polypeptide or a reference polynucleotide by one or more modifications for example, substitutions, insertions or deletions.
  • “In combination with” means that two or more therapeutics are administered to a subject together in a mixture, concurrently as single agents or sequentially as single agents in any order. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • “Treat” or “treatment” refers to therapeutic treatment wherein the object is to slow down (lessen) an undesired physiological change or disease, or to provide a beneficial or desired clinical outcome during treatment. Beneficial or desired clinical outcomes include alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state. Those in need of treatment include those subjects already with the undesired physiological change or disease as well as those subjects prone to have the physiological change or disease.
  • “Therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result. A therapeutically effective amount may vary according to factors such as the disease severity, age, sex, and weight of the individual, and the ability of a therapeutic or a combination of therapeutics to elicit a desired response in the individual. Exemplary indicators of an effective therapeutic or combination of therapeutics include, for example, improved well-being of the subject, reduction in symptoms of the disease, such as reduction in sneezing, coughing, sinus congestion, mucus production in the sinuses (rhinitis) or lungs (asthma), itching, swelling, and/or decreased IgE levels in a subject.
  • “Patient” includes any human or nonhuman animal. “Nonhuman animal” includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc. The terms “patient” and “subject” are used interchangeably.
  • “Specific binding” or “specifically binds” or “binds” refers to an antibody binding to an antigen or an epitope within the antigen with greater affinity than for other antigens. Typically, the antibody binds to the antigen or the epitope within the antigen with an equilibrium dissociation constant (KD) of about 1×10−8 M or less, for example about 1×10−9 M or less, about 1×10−10 M or less, about 1×10−11 M or less, or about 1×10−12 M or less, typically with the KD that is at least one hundred-fold less than its KD for binding to a non-specific antigen (e.g., BSA, casein). The dissociation constant may be measured using standard procedures. Antibodies that specifically bind to the antigen or the epitope within the antigen may, however, have cross-reactivity to other related antigens, for example to the same antigen from other species (homologs), such as human or monkey, for example Macaca fascicularis (cynomolgus, cyno), Pan troglodytes (chimpanzee, chimp) or Callithrix jacchus (common marmoset, marmoset). While a monospecific antibody specifically binds one antigen or one epitope, a bispecific antibody specifically binds two distinct antigens or two distinct epitopes.
  • “An IgE-mediated disease” refers to a disease that is mediated, at least in part, by an increase in the level of IgE in a subject. “Increase in the level of IgE” refers to the level of total IgE of >_2 kU/L and/or the level of allergen specific IgE of ≥0.35 kU/L using ImmunoCAP assay (ThermoFisher, Uppsala, Sweden) using methodology described herein and according to the manufacturer's instructions. IgE-mediated diseases include disorders associated with increased IgE levels or activity in which atypical symptoms may manifest due to levels of IgE locally and/or systemically in the body even if the threshold of IgE of >2 kU/L is not achieved systemically.
  • The current invention is based, at least in part, on the identification that treatment of a subject with an antibody that specifically binds CD38 reduces total IgE and allergen-specific IgE in the subject over time. By not wishing to be bound by any particular theory, it is believed that the antibody that specifically binds CD38 mediates killing of the B cells expressing and/or secreting IgE.
  • Allergic diseases are conventionally described as IgE-mediated diseases. Clinical manifestations of allergic diseases include allergic asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and anaphylactic reactions to insect bites or drugs. Allergic diseases are caused by hypersensitivity reactions of the immune system to specific allergens (such as pollen, stings, drugs, or food). The most severe form of an allergic disorder is anaphylactic shock.
  • The specific immune response that defines allergy is characterized by the presence of immunoglobulin type E (IgE) directed to an allergen (sensitization). After sensitization, exposure to the allergen induces crosslinking of IgE bound to mast cells and basophils causing a wide release of vasoactive mediators, such as histamine. Therefore, therapy directed to IgE has been a research focus for several years. Therapy using monoclonal antibodies against IgE (Xolair® (omalizumab)) has proven to be effective in allergic asthma [4], with effectiveness also being researched in other (atopic) diseases, such as allergic rhinitis, and atopic dermatitis (Baena-Cagnani CE et al., Curr Opin Allergy Clin Immunol 2014; 14:149-54. doi:10.1097/ACI.0000000000000044). Furthermore, the role of Xolair® (omalizumab) in (oral) immunotherapy (IT) for food allergy is actively investigated, with first results demonstrating a decrease in adverse effects during treatment, similar to previous studies on IT for aeroallergens (Wood R A et al., J Allergy Clin Immunol 2015. doi:10.1016/j.jaci.2015.10.005). Another monoclonal antibody currently under investigation is quilizumab, which targets membrane IgE-expressing cells and thereby disrupting IgE production (Gauvreau G M et al., Sci Transl Med 2014; 6:243ra85. doi:10.1126/scitranslmed.3008961).
  • A different method of targeting IgE may be by depleting plasma cells that produce (specific) IgE. The effects of B-cell depletion on IgE levels and clinical symptoms of atopic diseases have been studied before. Rituxan® (rituximab), for example, did not significantly decrease serum IgE levels compared to placebo at three or six months in patients with Idiopathic Thrombocytopenic Purpura (Dasgupta A et al., Allergy Asthma Clin Immunol 2013; 9:39. doi:10.1186/1710-1492-9-39). VELCADE® (bortezomib), targeting (malignant) plasma cells via proteasome inhibition, is also being investigated as a treatment option in (auto) antibody mediated diseases (Rosenberg AS et al., Clin Immunol 2016. doi:10.1016/j.clim.2016.02.009). In a mouse model for chronic asthma, bortezomib treatment reduced specific IgE levels (Wegmann M et al., Int Arch Allergy Immunol 2012; 158:43-53. doi:10.1159/000330103).
  • The invention provides a method of treating an IgE-mediated disease, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody that specifically binds CD38 for a time sufficient to treat the IgE-mediated disease.
  • The invention also provides for an antibody that specifically binds CD38 for use in treating a subject having an IgE-mediated disease.
  • The invention also provides use of an antibody that specifically binds CD38 in the manufacture of a medicament for the treatment of an IgE-mediated disease.
  • The invention also provides use of an antibody that specifically binds CD38 in the preparation of a pharmaceutical composition for the treatment of an IgE-mediated disease.
  • In some embodiments, the IgE-mediated disease is an allergic response to an allergen.
  • Exemplary allergens include airborne allergens, such as those of house dust mite, pets and pollens, for example house dust mites allergens obtained from Dermatophagoides spp or D. pteronyssinus, D. farinae and D. microceras, Euroglyphus maynei or Blomia sp., allergens from insects present in cockroach or Hymenoptera, allergens from pollen, such as pollens of tree, grass and weed, allergens from animals, especially in cat, dog, horse and rodent, allergens from fungi, such as from Aspergillus, Altemaria or Cladosporium, and occupational allergens including animal and plant antigens as well as drugs, detergents, metals and immunoenhancers such as isocyanates and allergens present in products such as latex or amylase.
  • Exemplary allergens also include ingested allergens responsible for food hypersensitivity, such as fruits, vegetables and milk, such as food allergens present in peanuts, fish e.g. codfish, egg white, crustacean e.g. shrimp, milk e.g. cow's milk, wheat, cereals, fruits of the Rosacea family (apple, plum, strawberry), vegetables of the Liliacea, Cruciferae, Solanaceae and Umbelliferae families, tree nuts, sesame, peanut, soybean and other legume family allergens, spices, melon, avocado, mango, fig, banana.
  • Non-antigen specific stimuli that can result in an IgE-mediated reaction include infection, irritants such as smoke, combustion fumes, diesel exhaust particles and sulphur dioxide, exercise, cold and emotional stress.
  • Specific hypersensitivity reactions in atopic and nonatopic individuals with a certain genetic background may result from exposure to proteins in foods (e.g., legumes, peanuts), venom (e.g., insect, snake), vaccines, hormones, antiserum, enzymes, latex, antibiotics, muscle relaxants, vitamins, cytotoxins, opiates, and polysaccharides such as dextrin, iron dextran and polygeline.
  • In some embodiments, the allergen is pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
  • IgE has also been associated with pathogenic mechanisms of inflammation and autoimmunity. In addition to SLE and RA, IgE autoantibodies have been detected in dermatological autoimmune disorders such as bullous pemphigoid (BP) and chronic spontaneous urticaria (CSU), in systemic sclerosis, thyroiditis, multiple sclerosis and atopic dermatitis Immune complexes of IgE autoantibodies and autoantigens may trigger mast cell and basophil degranulation and induce IFNα, TNF and IL-6 production by dendritic cells (DC). IgE can promote antigen cross-presentation triggering both CD4 and CD8 T cell responses, and drive B cell expansion and plasma cell differentiation via DC activation. (reviewed in Ettinger et al., Autoimmunity 50:25-35, 2017; Holgate, World Allergy Organization Journal 7:17, 2014). Xolair® (Omalizumab), an anti-IgE antibody has been reported to exhibit efficacy or is being investigated in at least non-allergic asthma, Churg-Strauss Syndrome, allergic rhinitis, atopic dermatitis, nasal polyposis, food allergy, chronic urticaria and angioedema, Kimura's disease, mastocytosis, anaphylaxis, systemic lupus erythematosus, Sjogren's Syndrome (Holgate, World Allergy Organization Journal 7:17, 2014; ClinicalTrials registry). Therefore, it can be expected that an anti-CD38 antibody depleting IgE producing cells would be efficacious in these diseases.
  • IgE-mediated diseases include, asthma, atopic dermatitis, allergic rhinitis, fibrosis (e.g., pulmonary fibrosis, such as IPF), allergic asthma, food allergy, anaphylaxis, contact dermatitis, allergic gastroenteropathy, allergic bronchopulmonary aspergillosis, allergic purpura (Henoch-Schonlein), ataxia-telangiectasia, Churg-Strauss Syndrome, eczema, enteritis, gastroenteropathy, graft-versus-host reaction, hyper-IgE (Job's) syndrome, hypersensitivity (e.g., anaphylactic hypersensitivity, candidiasis, vasculitis), IgE myeloma, inflammatory bowel disease (e.g., Crohn's disease, ulcerative colitis, indeterminate colitis and infectious colitis), mucositis (e.g., oral mucositis, gastrointestinal mucositis, nasal mucositis and proctitis), necrotizing enterocolitis and esophagitis, parasitic diseases (e.g., trypanosomiasis), hypersensitivity vasculitis, urticaria, cholinergic urticaria, Wiskott-Aldrich syndrome, lupus, type I diabetes, Kimura's disease, nasal polyposis, eosinophilic gastroenteritis, eosinophilic otitis media, latex allergy, Sjogren's syndrome and rheumatoid arthritis, mastocytosis, cutaneous mastocytosis, chronic or recurrent idiopathic angioedema.
  • In some embodiments, lupus may be systemic lupus erythematosus (SLE), discoid lupus erythematosus, subacute cutaneous lupus erythematosus, neonatal lupus or drug-induced lupus.
  • Additionally, disorders that may be treatable by lowering IgE levels, regardless of whether the disorders themselves are associated with elevated IgE are within the scope of IgE-mediated disease.
  • In some embodiments, the IgE-mediated disease is allergic asthma.
  • In some embodiments, the IgE-mediated disease is urticaria.
  • In some embodiments, the IgE-mediated disease is angioedema.
  • In some embodiments, the IgE-mediated disease is food allergy.
  • In some embodiments, the IgE-mediated disease is atopic dermatitis.
  • In some embodiments, the IgE-mediated disease is anaphylaxis.
  • In some embodiments, the IgE-mediated disease is cutaneous mastocytosis.
  • In some embodiments, the IgE-mediated disease is allergic rhinitis.
  • In some embodiments, the IgE-mediated disease is nasal polyposis.
  • In some embodiments, the IgE-mediated disease is Kimura's disease.
  • In some embodiments, the IgE-mediated disease is eosinophilic otitis media.
  • In some embodiments, the IgE-mediated disease is eosinophilic gastroenteritis.
  • In some embodiments, the IgE-mediated disease is latex allergy.
  • In some embodiments, the IgE-mediated disease is bronchopulmonary allergic aspergillosis.
  • In some embodiments, the IgE-mediated disease is bullous pemphigoid (BP).
  • In some embodiments, the IgE-mediated disease is systemic lupus erythematosus (SLE).
  • In some embodiments, the IgE-mediated disease is rheumatoid arthritis (RA).
  • In some embodiments, the IgE-mediated disease is acute or chronic.
  • In some embodiments, the IgE-mediated disease is an acute disease.
  • In some embodiments, the IgE-mediated disease is a chronic disease.
  • In some embodiments, the antibody that specifically binds CD38 competes for binding to CD38 with an antibody comprising a heavy chain variable region (VH) of SEQ ID NO: 4 and a light chain variable region (VL) of SEQ ID NO: 5.
  • In some embodiments, the antibody that specifically binds CD38 binds at least to the region SKRNIQFSCKNIYR (SEQ ID NO: 2) and the region EKVQTLEAWVIHGG (SEQ ID NO: 3) of CD38 (SEQ ID NO: 1).
  • In some embodiments, the antibody that specifically binds CD38 comprises a heavy chain complementarity determining region 1 (HCDR1), a HCDR2 and a HCDR3 amino acid sequences of SEQ ID NOs: 6, 7 and 8, respectively.
  • In some embodiments, the antibody that specifically binds CD38 comprises a light chain complementarity determining region 1 (LCDR1), a LCDR2 and a LCDR3 amino acid sequences of SEQ ID NOs: 9, 10 and 11, respectively.
  • In some embodiments, the antibody that specifically binds CD38 comprises the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2, and the LCDR3 amino acid sequences of SEQ ID NOs: 6, 7, 8, 9, 10 and 11, respectively.
  • In some embodiments, the antibody that specifically binds CD38 comprises the VH that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 4 and the VL that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 5.
  • In some embodiments, the antibody that specifically binds CD38 comprises the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5.
  • In some embodiments, the antibody that specifically binds CD38 comprises a heavy chain that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 12 and a light chain that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 13.
  • In some embodiments, the antibody that specifically binds CD38 comprises the heavy chain of SEQ ID NO: 12 and the light chain of SEQ ID NO: 13.
  • An exemplary antibody that specifically binds CD38 that may be used in the methods of the invention is DARZALEX™ (daratumumab). DARZALEX™ (daratumumab) comprises the heavy chain variable region (VH) and the light chain variable region (VL) amino acid sequences shown in SEQ ID NO: 4 and 5, respectively, heavy chain CDRs HCDR1, HCDR2 and HCDR3 of SEQ ID NOs: 6, 7 and 8, respectively, and light chain CDRs LCDR1, LCDR2 and LCDR3 of SEQ ID NOs: 9, 10 and 11, respectively, and is of IgG1/κ subtype and described in U.S. Pat. No. 7,829,673. DARZALEX™ (daratumumab) heavy chain amino acid sequence is shown in SEQ ID NO: 12 and light chain amino acid sequence shown in SEQ ID NO: 13.
  • SEQ ID NO: 2
    SKRNIQFSCKNIYR
    SEQ ID NO: 3
    EKVQTLEAWVIHGG
    SEQ ID NO: 4
    EVQLLESGGGLVQPGGSLRLSCAVSGFTFNSFAMSWVRQAPGKGLEW
    VSAISGSGGGTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY
    FCAKDKILWFGEPVFDYWGQGTLVTVSS
    SEQ ID NO: 5
    EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLL
    IYDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNW
    PPTFGQGTKVEIK
    SEQ ID NO: 6
    SFAMS
    SEQ ID NO: 7
    AISGSGGGTYYADSVKG
    SEQ ID NO: 8
    DKILWFGEPVFDY
    SEQ ID NO: 9
    RASQSVSSYLA
    SEQ ID NO: 10
    DASNRAT
    SEQ ID NO: 11
    QQRSNWPPTF
    SEQ ID NO: 12
    EVQLLESGGGLVQPGGSLRLSCAVSGFTFNSFAMSWVRQAPGKGLEW
    VSAISGSGGGTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY
    FCAKDKILWFGEPVFDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS
    GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLS
    SVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCP
    APELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNW
    YVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVS
    NKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKG
    FYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ
    QGNVFSCSVMHEALHNHYTQKSLSLSPGK
    SEQ ID NO: 13
    EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLL
    IYDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNW
    PPTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYP
    REAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEK
    HKVYACEVTHQGLSSPVTKSFNRGEC
  • Other exemplary antibodies that specifically bind CD38 that may be used in the methods of the invention are:
  • mAb003 comprising the VH and the VL sequences of SEQ ID NOs: 14 and 15, respectively and described in U.S. Pat. No. 7,829,673. The VH and the VL of mAb003 may be expressed as IgG1/κ.
  • SEQ ID NO: 14
    QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAFSWVRQAPGQGLEW
    MGRVIPFLGIANSAQKFQGRVTITADKSTSTAYMDLSSLRSEDTAVY
    YCARDDIAALGPFDYWGQGTLVTVSSAS
    SEQ ID NO: 15
    DIQMTQSPSSLSASVGDRVTITCRASQGISSWLAWYQQKPEKAPKSL
    IYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQYNSY
    PRTFGQGTKVEIK

    mAb024 comprising the VH and the VL sequences of SEQ ID NOs: 16 and 17, respectively, described in U.S. Pat. No. 7,829,673. The VH and the VL of mAb024 may be expressed as IgG1/κ.
  • SEQ ID NO: 16
    EVQLVQSGAEVKKPGESLKISCKGSGYSFSNYWIGWVRQMPGKGLEW
    MGIIYPHDSDARYSPSFQGQVTFSADKSISTAYLQWSSLKASDTAMY
    YCARHVGWGSRYWYFDLWGRGTLVTVSS
    SEQ ID NO: 17
    EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPGLL
    IYDASNRASGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNW
    PLTFGGGTKVEIK

    MOR-202 (MOR-03087) comprising the VH and the VL sequences of SEQ ID NOs: 18 and 19, respectively, described in US. Pat. No. 8,088,896. The VH and the VL of MOR-202 may be expressed as IgG1/κ.
  • SEQ ID NO: 18
    QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYYMNWVRQAPGKGLEW
    VSGISGDPSNTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY
    YCARDLPLVYTGFAYWGQGTLVTVSS
    SEQ ID NO: 19
    DIELTQPPSVSVAPGQTARISCSGDNLRHYYVYWYQQKPGQAPVLVI
    YGDSKRPSGIPERFSGSNSGNTATLTISGTQAEDEADYYCQTYTGGA
    SLVFGGGTKLTVLGQ

    Isatuximab; comprising the VH and the VL sequences of SEQ ID NOs: 20 and 21, respectively, described in U.S. Pat. No. 8,153,765. The VH and the VL of Isatuximab may be expressed as IgG1/κ.
  • SEQ ID NO: 20
    QVQLVQSGAEVAKPGTSVKLSCKASGYTFTDYWMQWVKQRPGQGLEW
    IGTIYPGDGDTGYAQKFQGKATLTADKSSKTVYMHLSSLASEDSAVY
    YCARGDYYGSNSLDYWGQGTSVTVSS
    SEQ ID NO: 21:
    DIVMTQSHLSMSTSLGDPVSITCKASQDVSTVVAWYQQKPGQSPRRL
    IYSASYRYIGVPDRFTGSGAGTDFTFTISSVQAEDLAVYYCQQHYSP
    PYTFGGGTKLEIK
  • Other exemplary anti-CD38 antibodies that may be used in the methods of the invention include those described in Int. Pat. Publ. No. WO05/103083, Intl. Pat. Publ. No. WO06/125640, Intl. Pat. Publ. No. WO07/042309, Intl. Pat. Publ. No. WO08/047242 or Intl. Pat. Publ. No. WO14/178820.
  • In some embodiments, the antibody that specifically binds CD38 is daratumumab or a biosimilar thereof.
  • “Biosimilar” (of an approved reference product/biological drug) refers to a biological product that is highly similar to the reference product notwithstanding minor differences in clinically inactive components with no clinically meaningful differences between the biosimilar and the reference product in terms of safety, purity and potency, based upon data derived from (a) analytical studies that demonstrate that the biological product is highly similar to the reference product notwithstanding minor differences in clinically inactive components; (b) animal studies (including the assessment of toxicity); and/or (c) a clinical study or studies (including the assessment of immunogenicity and pharmacokinetics or pharmacodynamics) that are sufficient to demonstrate safety, purity, and potency in one or more appropriate conditions of use for which the reference product is licensed and intended to be used and for which licensure is sought for the biosimilar. The biosimilar may be an interchangeable product that may be substituted for the reference product at the pharmacy without the intervention of the prescribing healthcare professional. To meet the additional standard of “interchangeability,” the biosimilar is to be expected to produce the same clinical result as the reference product in any given patient and, if the biosimilar is administered more than once to an individual, the risk in terms of safety or diminished efficacy of alternating or switching between the use of the biosimilar and the reference product is not greater than the risk of using the reference product without such alternation or switch. The biosimilar utilizes the same mechanisms of action for the proposed conditions of use to the extend the mechanisms are known for the reference product. The condition or conditions of use prescribed, recommended, or suggested in the labeling proposed for the biosimilar have been previously approved for the reference product. The route of administration, the dosage form, and/or the strength of the biosimilar are the same as those of the reference product and the biosimilar is manufactured, processed, packed or held in a facility that meets standards designed to assure that the biosimilar continues to be safe, pure and potent. The biosimilar may include minor modifications in the amino acid sequence when compared to the reference product, such as N- or C-terminal truncations that are not expected to change the biosimilar performance. The reference product may be approved in at least one of the U.S., Europe, or Japan.
  • In some embodiments, the antibody that specifically binds CD38 is a non-agonistic antibody.
  • A non-agonistic antibody that specifically binds CD38 refers to an antibody which upon binding to CD38 does not induce significant proliferation of a sample of peripheral blood mononuclear cells in vitro when compared to the proliferation induced by an isotype control antibody or medium alone.
  • In some embodiments, the non-agonistic antibody that specifically binds CD38 induces proliferation of peripheral blood mononuclear cells (PBMCs) in a statistically insignificant manner. PBMC proliferation may be assessed by isolating PBMCs from healthy donors and culturing the cells at 1×105 cells/well in flat bottom 96-well plates in the presence or absence of a test antibody in 200 μl RPMI. After a four-day incubation at 37° C., 30 μl 3H-thymidine (16.7 μCi/ml) may be added, and culture may be continued overnight. 3H-thymidine incorporation may be assessed using a Packard Cobra gamma counter (Packard Instruments, Meriden, DT, USA), according to the manufacturer's instructions. Data may be calculated as the mean cpm (±SEM) of PBMCs obtained from several donors. Statistical significance or insignificance between samples cultured in the presence or absence of the test antibody is calculated using standard methods.
  • Antibodies that specifically bind CD38 that can be used in the methods of the invention may also be selected de novo from, e.g., a phage display library, where the phage is engineered to express human immunoglobulins or portions thereof such as Fabs, single chain antibodies (scFv), or unpaired or paired antibody variable regions (Knappik et al., J Mol Biol 296:57-86, 2000; Krebs et al., J Immunol Meth 254:67-84, 2001; Vaughan et al., Nature Biotechnology 14:309-314, 1996; Sheets et al., PITAS (USA) 95:6157-6162, 1998; Hoogenboom and Winter, J Mol Biol 227:381, 1991; Marks et al., J Mol Biol 222:581, 1991). CD38 binding variable domains may be isolated from e.g., phage display libraries expressing antibody heavy and light chain variable regions as fusion proteins with bacteriophage pIX coat protein as described in Shi et al., J. Mol. Biol. 397:385-96, 2010 and Intl. Pat. Publ. No. WO09/085462). The antibody libraries may be screened for binding to CD38 extracellular domain, the obtained positive clones further characterized, Fabs isolated from the clone lysates, and subsequently cloned as full length antibodies. Such phage display methods for isolating human antibodies are established in the art. See for example: U.S. Pat. Nos. 5,223,409, 5,403,484, 5,571,698, 5,427,908, 5,580,717, 5,969,108, 6,172,197, 5,885,793, 6,521,404, 6,544,731, 6,555,313, 6,582,915, and 6,593,081.
  • Antibodies may be evaluated for their competition with a reference antibody, for example DARZALEX™ (daratumumab) having the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5 for binding to CD38 using well known in vitro methods. In an exemplary method, CHO cells recombinantly expressing CD38 may be incubated with an unlabeled reference antibody for 15 min at 4° C., followed by incubation with an excess of a fluorescently labeled test antibody for 45 min at 4° C. After washing in PBS/BSA, fluorescence may be measured by flow cytometry using standard methods. In another exemplary method, the extracellular domain of CD38 may be coated on the surface of an ELISA plate. Excess of an unlabeled reference antibody may be added for about 15 minutes and subsequently a biotinylated test antibody may be added. After washes in PBS/Tween, binding of the biotinylated test antibody may be detected using horseradish peroxidase (HRP)-conjugated streptavidin and the signal detected using standard methods. It is readily apparent that in the competition assays, the reference antibody may be labelled and the test antibody unlabeled. The test antibody competes with the reference antibody when the reference antibody inhibits binding of the test antibody, or the test antibody inhibits binding of the reference antibody to CD38 by at least 80%, for example 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%. The epitope of the test antibody may further be defined for example by peptide mapping or hydrogen/deuterium protection assays using known methods, or by crystal structure determination.
  • Antibodies binding to the region SKRNIQFSCKNIYR (SEQ ID NO: 2) and the region EKVQTLEAWVIHGG (SEQ ID NO: 3) of CD38 (SEQ ID NO: 1) may be generated for example by immunizing mice with peptides having the amino acid sequences shown in SEQ ID NOs: 2 and 3 using standard methods and those described herein, and characterizing the obtained antibodies for binding to the peptides using for example known ELISA or mutagenesis studies.
  • Antibodies that are substantially identical to the antibody comprising the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5 may be used in the methods of the invention. The term “substantially identical” as used herein means that the antibody VH or VL amino acid sequences being compared are identical or have “insubstantial differences”. Insubstantial differences are substitutions of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in the antibody VL and/or VL that do not adversely affect antibody properties. Percent identity may be determined for example by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9.0.0 (Invitrogen, Carlsbad, Calif.). The protein sequences of the present invention may be used as a query sequence to perform a search against public or patent databases to, for example, identify related sequences. Exemplary programs used to perform such searches are the XBLAST or BLASTP programs (http_//www_ncbi_nlm/nih_gov), or the GenomeQuest™ (GenomeQuest, Westborough, Mass.) suite using the default settings. Exemplary substitutions that may be made to the antibodies that specifically bind CD38 used in the methods of the invention are for example conservative substitutions with an amino acid having similar charge, hydrophobic, or stereochemical characteristics. Conservative substitutions may also be made to improve antibody properties, for example stability or affinity, or to improve antibody effector functions. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acid substitutions may be made for example to the heavy or the light chain of the antibodies that specifically binds CD38. Furthermore, any native residue in the heavy or light chain may also be substituted with alanine, as has been previously described for alanine scanning mutagenesis (MacLennan et al., Acta Physiol Scand Suppl 643:55-67, 1998; Sasaki et al., Adv Biophys 35:1-24, 1998). Desired amino acid substitutions may be determined by those skilled in the art at the time such substitutions are desired. Amino acid substitutions may be done for example by PCR mutagenesis (U.S. Pat. No. 4,683,195). Libraries of variants may be generated using well known methods, for example using random (NNK) or non-random codons, for example DVK codons, which encode 11 amino acids (Ala, Cys, Asp, Glu, Gly, Lys, Asn, Arg, Ser, Tyr, Trp) and screening the libraries for variants with desired properties. The generated variants may be tested for their binding to CD38, their ability to induce ADCC, ADCP or apoptosis, or modulate CD38 enzymatic activity in vitro using methods described herein.
  • “Conservative modifications” refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequences. Conservative modifications include amino acid substitutions, additions and deletions. Conservative substitutions are those in which the amino acid is replaced with an amino acid residue having a similar side chain. The families of amino acid residues having similar side chains are well defined and include amino acids with acidic side chains (e.g., aspartic acid, glutamic acid), basic side chains (e.g., lysine, arginine, histidine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), uncharged polar side chains (e.g., glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine, tryptophan), aromatic side chains (e.g., phenylalanine, tryptophan, histidine, tyrosine), aliphatic side chains (e.g., glycine, alanine, valine, leucine, isoleucine, serine, threonine), amide (e.g., asparagine, glutamine), beta-branched side chains (e.g., threonine, valine, isoleucine) and sulfur-containing side chains (cysteine, methionine). Furthermore, any native residue in the polypeptide may also be substituted with alanine, as has been previously described for alanine scanning mutagenesis (MacLennan et al., (1988) Acta Physiol Scand Suppl 643:55-67; Sasaki et al., (1988) Adv Biophys 35:1-24). Amino acid substitutions to the antibodies of the invention may be made by known methods for example by PCR mutagenesis (U.S. Pat. No. 4,683,195). Alternatively, libraries of variants may be generated for example using random (NNK) or non-random codons, for example DVK codons, which encode 11 amino acids (Ala, Cys, Asp, Glu, Gly, Lys, Asn, Arg, Ser, Tyr, Trp). The resulting antibody variants may be tested for their characteristics using assays described herein.
  • In some embodiments, the antibody may bind CD38 with a dissociation constant (KD) of less than about 1×10−7 M, 1×10−8 M, 1×10−9 M, 1×10−10 M, 1×10−11 M, 1×10−12 M, 1×10−13 M, 1×10−14 M or 1×10−15, as determined by surface plasmon resonance or the Kinexa method, as practiced by those of skill in the art. In some embodiments, the antibody binds human CD38 with a KD of less than about 1×10−8 M. In some embodiments, the antibody binds human CD38 with a KD of less than about 1×10−9 M.
  • KinExA instrumentation, ELISA or competitive binding assays known to those skilled in the art. The measured affinity of a particular antibody/CD38 interaction may vary if measured under different conditions (e.g., osmolarity, pH). Thus, measurements of affinity and other binding parameters (e.g., KD, Kon, Koff) are typically made with standardized conditions and a standardized buffer, such as the buffer described herein. Skilled in the art will appreciate that the internal error for affinity measurements for example using Biacore 3000 or ProteOn (measured as standard deviation, SD) may typically be within 5-33% for measurements within the typical limits of detection. Therefore the term “about” in the context of KD reflects the typical standard deviation in the assay. For example, the typical SD for a KD of 1×10−9 M is up to ±0.33×10−9 M.
  • In some embodiments, the antibody that specifically binds CD38 is an IgG1, IgG2, IgG3 or IgG4 isotype.
  • In some embodiments, the antibody that specifically binds CD38 is an IgG1 isotype.
  • In some embodiments, the antibody that specifically binds CD38 is an IgG2 isotype.
  • In some embodiments, the antibody that specifically binds CD38 is an IgG3isotype.
  • In some embodiments, the antibody that specifically binds CD38 is an IgG4 isotype.
  • The Fc portion of the antibody may mediate antibody effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) or complement dependent cytotoxicity (CDC). Such function may be mediated by binding of an Fc effector domain(s) to an Fc receptor on an immune cell with phagocytic or lytic activity or by binding of an Fc effector domain(s) to components of the complement system. Typically, the effect(s) mediated by the Fc-binding cells or complement components result in inhibition and/or depletion of target cells, for example CD38-expressing cells. Human IgG isotypes IgG1, IgG2, IgG3 and IgG4 exhibit differential capacity for effector functions. ADCC may be mediated by IgG1 and IgG3, ADCP may be mediated by IgG 1, IgG2, IgG3 and IgG4, and CDC may be mediated by IgG1 and IgG3.
  • “Antibody-dependent cellular cytotoxicity”, “antibody-dependent cell-mediated cytotoxicity” or “ADCC” is a mechanism for inducing cell death that depends upon the interaction of antibody-coated target cells with effector cells possessing lytic activity, such as natural killer cells, monocytes, macrophages and neutrophils via Fc gamma receptors (FcγR) expressed on effector cells. For example, NK cells express FcγRIIIa, whereas monocytes express FcγRT, FcγRII and FcγRIIIa. Death of the antibody-coated target cell, such as CD38-expressing cells, occurs as a result of effector cell activity through the secretion of membrane pore-forming proteins and proteases. To assess ADCC activity of an antibody that specifically binds CD38, the antibody may be added to CD38-expressing cells in combination with immune effector cells, which may be activated by the antigen antibody complexes resulting in cytolysis of the target cell. Cytolysis is generally detected by the release of label (e.g. radioactive substrates, fluorescent dyes or natural intracellular proteins) from the lysed cells. Exemplary effector cells for such assays include peripheral blood mononuclear cells (PBMC) and NK cells. Exemplary target cells include B cells expressing CD38. In an exemplary assay, target cells are labeled with 20 μCi of 51Cr for 2 hours and washed extensively. Cell concentration of the target cells may be adjusted to 1×106 cells/ml, and antibodies specifically binding CD38 at various concentrations are added. Assays are started by adding target cells at an effector:target cell ratio of 40:1. After incubation for 3 hr at 37° C. assays are stopped by centrifugation, and 51Cr release from lysed cells are measured in a scintillation counter. Percentage of cellular cytotoxicity may be calculated as % maximal lysis which may be induced by adding 3% perchloric acid to target cells.
  • “Antibody-dependent cellular phagocytosis” (“ADCP”) refers to a mechanism of elimination of antibody-coated target cells by internalization by phagocytic cells, such as macrophages or dendritic cells. ADCP may be evaluated using CD38-positive cells engineered to express GFP or other labeled molecule as target cells. Effctor:target cell ratio may be for example 4:1. Effector cells may be incubated with target cells for 4 hours with or without antibodies that specifically bind CD38. After incubation, cells may be detached using accutase. Macrophages may be identified with anti-CD11b and anti-CD14 antibodies coupled to a fluorescent label, and percent phagocytosis may be determined based on % GFP fluorescent in the CD11+CD14+ macrophages using standard methods.
  • “Complement-dependent cytotoxicity”, or “CDC”, refers to a mechanism for inducing cell death in which an Fc effector domain of a target-bound antibody binds and activates complement component C1q which in turn activates the complement cascade leading to target cell death. Activation of complement may also result in deposition of complement components on the target cell surface that facilitate ADCC by binding complement receptors (e.g., CR3) on leukocytes.
  • The ability of monoclonal antibodies to induce ADCC may be enhanced by engineering their oligosaccharide component. Human IgG1 or IgG3 are N-glycosylated at Asn297 with most the glycans in the well-known biantennary G0, G0F, G1, G1F, G2 or G2F forms. Antibodies produced by non-engineered CHO cells typically have a glycan fucose content of about at least 85%. The removal of the core fucose from the biantennary complex-type oligosaccharides attached to the Fc regions enhances the ADCC of antibodies via improved FcγRIIIa binding without altering antigen binding or CDC activity. Such mAbs may be achieved using different methods reported to lead to the successful expression of relatively high defucosylated antibodies bearing the biantennary complex-type of Fc oligosaccharides such as control of culture osmolality (Konno et al., Cytotechnology 64:249-65, 2012), application of a variant CHO line Lec13 as the host cell line (Shields et al., J Biol Chem 277:26733-26740, 2002), application of a variant CHO line EB66 as the host cell line (Olivier et al., MAbs; 2(4), 2010; Epub ahead of print; PMID:20562582), application of a rat hybridoma cell line YB2/0 as the host cell line (Shinkawa et al., J Biol Chem 278:3466-3473, 2003), introduction of small interfering RNA specifically against the α 1,6-fucosyltrasferase (FUT8) gene (Mori et al., Biotechnol Bioeng88:901-908, 2004), or coexpression of β-1,4-N-acetylglucosaminyltransferase III and Golgi α-mannosidase II or a potent alpha-mannosidase I inhibitor, kifunensine (Ferrara et al., J Biol Chem 281:5032-5036, 2006, Ferrara et al., Biotechnol Bioeng 93:851-861, 2006; Xhou et al., Biotechnol Bioeng 99:652-65, 2008). ADCC elicited by anti-CD38 antibodies used in the methods of the invention may also be enhanced by certain substitutions in the antibody Fc. Exemplary substitutions are for example substitutions at amino acid positions 256, 290, 298, 312, 356, 330, 333, 334, 360, 378 or 430 (residue numbering according to the EU index) as described in U.S. Pat. No. 6,737,056.
  • In some embodiments, the antibody that specifically binds CD38 comprises a substitution in the antibody Fc.
  • In some embodiments, the antibody that specifically binds CD38 comprises a substitution in the antibody Fc at amino acid positions 256, 290, 298, 312, 356, 330, 333, 334, 360, 378 or 430 (residue numbering according to the EU index).
  • In some embodiments, the antibody that specifically binds CD38 has a biantennary glycan structure with fucose content of about between 0% to about 15%, for example 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or 0%.
  • In some embodiments, the antibody that specifically binds CD38 has a biantennary glycan structure with fucose content of about 50%, 40%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or 0%
  • Substitutions in the Fc and reduced fucose content may enhance the ADCC activity of the antibody that specifically binds CD38.
  • “Fucose content” means the amount of the fucose monosaccharide within the sugar chain at Asn297. The relative amount of fucose is the percentage of fucose-containing structures related to all glycostructures. These may be characterized and quantified by multiple methods, for example: 1) using MALDI-TOF of N-glycosidase F treated sample (e.g. complex, hybrid and oligo- and high-mannose structures) as described in Intl. Pat. Publ. No. WO2008/077546; 2) by enzymatic release of the Asn297 glycans with subsequent derivatization and detection/quantitation by HPLC (UPLC) with fluorescence detection and/or HPLC-MS (UPLC-MS); 3) intact protein analysis of the native or reduced mAb, with or without treatment of the Asn297 glycans with Endo S or other enzyme that cleaves between the first and the second G1cNAc monosaccharides, leaving the fucose attached to the first G1cNAc; 4) digestion of the mAb to constituent peptides by enzymatic digestion (e.g., trypsin or endopeptidase Lys-C), and subsequent separation, detection and quantitation by HPLC-MS (UPLC-MS) or 5) separation of the mAb oligosaccharides from the mAb protein by specific enzymatic deglycosylation with PNGase F at Asn 297. The oligosaccharides released may be labeled with a fluorophore, separated and identified by various complementary techniques which allow: fine characterization of the glycan structures by matrix-assisted laser desorption ionization (MALDI) mass spectrometry by comparison of the experimental masses with the theoretical masses, determination of the degree of sialylation by ion exchange HPLC (GlycoSep C), separation and quantification of the oligosacharride forms according to hydrophilicity criteria by normal-phase HPLC (GlycoSep N), and separation and quantification of the oligosaccharides by high performance capillary electrophoresis-laser induced fluorescence (HPCE-LIF).
  • “Low fucose” or “low fucose content” as used herein refers to antibodies with fucose content of about 0% -15%.
  • “Normal fucose” or ‘normal fucose content” as used herein refers to antibodies with fucose content of about over 50%, typically about over 60%, 70%, 80% or over 85%.
  • The antibody that specifically binds CD38 used in the methods of the invention may induce killing of CD38-expressing IgE producing cells by apoptosis. Methods for evaluating apoptosis are well known, and include for example annexin IV staining using standard methods. The antibodies that specifically bind human CD38 may induce apoptosis in about 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% of cells.
  • In some embodiments, the antibody that specifically binds human CD38 is a bispecific antibody. The VL and/or the VH regions of the existing antibodies that specifically bind CD38 or the VL and VH regions identified de novo as described herein may be engineered into bispecific full length antibodies. Such bispecific antibodies may be made by modulating the CH3 interactions between the monospecific antibody heavy chains to form bispecific antibodies using technologies such as those described in U.S. Pat. No. 7,695,936; Intl. Pat. Publ. No. WO04/111233; U.S. Pat. Publ. No. US2010/0015133; U.S. Pat. Publ. No. US2007/0287170; Intl. Pat. Publ. No. WO2008/119353; U.S. Pat. Publ. No. US2009/0182127; U.S. Pat. Publ. No. US2010/0286374; U.S. Pat. Publ. No. US2011/0123532; Intl. Pat. Publ. No. WO2011/131746; Int. Pat. Publ. No. WO2011/143545; or U.S. Pat. Publ. No. US2012/0149876. Additional bispecific structures into which the VL and/or the VH regions of the antibodies of the invention may be incorporated are for example Dual Variable Domain Immunoglobulins (Intl. Pat. Publ. No. WO2009/134776), or structures that include various dimerization domains to connect the two antibody arms with different specificity, such as leucine zipper or collagen dimerization domains (Int. Pat. Publ. No. WO2012/022811, U.S. Pat. Nos. 5,932,448; 6,833,441).
  • For example, bispecific antibodies may be generated in vitro in a cell-free environment by introducing asymmetrical mutations in the CH3 regions of two monospecific homodimeric antibodies and forming the bispecific heterodimeric antibody from two parental monospecific homodimeric antibodies in reducing conditions to allow disulfide bond isomerization according to methods described in Intl. Pat. Publ. No. WO2011/131746. In the methods, the first monospecific bivalent antibody (e.g., anti-CD38 antibody) and the second monospecific bivalent antibody are engineered to have certain substitutions at the CH3 domain that promote heterodimer stability; the antibodies are incubated together under reducing conditions sufficient to allow the cysteines in the hinge region to undergo disulfide bond isomerization; thereby generating the bispecific antibody by Fab arm exchange. The incubation conditions may optimally be restored to non-reducing. Exemplary reducing agents that may be used are 2-mercaptoethylamine (2-MEA), dithiothreitol (DTT), dithioerythritol (DTE), glutathione, tris(2-carboxyethyl) phosphine (TCEP), L-cysteine and beta-mercaptoethanol, preferably a reducing agent selected from the group consisting of: 2-mercaptoethylamine, dithiothreitol and tris(2-carboxyethyl)phosphine. For example, incubation for at least 90 min at a temperature of at least 20° C. in the presence of at least 25 mM 2-MEA or in the presence of at least 0.5 mM dithiothreitol at a pH of from 5-8, for example at pH of 7.0 or at pH of 7.4 may be used.
  • Exemplary CH3 mutations that may be used in a first heavy chain and in a second heavy chain of the bispecific antibody are K409R and/or F405L.
  • The methods of the invention may be used to treat an animal patient belonging to any classification. Examples of such animals include mammals such as humans, rodents, dogs, cats and farm animals.
  • Administration/Pharmaceutical Compositions
  • In the methods of the invention, the antibodies that specifically bind CD38 may be provided in suitable pharmaceutical compositions comprising the antibody and a pharmaceutically acceptable carrier. The carrier may be diluent, adjuvant, excipient, or vehicle with which the antibodies that specifically bind CD38 are administered. Such vehicles may be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. For example, 0.4% saline and 0.3% glycine may be used. These solutions are sterile and generally free of particulate matter. They may be sterilized by conventional, well-known sterilization techniques (e.g., filtration). The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc. The concentration of the molecules or antibodies of the invention in such pharmaceutical formulation may vary widely, i.e., from less than about 0.5%, usually to at least about 1% to as much as 15 or 20%, 25%, 30%, 35%, 40%, 45% or 50% by weight and will be selected primarily based on required dose, fluid volumes, viscosities, etc., according to the particular mode of administration selected. Suitable vehicles and formulations, inclusive of other human proteins, e.g., human serum albumin, are described, for example, in e.g. Remington: The Science and Practice of Pharmacy, 21st Edition, Troy, D. B. ed., Lipincott Williams and Wilkins, Philadelphia, Pa. 2006, Part 5, Pharmaceutical Manufacturing pp 691-1092, see especially pp. 958-989.
  • The mode of administration of the antibodies that specifically bind CD38 may be any suitable route such as parenteral administration, e.g., intradermal, intramuscular, intraperitoneal, intravenous or subcutaneous, pulmonary, transmucosal (oral, intranasal, intravaginal, rectal) or other means appreciated by the skilled artisan, as well known in the art.
  • The antibodies that specifically bind CD38 may be administered to a subject by any suitable route, for example parentally by intravenous (i.v.) infusion or bolus injection, intramuscularly or subcutaneously or intraperitoneally. i.v. infusion may be given over for example 15, 30, 60, 90, 120, 180, or 240 minutes, or from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 hours.
  • The dose given to a subject is sufficient to alleviate or at least partially arrest the disease being treated (“therapeutically effective amount”) and may be sometimes 0.005 mg to about 100 mg/kg, e.g. about 0.05 mg to about 30 mg/kg or about 5 mg to about 25 mg/kg, or about 4 mg/kg, about 8 mg/kg, about 16 mg/kg or about 24 mg/kg, or for example about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mg/kg, but may even higher, for example about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 40, 50, 60, 70, 80, 90 or 100 mg/kg.
  • A fixed unit dose may also be given, for example, 50, 100, 200, 500 or 1000 mg, or the dose may be based on the patient's surface area, e.g., 500, 400, 300, 250, 200, or 100 mg/m2. Usually between 1 and 8 doses, (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) may be administered, but 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more doses may be given.
  • The administration of the antibodies that specifically bind CD38 in the methods of the invention may be repeated after one day, two days, three days, four days, five days, six days, one week, two weeks, three weeks, one month, five weeks, six weeks, seven weeks, two months, three months, four months, five months, six months or longer. Repeated courses of treatment are also possible, as is chronic administration. The repeated administration may be at the same dose or at a different dose. For example, the antibodies that specifically bind CD38 may be administered at 8 mg/kg or at 16 mg/kg at weekly interval for 8 weeks, followed by administration at 8 mg/kg or at 16 mg/kg every two weeks for an additional 16 weeks, followed by administration at 8 mg/kg or at 16 mg/kg every four weeks by intravenous infusion.
  • The antibodies that specifically bind CD38 may be administered as maintenance therapy, such as, e.g., once a week for a period of 6 months or more.
  • For example, the antibodies that specifically bind CD38 may be provided as a daily dosage in an amount of about 0.1-100 mg/kg, such as 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 after initiation of treatment, or any combination thereof, using single or divided doses of every 24, 12, 8, 6, 4, or 2 hours, or any combination thereof.
  • The antibodies that specifically bind CD38 may be lyophilized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional protein preparations and well known lyophilization and reconstitution techniques can be employed.
  • The antibodies that specifically bind CD38 may be administered prophylactically to reduce the risk of developing an IgE-mediated disease.
  • The antibodies that specifically bind CD38 may also be administered within seconds, minutes, or hours following a subject's exposure to an allergen, or when at least one symptom of the IgE-mediated disease is present in a subject. Thus, the methods used herein are useful in the treatment of both an acute exposure to an allergen and chronic (e.g., seasonal) exposure to an allergen.
  • The antibodies that specifically bind CD38 may be administered in combination with a second therapeutic agent.
  • The second therapeutic agent may be a standard of care treatment for the IgE-mediated disease, such as allergy, allergic asthma, urticarial, angioedema, autoimmune and inflammatory diseases.
  • Subcutaneous Administration of Pharmaceutical Compositions Comprising an Antibody that Specifically Binds CD38 and a Hyaluronidase
  • The antibody that specifically binds CD38 may be administered as a pharmaceutical composition comprising the antibody that specifically binds CD38 and a hyaluronidase subcutaneously.
  • The concentration of the antibody that specifically binds CD38 in the pharmaceutical composition administered subcutaneously may be about 20 mg/ml.
  • The pharmaceutical composition administered subcutaneously may comprise between about 1,200 mg-1,800 mg of the antibody that specifically binds CD38.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,200 mg of the antibody that specifically binds CD38.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,600 mg of the antibody that specifically binds CD38.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,800 mg of the antibody that specifically binds CD38.
  • The pharmaceutical composition administered subcutaneously may comprise between about 30,000 U-45,000 U of the hyaluronidase.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,200 mg of the antibody that specifically binds CD38and about 30,000 U of the hyaluronidase.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,800 mg of the antibody that specifically binds CD38 and about 45,000 U of the hyaluronidase.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 30,000 U of the hyaluronidase.
  • The pharmaceutical composition administered subcutaneously may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 45,000 U of the hyaluronidase.
  • The pharmaceutical composition administered subcutaneously may comprise the hyaluronidase rHuPH20 having the amino acid sequence of SEQ ID NO: 22.
  • rHuPH20 is a recombinant hyaluronidase (HYLENEX® recombinant) and is described in Int. Pat. Publ. No. WO2004/078140.
  • Hyaluronidase is an enzyme that degrades hyaluronic acid (EC 3.2.1.35) and lowers the viscosity of hyaluronan in the extracellular matrix, thereby increasing tissue permeability.
  • SEQ ID NO: 22
    MGVLKFKHIFFRSFVKSSGVSQIVFTFLLIPCCLTLNFRAPPVIPNV
    PFLWAWNAPSEFCLGKFDEPLDMSLFSFIGSPRINATGQGVTIFYVD
    RLGYYPYIDSITGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLG
    MAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQLSLTEATEKA
    KQEFEKAGKDFLVETIKLGKLLRPNHLWGYYLFPDCYNHHYKKPGYN
    GSCFNVEIKRNDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV
    REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQDELVYTFGETV
    ALGASGIVIWGTLSIMRSMKSCLLLDNYMETILNPYIINVTLAAKMC
    SQVLCQEQGVCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTL
    EDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVCIADGVCIDAFLK
    PPMETEEPQIFYNASPSTLSATMFIVSILFLIISSVASL
  • The administration of the pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be repeated after one day, two days, three days, four days, five days, six days, one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, two months, three months, four months, five months, six months or longer. Repeated courses of treatment are also possible, as is chronic administration. The repeated administration may be at the same dose or at a different dose. For example, the pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be administered once weekly for eight weeks, followed by once in two weeks for 16 weeks, followed by once in four weeks. The pharmaceutical compositions to be administered may comprise about 1,200 mg of the antibody that specifically binds CD38 and about 30,000 U of hyaluronidase, wherein the concentration of the antibody that specifically binds CD38 in the pharmaceutical composition is about 20 mg/ml. The pharmaceutical compositions to be administered may comprise about 1,800 mg of the antibody that specifically binds CD38 and about 45,000 U of hyaluronidase. The pharmaceutical compositions to be administered may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 30,000 U of hyaluronidase. The pharmaceutical compositions to be administered may comprise about 1,600 mg of the antibody that specifically binds CD38 and about 45,000 U of hyaluronidase.
  • The pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be administered subcutaneously to the abdominal region.
  • The pharmaceutical composition comprising the antibody that specifically binds CD38 and the hyaluronidase may be administered in a total volume of about 80 ml, 90 ml, 100 ml, 110 ml or 120 ml.
  • For administration, 20 mg/ml of the antibody that specifically binds CD38 antibody in 25 mM sodium acetate, 60 mM sodium chloride, 140 mM D-mannitol, 0.04% polysorbate 20, pH 5.5 may be mixed with rHuPH20, 1.0 mg/mL (75-150 kU/mL) in 10 mM L-Histidine, 130 mM NaCl, 10 mM L-Methionine, 0.02% Polysorbate 80, pH 6.5 prior to administration of the mixture to a subject.
  • While having described the invention in general terms, the embodiments of the invention will be further disclosed in the following examples that should not be construed as limiting the scope of the claims.
  • Example 1. Daratumumab Reduces IgE Levels in Allergic Patients
  • The effect of DARZALEX™ (dratumumab) treatment on total and specific IgE levels via the depletion of IgE-producing plasma cells was evaluated. This pilot study could demonstrate the potential value of daratumumab in management of patients with severe IgE-mediated disease.
  • Residual blood samples were collected from patients with multiple myeloma (MM) treated with daratumumab monotherapy or daratumumab plus lenalidomide-dexamethsone. Routine blood analysis was performed at baseline and after every four weeks of daratumumab treatment.
  • In the baseline samples, total IgE was determined and an ImmunoCAP Phadiatop (ThermoFisher, Uppsala, Sweden) was performed, according to manufacturer's instructions, to indicate sensitization to common inhalation allergens. If the Phadiatop was positive, specific IgE (sIgE) against birch pollen, timothy grass pollen and house dust mite was determined using ImmunoCAP (ThermoFisher, Uppsala, Sweden). These are among the most frequently recognized inhalant allergens in the Netherlands.
  • Determination of sIgE was performed by the ImmunoCAP technique for both the Phadiatop as the specific/total IgE according to the manufacturer's instructions (Thermo Fisher Scientific, Uppsala Sweden). This technique uses allergens covalently coupled to a solid phase matrix consisting of a cellulose derivative enclosed in a capsule. Analysis of binding of sIgE to the coated allergens is quantified by enzyme-labelled anti-IgE using fluorescence as read-out. sIgE levels ≥0.35 kU/L were defined as positive test results.
  • Catalogue Numbers:
      • Total IgE: 14-4509-01
      • Phadiatop: 14-4405-35
      • Birch sIgE: 14-4102-01 (t3)
      • Timothy grass pollen sIgE: 14-4100-01 (g6)
      • House dust mite sIgE: 14-4107-01 (d1)
  • A total of eight patients were included; five treated with daratumumab monotherapy and three with daratumumab plus lenalidomide-dexamethasone. Four patients had a detectable IgE (≥2 kU/L) at baseline and are listed in Table 1. Only for the first patient (patient 1), specific IgE levels were elevated above reference levels and specific IgE against inhalant allergens were detected. Additional samples from patient 1 at week 4, 8, 12, 16 and 20 were analyzed, demonstrating a decrease of more than 80% in both total IgE and specific IgE levels for timothy grass pollen and house dust mite after 20 weeks (Table 2 and FIG. 1). The other three patients with detectable IgE levels also demonstrated a decrease in total IgE after eight weeks of treatment. For patient 2, total IgE levels decreased with 88% (41 to 5 kU/L). For the other two patients, baseline IgE levels were very low and dropped below detection limit after eight weeks. Percentage of benign and malignant plasma cells were reduced with daratumumab treatment in all patients (Table 3). Patient 1 M-protein and free kappa chain levels were reduced over time (Table 4).
  • In conclusion, this proof of concept demonstrates that levels of total and specific IgE gradually decrease during daratumumab treatment in a single patient sensitized to two common inhalant allergens. This patient was co-treated with lenalidomide and dexamethasone. The effect of lenalidomide on IgE levels is unknown, but glucocorticoids are known to increase IgE synthesis (Hemady Z et al., J Allergy Clin Immunol 1985; 75:304-12. doi:10.1016/0091-6749(85)90062-4; Wu C Y et al., J Clin Invest 1991; 87:870-7. doi:10.1172/JCI115092). IgE depletion by omalizumab treatment resulted in clinical improvement of disease, as well as increased quality of life and reduced socio-economic burden of disease, both in clinical trials as well as daily practice studies (Abraham I et al., Allergy 2015:n/a-n/a. doi:10.1111/a11.12815). The effect of plasma cell depletion by daratumumab on clinical parameters of allergy has yet to be investigated.
  • TABLE 1
    Total Tim. grass HDM
    IgE Birch sIgE sIgE sIgE
    Patient Study (kU/L) Phadiatop (kU/L) (kU/L) (kU/L)
    1 D + LD* 356 Positive Negative 9.83 3.23
    2 D + LD* 41 Negative
    3 D** 3 Negative
    4 D + LD* 7 Negative
    Abbreviations:
    D: daratumumab monotherapy, D + L: daratumumab plus lenalidomide-dexamethasone,
    Birch: birch pollen, Tim. grass: timothy grass pollen,
    HDM: house dust mite
    Reference values:
    Total IgE: <100 kU/L; ImmunoCAP sIgE birch/tim. grass/hdm: <0.35 kU/L
    *Patient was enrolled in the GEN503 study and received daratumumab 16 mg/kg and
    lenalidomide 25 mg.
    **Patient was in the GEN501 phase II study and received daratumumab 16 mg/kg.
  • TABLE 2
    Week
    W0 W4 W8 W12 W16 W20
    Total IgE 356 197 185 145 95 44
    Tim. grass 9.83 6.07 6.88 6.63 4.1 1.67
    HDM 3.23 1.93 1.54 1.06 0.73 0.4
    Abbreviations:
    Tim. grass: timothy grass pollen,
    HDM: house dust mite
    Reference values:
    Total IgE: <100 kU/L; ImmunoCAP sIgE tim. grass/hdm: <0.35 kU/L
  • TABLE 3
    % malignant plasma
    Response % benign plasma cells cells
    Start to Pre- After Pre- After
    Patient treatment treatment treatment W8/12 treatment W8/12
    1 April 2014 CR 0.420 0.240 0.226 0.015
    2 April 2014 CR 0.168 0.033 2.485 0.539
    3 April 2014 PD 0.032 0 2.543 0.283
    4 June 2014 CR 0.055 0 6.214 0.025
    CR: complete response,
    PD: progressive disease.
  • TABLE 4
    Time M-protein levels Free kappa
    point quantitative (g/l) chains (mg/L)
    W0 16  52.96
    W4 5 14.52
    W8 3 16.40
    W12 Not quantifiable 14.28
    W16 Not quantifiable 13.58
    W20 Not quantifiable 12.69

Claims (36)

1. A method of treating an IgE-mediated disease, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody that specifically binds CD38 for a time sufficient to treat the IgE-mediated disease.
2. The method of claim 1, wherein the IgE-mediated disease is an allergic response to an allergen.
3. The method of claim 2, wherein the allergen is a pollen, a dust mite, a food allergen, a plant allergen, animal dander, insect stings, a fungus, a spore, a mold, latex, or a drug.
4. The method of claim 1, wherein the IgE-mediated disease is allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis, bullous pemphigoid, systemic lupus erythematosus, lupus, rheumatoid arthritis or an autoimmune disease.
5. The method of claim 1, wherein the IgE-mediated disease is autoimmune disease.
6. The method of claim 1, wherein the IgE-mediated disease is lupus.
7. The method of claim 6, wherein lupus is systemic lupus erythematosus (SLE), discoid lupus erythematosus, subacute cutaneous lupus erythematosus, neonatal lupus or drug-induced lupus.
8. The method of claim 1, wherein the IgE-mediated disease is rheumatoid arthritis.
9. The method of claim 1, wherein the IgE-mediated disease is acute or chronic.
10. The method of claim 1, wherein the antibody that specifically binds CD38 is administered to the subject following exposure to an allergen.
11. The method of claim 1 wherein the antibody that specifically binds CD38 competes for binding to CD38 with an antibody comprising a heavy chain variable region (VH) of SEQ ID NO: 4 and a light chain variable region (VL) of SEQ ID NO: 5.
12. The method of claim 1, wherein the antibody that specifically binds CD38 comprises a heavy chain complementarity determining region 1 (HCDR1), a HCDR2 and a HCDR3 amino acid sequences of SEQ ID NOs: 6, 7 and 8, respectively, and a light chain complementarity determining region 1 (LCDR1), a LCDR2 and a LCDR3 amino acid sequences of SEQ ID NOs: 9, 10 and 11, respectively.
13. The method claim 1, wherein the antibody that specifically binds CD38 comprises the VH having an amino acid sequence that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 4 and the VL having an amino acid sequence that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 5.
14. The method of claim 1, wherein the antibody that specifically binds CD38 is daratumumab or biosimilar thereof.
15. The method of claim 1, wherein the antibody that specifically binds CD38 is a non-agonistic antibody.
16. The method of claim 1, wherein the antibody that specifically binds CD38 comprises the VH of SEQ ID NO: 4 and the VL of SEQ ID NO: 5.
17. The method of claim 1, wherein the antibody that specifically binds CD38 comprises a heavy chain having an amino acid sequence that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 12 and a light chain having an amino acid sequence that is 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 13.
18. The method of claim 1, wherein the antibody that specifically binds CD38 comprises the heavy chain of SEQ ID NO: 12 and the light chain of SEQ ID NO: 13.
19. The method of claim 1, wherein the antibody that specifically binds CD38 comprises:
a. the VH of SEQ ID NO: 14 and the VL of SEQ ID NO: 15;
b. the VH of SEQ ID NO: 16 and the VL of SEQ ID NO: 17;
c. the VH of SEQ ID NO: 18 and the VL of SEQ ID NO: 19; or
d. the VH of SEQ ID NO: 20 and the VL of SEQ ID NO: 21.
20. The method of claim 1, wherein the antibody that specifically binds CD38 is an IgG1, IgG2, IgG3 or IgG4 isotype.
21. The method of claim 1, wherein the antibody that specifically binds CD38 induces killing of IgE producing CD38+ cells.
22. The method of claim 21, wherein the antibody that specifically binds CD38 induces killing of IgE producing CD38+ cells by antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement dependent cytotoxicity (CDC), apoptosis, or modulation of CD38 enzymatic activity.
23. The method of claim 22, wherein the antibody that specifically binds CD38 induces killing of IgE producing CD38+ cells by ADCC.
24. The method of claim 1, wherein the antibody that specifically binds CD38 is administered to the subject prophylactically.
25. The method of claim 1, wherein the antibody that specifically binds CD38 is administered intravenously.
26. The method of claim 1, wherein the antibody that specifically binds CD38 is administered subcutaneously in a pharmaceutical composition comprising the antibody that specifically binds CD38 and a hyaluronidase.
27. The method of claim 26, wherein the hyaluronidase is rHuPH20 of SEQ ID NO: 22.
28. The method of claim 1, wherein the antibody that specifically binds CD38 is administered with a second therapeutic agent.
29. The method of claim 28, wherein the second therapeutic agent is a standard of care treatment for the IgE-mediated disease.
30. The method of claim 28, wherein the antibody that specifically binds CD38 and the second therapeutic agent are administered simultaneously, sequentially or separately.
31. An antibody that specifically binds CD38 for use in the treatment of an IgE-mediated disease.
32. The antibody of claim 31 for use in the treatment of an IgE-mediated disease, wherein the antibody comprises
a. the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2 and the LCDR3 of SEQ ID NOs: 6, 7, 8, 9, 10 and 11;
b. the VH and the VL of SEQ ID NOs: 4 and 5, respectively;
c. the HC and the LC of SEQ ID NOs: 12 and 13, respectively; and/or
d. daratumumab or a biosimilar thereof.
33. The antibody of claim 31 for use in the treatment of an IgE-mediated disease, wherein the IgE-mediated disease is an allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis, bullous pemphigoid, systemic lupus erythematosus, lupus, rheumatoid arthritis or an autoimmune disease.
34. A pharmaceutical composition for the treatment of an IgE-mediated disease comprising an antibody that specifically binds CD38.
35. The pharmaceutical composition of claim 34, wherein the antibody comprises
a. the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2 and the LCDR3 of SEQ ID NOs: 6, 7, 8, 9, 10 and 11;
b. the VH and the VL of SEQ ID NOs: 4 and 5, respectively;
c. the HC and the LC of SEQ ID NOs: 12 and 13, respectively; and/or
d. daratumumab or biosimilar thereof.
36. The pharmaceutical composition of claim 34, wherein the IgE-mediated disease is an allergic asthma, urticaria, angioedema, food allergy, an allergic response, atopic dermatitis, anaphylaxis, cutaneous mastocystosis, allergic rhinitis, nasal polyposis, Kimura's disease, eosinophilic otitis media, eosinophilic gastroenteritis, latex allergy, bronchopulmonary allergic aspergillosis, bullous pemphigoid, systemic lupus erythematosus, lupus, rheumatoid arthritis or an autoimmune disease.
US16/312,133 2016-06-28 2017-06-28 Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38 Abandoned US20190233533A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/312,133 US20190233533A1 (en) 2016-06-28 2017-06-28 Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662355526P 2016-06-28 2016-06-28
PCT/EP2017/066063 WO2018002181A1 (en) 2016-06-28 2017-06-28 TREATMENT OF IgE-MEDIATED DISEASES WITH ANTIBODIES THAT SPECIFICALLY BIND CD38
US16/312,133 US20190233533A1 (en) 2016-06-28 2017-06-28 Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38

Publications (1)

Publication Number Publication Date
US20190233533A1 true US20190233533A1 (en) 2019-08-01

Family

ID=59298446

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/312,133 Abandoned US20190233533A1 (en) 2016-06-28 2017-06-28 Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38

Country Status (4)

Country Link
US (1) US20190233533A1 (en)
EP (1) EP3474895A1 (en)
JP (1) JP2019527678A (en)
WO (1) WO2018002181A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10556961B2 (en) 2014-02-28 2020-02-11 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
US10604580B2 (en) 2014-09-09 2020-03-31 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
US10668149B2 (en) 2015-06-22 2020-06-02 Janssen Biotech, Inc. Combination therapies for heme malignancies with anti-CD38 antibodies and survivin inhibitors
US10766965B2 (en) 2015-05-20 2020-09-08 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of light chain amyloidosis and other CD38-positive hematological malignancies
US10781261B2 (en) 2015-11-03 2020-09-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US10793630B2 (en) 2014-12-04 2020-10-06 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute myeloid leukemia
US10800851B2 (en) 2014-02-28 2020-10-13 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
US20210040224A1 (en) * 2018-03-13 2021-02-11 Trillium Therapeutics Inc. Cd47 blockade therapy with cd38 antibody
US11021543B2 (en) 2015-06-24 2021-06-01 Janssen Biotech, Inc. Immune modulation and treatment of solid tumors with antibodies that specifically bind CD38
US11566079B2 (en) 2015-11-03 2023-01-31 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US20240000934A1 (en) * 2020-12-28 2024-01-04 Bristol-Myers Squibb Company Antibody Compositions and Methods of Use Thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020194241A1 (en) * 2019-03-28 2020-10-01 Janssen Biotech, Inc. Clinically proven subcutaneous pharmaceutical compositions comprising anti-cd38 antibodies and their uses
KR20230142834A (en) 2021-01-14 2023-10-11 모르포시스 아게 Anti-CD38 antibodies and uses thereof
TW202302642A (en) 2021-03-01 2023-01-16 德商莫菲西斯公司 Anti-cd38 antibodies for use in the treatment of antibody-mediated transplant rejection

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011034604A2 (en) * 2009-09-17 2011-03-24 Baxter Healthcare, S.A. Stable co-formulation of hyaluronidase and immunoglobulin, and methods of use thereof

Family Cites Families (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
WO1988001649A1 (en) 1986-09-02 1988-03-10 Genex Corporation Single polypeptide chain binding molecules
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
ATE463573T1 (en) 1991-12-02 2010-04-15 Medimmune Ltd PRODUCTION OF AUTOANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES
DK0672142T3 (en) 1992-12-04 2001-06-18 Medical Res Council Multivalent and multi-specific binding proteins as well as their preparation and use
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
AUPO591797A0 (en) 1997-03-27 1997-04-24 Commonwealth Scientific And Industrial Research Organisation High avidity polyvalent and polyspecific reagents
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6833441B2 (en) 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
EP2330213A1 (en) 2003-03-05 2011-06-08 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US8597911B2 (en) 2003-06-11 2013-12-03 Chugai Seiyaku Kabushiki Kaisha Process for producing antibodies
KR20140066259A (en) 2004-02-06 2014-05-30 모르포시스 아게 Anti-cd38 human antibodies and uses therefor
CA2602375C (en) * 2005-03-23 2018-07-24 Genmab A/S Antibodies against cd38 for treatment of multiple myeloma
EP3050963B1 (en) 2005-03-31 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
US20090123950A1 (en) 2005-05-24 2009-05-14 Morphosys Ag Generation And Profiling Of Fully Human Hucal Gold®-Derived Therapeutic Antibodies Specific For Human CD38
LT2860192T (en) 2005-10-12 2017-12-11 Morphosys Ag Generation and profiling of fully human HuCAL GOLD-derived therapeutic antibodies specific for human CD38
ES2395969T3 (en) 2006-03-24 2013-02-18 Merck Patent Gmbh Genetically modified heterodimeric protein domains
JP2009541275A (en) 2006-06-22 2009-11-26 ノボ・ノルデイスク・エー/エス Production of bispecific antibodies
EP1914242A1 (en) * 2006-10-19 2008-04-23 Sanofi-Aventis Novel anti-CD38 antibodies for the treatment of cancer
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
CA2681974C (en) 2007-03-29 2019-12-31 Genmab A/S Bispecific antibodies and methods for production thereof
AU2008343589A1 (en) 2007-12-19 2009-07-09 Centocor Ortho Biotech Inc. Design and generation of human de novo pIX phage display libraries via fusion to pIX or pVII, vectors, antibodies and methods
PT2235064E (en) 2008-01-07 2016-03-01 Amgen Inc Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
BRPI0910482A2 (en) 2008-04-29 2019-09-24 Abbott Lab double variable domain immunoglobins and their uses
ES2708124T3 (en) 2009-04-27 2019-04-08 Oncomed Pharm Inc Procedure for preparing heteromultimeric molecules
CA2796181C (en) 2010-04-20 2023-01-03 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
US9527926B2 (en) 2010-05-14 2016-12-27 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
SI2580243T1 (en) * 2010-06-09 2020-02-28 Genmab A/S Antibodies against human cd38
EP2420253A1 (en) 2010-08-20 2012-02-22 Leadartis, S.L. Engineering multifunctional and multivalent molecules with collagen XV trimerization domain
EP2635607B1 (en) 2010-11-05 2019-09-04 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
JOP20210044A1 (en) * 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
MX370377B (en) 2013-04-29 2019-12-11 Teva Pharmaceuticals Australia Pty Ltd Anti-cd38 antibodies and fusions to attenuated interferon alpha-2b.
WO2015195555A1 (en) * 2014-06-16 2015-12-23 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Blocking cd38 using anti-cd38 antibody conjugated to protein g to protect nk cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011034604A2 (en) * 2009-09-17 2011-03-24 Baxter Healthcare, S.A. Stable co-formulation of hyaluronidase and immunoglobulin, and methods of use thereof

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10556961B2 (en) 2014-02-28 2020-02-11 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
US10800851B2 (en) 2014-02-28 2020-10-13 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
US11713355B2 (en) 2014-02-28 2023-08-01 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
US10604580B2 (en) 2014-09-09 2020-03-31 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
US10793630B2 (en) 2014-12-04 2020-10-06 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute myeloid leukemia
US10766965B2 (en) 2015-05-20 2020-09-08 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of light chain amyloidosis and other CD38-positive hematological malignancies
US10668149B2 (en) 2015-06-22 2020-06-02 Janssen Biotech, Inc. Combination therapies for heme malignancies with anti-CD38 antibodies and survivin inhibitors
US11021543B2 (en) 2015-06-24 2021-06-01 Janssen Biotech, Inc. Immune modulation and treatment of solid tumors with antibodies that specifically bind CD38
US10781261B2 (en) 2015-11-03 2020-09-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11566079B2 (en) 2015-11-03 2023-01-31 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11708420B2 (en) 2015-11-03 2023-07-25 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11708419B2 (en) 2015-11-03 2023-07-25 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11732051B2 (en) 2015-11-03 2023-08-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US20210040224A1 (en) * 2018-03-13 2021-02-11 Trillium Therapeutics Inc. Cd47 blockade therapy with cd38 antibody
US20240000934A1 (en) * 2020-12-28 2024-01-04 Bristol-Myers Squibb Company Antibody Compositions and Methods of Use Thereof

Also Published As

Publication number Publication date
JP2019527678A (en) 2019-10-03
WO2018002181A1 (en) 2018-01-04
EP3474895A1 (en) 2019-05-01

Similar Documents

Publication Publication Date Title
US20190233533A1 (en) Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38
US10604580B2 (en) Combination therapies with anti-CD38 antibodies
CN108136218B (en) anti-CD 38 antibodies for the treatment of light chain amyloidosis and other CD 38-positive hematologic malignancies
JP6963577B2 (en) Anti-Siglec-8 antibody and how to use it
CA3044851A1 (en) Methods of treating inflammatory conditions
EP3954710A2 (en) Antibodies to canine interleukin-4 receptor alpha
JP2021525082A (en) Anti-SIRPA antibody and its usage
JP2017501744A5 (en)
RU2737378C2 (en) Hybrid proteins of human protein fragments to create ordered multimerized compositions of immunoglobulin fc regions with enhanced binding with a complement system
TR201808486T4 (en) Antibodies against bradykinin B1 receptor ligands.
Zhang et al. A recombinant human IgG1 Fc multimer designed to mimic the active fraction of IVIG in autoimmunity
D Brightbill et al. Quilizumab is an afucosylated humanized anti-M1 prime therapeutic antibody
AU2010279892A1 (en) Humanized anti-amyloid-b oligomer antibody
KR20200016232A (en) Methods and compositions for treating allergic eye diseases
US20230391884A1 (en) Treatment Of IgE-Mediated Diseases With Antibodies That Specifically Bind CD38
US20220127365A1 (en) Anti-cd94 antibodies and methods of use thereof
US20180051069A1 (en) Compositions and methods for treating allergic inflammation
WO2021062268A1 (en) Therapeutic neutralization antibodies for the treatment of peanut allergy
CA3109513A1 (en) Anti-fc epsilon-r1 alpha (fcer1a) antibodies, bispecific antigen-binding molecules that bind fcer1a and cd3, and uses thereof
EA045206B1 (en) METHODS AND COMPOSITIONS FOR TREATING ALLERGIC EYE DISEASES
NZ794842A (en) Methods of treating inflammatory conditions
EA044647B1 (en) ANTIBODY THERAPY FOR TRANSTHYRETIN (TTR) AMYLOIDOSIS, AS WELL AS ANTIBODIES OBTAINED FROM HUMAN FOR THE SPECIFIED THERAPY

Legal Events

Date Code Title Description
AS Assignment

Owner name: UMC UTRECHT HOLDING B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:OTTEN, HENNY G.;REEL/FRAME:049484/0770

Effective date: 20190614

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION