EP3661551A1 - Méthodes de traitement de l'oesophagite à éosinophiles active - Google Patents

Méthodes de traitement de l'oesophagite à éosinophiles active

Info

Publication number
EP3661551A1
EP3661551A1 EP18750578.9A EP18750578A EP3661551A1 EP 3661551 A1 EP3661551 A1 EP 3661551A1 EP 18750578 A EP18750578 A EP 18750578A EP 3661551 A1 EP3661551 A1 EP 3661551A1
Authority
EP
European Patent Office
Prior art keywords
patient
eoe
pathway inhibitor
inhibitor
antibody
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.)
Pending
Application number
EP18750578.9A
Other languages
German (de)
English (en)
Inventor
Allen Radin
Jennifer D. Hamilton
Leda MANNENT
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.)
Sanofi Biotechnology SAS
Regeneron Pharmaceuticals Inc
Original Assignee
Sanofi Biotechnology SAS
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanofi Biotechnology SAS, Regeneron Pharmaceuticals Inc filed Critical Sanofi Biotechnology SAS
Priority claimed from PCT/US2018/045195 external-priority patent/WO2019028367A1/fr
Publication of EP3661551A1 publication Critical patent/EP3661551A1/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • 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
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/577Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 tolerising response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins 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
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to the use of interleukin-4/ interleukin-13 pathway inhibitors to treat or prevent active eosinophilic esophagitis in a subject in need thereof.
  • Esophageal stricture (narrowing of the esophagus) results from injury to the esophageal lining and leads to, inter alia, difficulty in swallowing (dysphagia), regurgitation of food or liquid, heartburn and unintended weight loss.
  • Treatment of esophageal stricture is very important as it reduces quality of life due to dysphagia, weight loss, and nutritional imbalance.
  • Esophageal stricture may be caused due to chronic ulceration or chronic inflammation, as a complication due to chemotherapy, radiotherapy, esophageal cancer or endoscopic surgery, peptic ulcers or gastroesophageal reflux.
  • Esophageal stricture is also caused by eosinophilic esophagitis.
  • Eosinophilic esophagitis is an emerging, chronic, immune-/antigen- mediated disease characterized by esophageal dysfunction and eosinophil inflammation in the esophagus (Liacouras et al 2011, The Journal of Allergy and Clinical Immunology. 128: 3-20 e6; quiz 1-2; Weinbrand-Goichberg et al 2013, Immunologic Research. 56: 249- 60; Zhang et al 2013, Digestive Diseases and Sciences 58: 1497-506).
  • EoE Eosinophilic esophagitis
  • Some patients may also have concomitant asthma or an atopic disease such as atopic dermatitis, or allergic rhinitis.
  • the symptomatic burden of EoE including food avoidance, eating modification behaviors, and social, emotional, financial, work and school, and sleep impacts, is also important and relevant to the EoE population and, if improved, may reflect a treatment benefit for EoE patients.
  • Esophageal dilation by endoscopy is currently used to treat esophageal stricture and increase esophageal distensibility.
  • it is a surgical procedure that is invasive and may lead to complications such as perforation and bleeding. Accordingly, there is an unmet need for safe and effective therapies that increase esophageal distensibility and treat esophageal stricture (e.g. , in eosinophilic esophagitis).
  • methods for increasing the distensibility of the esophagus.
  • the methods comprise: (a) selecting a patient with moderate-to-severe eosinophilic esophagitis (EoE), wherein the patient in need thereof has an attribute or is selected on the basis of a criterion selected from the group consisting of: (i) the patient has > 15 eosinophils per high powered field (hpf) in the esophagus prior to or at the time of the treatment ("baseline”); (ii) the patient exhibits at least one episode of dysphagia per week; and (iii) the patient has a Straumann Dysphagia Instrument (SDI) score >2; and (b) administering a therapeutically effective amount of a pharmaceutical composition comprising an interleukin-4/ interleukin-13 (IL-4/IL-13) pathway inhibitor to the patient in need thereof, thereby increasing esophage
  • EoE moderate-to-severe eosinophil
  • the patient has active EoE. In one embodiment, the patient is >18 years of age. In one embodiment, the patient has been treated previously with proton pump inhibitors (PPIs). In one embodiment, the patient has had at least one prior esophageal dilation.
  • PPIs proton pump inhibitors
  • the patient has a characteristic selected from the group consisting of: (1) prior treatment with at least one of PPIs, esophageal dilation, corticosteroids, allergen withdrawal and/or diet modification; (2) the patient is unresponsive or resistant to prior treatment with PPIs or esophageal dilation; (3) the patient has an Eosinophilic Esophagitis Severity and Activity Index (EEsAI) score >30, >40, or >50; (4) the patient suffers from EoE for at least 3 years; (5) the patient, prior to or at the time of administration of the IL-4/IL-13 pathway inhibitor, has or is diagnosed with a disease or disorder selected from the group consisting of food allergy, atopic dermatitis, asthma, allergic rhinitis and allergic conjunctivitis; and (6) the patient has an elevated level of a biomarker selected from the group consisting of eotaxin- 3, periostin, serum IgE (total and allergen-specific), IL-13, IL-5
  • Eosinophilic Esophagitis Severity and Activity Index (EEsAI) score >30, >40, or >50;
  • the patient prior to or at the time of administration of the IL-4/IL-13 pathway inhibitor, has or is diagnosed with a disease or disorder selected from the group consisting of food allergy, atopic dermatitis, asthma, allergic rhinitis, and/or allergic conjunctivitis; and/or (6) the patient has an elevated level of at least one biomarker selected from the group consisting of eotaxin-3, periostin, serum IgE (total and allergen-specific), IL-13, IL-5, serum thymus and activation regulated chemokine (TARC), thymic stromal lymphopoietin (TSLP), serum eosinophilic cationic protein (ECP), and/or eosinophil-derived neurotoxin (EDN),
  • a disease or disorder selected from the group consisting of food allergy, atopic dermatitis, asthma, allergic rhinitis, and/or allergic conjunctivitis
  • the patient has an elevated level of at least
  • the patient has at least characteristic (1) or characteristic (2) or characteristic (3) or characteristic (4) or characteristic (5) or characteristic (6).
  • the patient can also, based on such language, have more than one of the six characteristics (for example, (1) and (2), or (4) and (5), or (1), (2), and (6), and so on). It is not, however, meant that the patient must have at least characteristic (1) and characteristic (2) and characteristic (3) and characteristic (4) and characteristic (5) and characteristic (6).
  • methods are provided for treating, preventing or ameliorating at least one symptom or indication of active eosinophilic esophagitis (EoE) in a subject.
  • the methods comprise selecting a patient with moderate-to-severe EoE, and administering a therapeutically effective amount of a pharmaceutical composition comprising an interleukin-4/ interleukin-13 (IL-4/IL-13) pathway inhibitor to the patient in need thereof.
  • IL-4/IL-13 interleukin-13
  • the patient in need thereof is selected on the basis of an attribute or criterion selected from the group consisting of: (1) the patient has > 15 eosinophils per high powered field (hpf) in the esophagus prior to or at the time of the treatment
  • baseline (“baseline”); (2) prior treatment with at least one of high dose proton pump inhibitors (PPIs), esophageal dilation, corticosteroids, allergen withdrawal and/or diet modification; (3) the patient exhibits at least one episode of dysphagia per week; (4) the patient is unresponsive or resistant to prior treatment with high dose PPIs or esophageal dilation; (5) the patient has a Straumann Dysphagia Instrument (SDI) score >5; (6) the patient has an Eosinophilic Esophagitis Severity and Activity Index (EEsAI) score >30, >40, or >50; (7) the patient suffers from EoE for at least 3 years; (8) the patient, prior to or at the time of administration of the IL-4/IL-13 pathway inhibitor, has or is diagnosed with a disease or disorder selected from the group consisting of food allergy, atopic dermatitis, asthma, allergic rhinitis and allergic conjunctivitis; and (9) the patient has an elevated level of a
  • methods for reducing dysphagia, the methods comprising selecting a patient with moderate-to-severe EoE wherein the patient (i) exhibits >1 episodes of dysphagia per week; (ii) has been treated previously with high-dose proton pump inhibitors (PPIs); and/or (iii) has had at least one prior esophageal dilation; and (b) administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to the patient in need thereof.
  • PPIs proton pump inhibitors
  • methods for improving a parameter, the methods comprising selecting a patient with moderate-to- severe EoE; and administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor, wherein the administration leads to an improvement in a parameter selected from the group consisting of: (a) reduction of at least 40% from baseline in dysphagia frequency and severity, as measured by Straumann Dysphagia Instrument (SDI) score; (b) reduction of 3 points from baseline in the SDI score; (c) reduction of more than 85% from baseline in peak intraepithelial eosinophil count in proximal, mid and/or distal regions of the esophagus; (d) increase of at least 10% from baseline in esophageal distensibility, as measured by impedance planimetry; (e) decrease of more than 50% from baseline in severity and extent of disease, as measured by EoE Histology Scoring System (HSS) score; and (f
  • methods are provided for reducing the eosinophilic infiltration of esophagus in a patient in need thereof.
  • methods are provided for reducing inflammation in the esophagus.
  • the methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor.
  • the eosinophilic infiltration of the esophagus is represented by greater than or equal to about 15 eosinophils per high powered field in the esophagus of the subject in need thereof.
  • the number of eosinophils is reduced >85% following administration of the IL-4/IL-13 pathway inhibitor.
  • the inflammation e.g. , mucosal inflammation
  • the inflammation is identified by endoscopy and features such as esophageal edema, rings, exudates, furrows and strictures (EREFS).
  • EREFS esophageal edema, rings, exudates, furrows and strictures
  • administration of the IL-4/IL-13 pathway inhibitor leads to reduction in the EREFS score to less than 8, less than 7, less than 6, less than 5, less than 4, less than 3, or less than 2 (disclosed elsewhere herein).
  • the EoE-associated biomarker is selected from the group consisting of, e.g. , circulating or esophagus eosinophils, eotaxin-3, periostin, serum IgE (total and allergen-specific), IL-13, IL-5, serum thymus and activation regulated chemokine (TARC; CCL17), thymic stromal lymphopoietin (TSLP), serum eosinophilic cationic protein (ECP), and eosinophil-derived neurotoxin (EDN).
  • the methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor.
  • the IL-4/IL-13 pathway inhibitor is administered in combination with a second therapeutic agent or therapy.
  • the subject in need thereof has a concurrent disease or disorder selected from the group consisting of food allergy, atopic dermatitis, asthma, allergic rhinitis, allergic conjunctivitis and inherited connective tissue disorders.
  • Exemplary IL-4/IL-13 pathway inhibitors that can be used in the context of the methods of the present invention include, but are not limited to, an anti-IL-4 antibody, an anti-IL-13 antibody, a bispecific anti-IL-4/IL-13 antibody and an IL-4 receptor (IL-4R) inhibitor.
  • the IL-4/IL-13 pathway inhibitor is an IL-4R inhibitor (such as an anti-IL-4R antibody).
  • Exemplary IL-4R inhibitors that can be used in the context of the methods of the present invention include, e.g., small molecule chemical inhibitors of IL-4R or its ligands (IL-4 and/or IL-13), or biological agents that target IL-4R or its ligands.
  • the IL-4R inhibitor is an antibody or antigen-binding protein that binds the IL-4Ra chain and blocks signaling by IL-4, IL-13, or both IL-4 and IL-13.
  • the anti-IL-4R antibody or antigen-binding protein comprises the heavy chain complementarity determining regions (HCDRs) of a heavy chain variable region (HCVR) comprising the amino acid sequence of SEQ ID NO: 1 and the light chain CDRs of a light chain variable region (LCVR) comprising the amino acid sequence of SEQ ID NO: 2.
  • HCDRs heavy chain complementarity determining regions
  • LCVR light chain variable region
  • antigen-binding protein that can be used in the context of the methods of the present invention is an anti-IL-4Ra antibody such as dupilumab.
  • the present invention provides use of an IL-4/IL-13 pathway inhibitor in the manufacture of a medicament to treat or inhibit or prevent active eosinophilic esophagitis in a subject, including humans.
  • the present invention provides use of an antibody or antigen-binding fragment thereof that binds to IL-4R in the manufacture of a medicament to treat or inhibit or prevent active eosinophilic esophagitis in a subject, including humans.
  • the present invention provides use of an IL-4/IL-13 pathway inhibitor in the manufacture of a medicament to increase esophageal distensibility in a subject, including humans.
  • the subject has active EoE.
  • the subject has moderate-to-severe EoE.
  • the present invention provides use of an antibody or antigen-binding fragment thereof that binds to IL-4R in the manufacture of a medicament to increase esophageal distensibility in a subject, including humans.
  • the subject has active EoE.
  • the subject has moderate-to-severe EoE
  • Figure 1 lists the components comprising the weekly Eosinophilic Esophagitis Severity and Activity Index (EEsAI) score.
  • EEsAI Activity Index
  • Figure 2 shows mean change from baseline in Straumann Dysphagia Instrument (SDI) scores during the 12-week treatment period in patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • SDI Straumann Dysphagia Instrument
  • Figure 3 shows mean percent change from baseline in dysphagia frequency and severity components of the Straumann Dysphagia Instrument (SDI) score at week 10 and week 12 upon administration of once-a-week (qw) 300 mg dupilumab vs placebo.
  • SDI Straumann Dysphagia Instrument
  • Figure 4 shows the mean percent change from baseline in EEsAI score during the 12-week treatment period in patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 5 shows mean percent change from baseline in total EoE Edema Rings Exudates Furrows and Strictures (EREFS) score as well as in the subcomponents of the EREFS score at week 12 in patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • ERP EoE Edema Rings Exudates Furrows and Strictures
  • Figure 6 shows mean change from baseline in total EoE Histology Scoring System (HSS) score for the grade (severity) subcomponent at week 12 in patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 7 shows mean change from baseline in total EoE HSS score for the stage (extent) subcomponent at week 12 in patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 8 is made up of Figures 8A, 8B, 8C, and 8D.
  • Figure 8A shows mean change from baseline in EoE HSS grade scores for basal zone hyperplasia
  • Figure 8B shows mean change from baseline in EoE HSS grade scores for eosinophil surface layering
  • Figure 8C shows mean change from baseline in EoE HSS grade scores for eosinophil inflammation
  • Figure 8D shows mean change from baseline in EoE HSS grade scores for eosinophil abscess in proximal, mid and distal regions of the esophagus sampled at week 12 from patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 9 is made up of Figures 9A, 9B, 9C, and 9D.
  • Figure 9A shows mean change from baseline in EoE HSS stage scores for basal zone hyperplasia
  • Figure 9B shows mean change from baseline in EoE HSS stage scores for eosinophil abscess
  • Figure 9C shows mean change from baseline in EoE HSS stage scores for eosinophil inflammation
  • Figure 9D shows mean change from baseline in EoE HSS stage scores for eosinophil surface layering in proximal, mid and distal regions of the esophagus sampled at week 12 from patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 10 is made up of Figures 10A, 10B, and IOC.
  • Figure 10A shows mean change from baseline in EoE HSS grade scores for dilated intercellular spaces;
  • Figure 10B shows mean change from baseline in EoE HSS grade scores for surface alteration and
  • Figure IOC shows mean change from baseline in EoE HSS grade scores for apoptotic epithelial cells in proximal, mid and distal regions of the esophagus sampled at week 12 from patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 11 is made up of Figures 11 A, 11B, and 11C.
  • Figure 11A shows mean change from baseline in EoE HSS stage scores for dilated intercellular spaces;
  • Figure 11B shows mean change from baseline in EoE HSS stage scores for surface alteration and
  • Figure 11C shows mean change from baseline in EoE HSS stage scores for apoptotic epithelial cells in proximal, mid and distal regions of the esophagus sampled at week 12 from patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • Figure 12 shows percent change from baseline in distensibility plateau at week 12 in patients administered once-a-week (qw) 300 mg dupilumab vs placebo.
  • the present invention includes methods for treating, preventing, or ameliorating at least one symptom or indication of active eosinophilic esophagitis (EoE) in a subject.
  • the methods according to this aspect of the invention comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL- 13 pathway inhibitor to the subject in need thereof.
  • the terms "treat”, “treating”, or the like mean to alleviate symptoms, eliminate the causation of symptoms either on a temporary or permanent basis, or to prevent or slow the appearance of symptoms of eosinophilic inflammation in the esophagus.
  • the present methods are useful for treating or ameliorating at least one symptom or indication of EoE including, but not limited to, eosinophilic infiltration of the esophagus, thickening of the esophageal wall, inflammation in the esophagus, appearance of trachea-like rings or ridges in the esophagus, chest and abdominal pain, food refusal, vomiting, dysphagia and food impaction.
  • EoE including, but not limited to, eosinophilic infiltration of the esophagus, thickening of the esophageal wall, inflammation in the esophagus, appearance of trachea-like rings or ridges in the esophagus, chest and abdominal pain, food refusal, vomiting, dysphagia and food impaction.
  • Eosinophilic Esophagitis means an inflammatory disease characterized by abnormal eosinophilic inflammation within the esophagus and esophageal dysfunction.
  • the primary symptoms of EoE include, but are not limited to, chest and abdominal pain, dysphagia, heartburn, food refusal, vomiting and food impaction.
  • the clinicopathology of EoE is characterized by presence of ridges or trachealike rings in the esophageal wall and eosinophilic infiltration in the esophageal mucosa.
  • EoE is diagnosed by endoscopy of the esophagus followed by microscopic and biochemical analysis of the esophageal mucosal lining. EoE may be classified as allergic or non-allergic depending upon the status of the subject.
  • the present invention includes methods to treat both allergic and non-allergic forms of EoE.
  • active EoE refers to the EoE disease in a patient wherein the patient has >15 eosinophils/high powered field (hpf) in an esophageal biopsy even after 8 weeks of treatment with proton pump inhibitors (PPIs).
  • PPIs proton pump inhibitors
  • the term also refers to the EoE disease in patients that exhibit frequent dysphagia, e.g. , the patient has 2, 3, 4, 5, or more episodes of dysphagia per week.
  • active EoE includes mild EoE as well as moderate-to-severe EoE.
  • Moderate-to-severe refers to EoE disease in patients that in addition to eosinophilia (e.g.., >15 eosinophils/hpf in the esophageal mucosa) and frequent episodes of dysphagia, have SDI score >2 and/or EEsAI score >30, have duration of EoE for at least 2 years, and/or are non-responsive or resistant to prior therapy (including PPIs or esophageal dilation).
  • eosinophilia e.g.., >15 eosinophils/hpf in the esophageal mucosa
  • EEsAI score >30
  • duration of EoE for at least 2 years and/or are non-responsive or resistant to prior therapy (including PPIs or esophageal dilation).
  • a subject in need thereof means a human or non- human mammal that exhibits one or more symptoms or indications of eosinophilic esophagitis, and/or who has been diagnosed with eosinophilic esophagitis (EoE).
  • the term "subject” is used interchangeably with the term "patient”.
  • the term "a subject in need thereof may also include, e.g. , patients who, prior to treatment, exhibit (or have exhibited) one or more indications of EoE such as, e.g. , esophageal overexpression of pro-inflammatory mediators such as mast cells, eosinophilic infiltration of the esophagus, thickening of the esophageal wall, dysphagia, food impaction and chest and abdominal pain and/or an elevated level of a EoE-associated biomarker.
  • the term specifically includes subjects who show the presence of > 15 eosinophils per high power field in the esophagus.
  • the term also includes subjects with elevated peripheral eosinophil counts (e.g. , >100, >150, >200, or >300 cells/ ⁇ ) or elevated serum IgE (>150kU/L).
  • the present methods may be used to treat patients who exhibit pathology and symptoms that are observed in subjects with chronic esophagitis including in gastroesophageal reflux disease (GERD).
  • GERD gastroesophageal reflux disease
  • the term "a subject in need thereof includes subjects that are non-responsive to or resistant to anti- GERD therapy.
  • the present methods may be used to treat subjects that are resistant to proton pump inhibitors (PPI).
  • PPI proton pump inhibitors
  • a subject in need thereof may include a subset of population that is more susceptible to EoE or may show an elevated level of an EoE-associated biomarker.
  • a subject in need thereof may include a subject suffering from an atopic disease or disorder such as food allergy, atopic dermatitis, asthma, allergic rhinitis and allergic conjunctivitis.
  • the term "a subject in need thereof includes a patient who, prior to or at the time of administration of the IL-4/IL-13 pathway inhibitor, has or is diagnosed with a disease or disorder selected from the group consisting of food allergy, atopic dermatitis, asthma, allergic rhinitis and allergic conjunctivitis.
  • a subject in need thereof may include patients with inherited connective tissue disorders. Such a subject population may show an elevated level of an EoE-associated biomarker such as, e.g. , IgE, eotaxin-3, periostin, IL-5, or IL-13.
  • EoE-associated biomarker such as, e.g. , IgE, eotaxin-3, periostin, IL-5, or IL-13.
  • a subject in need thereof includes a patient susceptible to an allergen.
  • a subject in need thereof includes a patient who may exhibit one of the following characteristics: (a) is prone to allergic reactions or responses when exposed to one or more allergens; (b) has previously exhibited an allergic response or reaction to one or more allergens; (c) has a known history of allergies; and/or (d) exhibits a sign or symptom of an allergic response or anaphylaxis.
  • the patient is allergic to an allergen associated with EoE or that renders the subject susceptible and/or prone to developing EoE.
  • allergen includes any substance, chemical, particle or composition that is capable of stimulating an allergic response in a susceptible individual. Allergens may be contained within or derived from a food item such as, e.g. , dairy products (e.g. , cow's milk), egg, wheat, soy, corn, rye, fish, shellfish, peanuts and tree nuts. Alternatively, an allergen may be contained within or derived from a non-food item such as, e.g. , dust (e.g., containing dust mite), pollen, insect venom (e.g. , venom of bees, wasps, mosquitoes, etc.), mold, animal dander, latex, medication, drugs, ragweed, grass and birch.
  • a food item such as, e.g. , dairy products (e.g. , cow's milk), egg, wheat, soy, corn, rye, fish, shellfish, peanuts and tree nuts.
  • an allergen may be contained within or
  • a subject in need thereof includes a subset of population that exhibits an allergic reaction to a food allergen.
  • a subject in need thereof may include a subject who has an allergy to an allergen contained in a food item including, but not limited to, a dairy product, egg, wheat, soy, corn, rye, fish, shellfish, peanut, a tree nut, beef, chicken, oat, barley, pork, green beans, and fruits such as apple and pineapple.
  • the term includes a subject allergic to a non-food allergen such as allergens derived from dust, mold, insects, plants including pollen, and pets such as cats and dogs.
  • a non-food allergen such as allergens derived from dust, mold, insects, plants including pollen, and pets such as cats and dogs.
  • non-food allergens also known as environmental allergens or aeroallergens
  • non-food allergens include, but are not limited to, house dust mite allergens, pollen allergens, animal dander allergens, insect venom, grass allergens, and latex.
  • the phrases "allergic response,” “allergic reaction,” “allergic symptom,” and the like include one or more signs or symptoms selected from the group consisting of urticaria (e.g. , hives), angioedema, rhinitis, asthma, vomiting, sneezing, runny nose, sinus inflammation, watery eyes, wheezing, bronchospasm, reduced peak expiratory flow (PEF), gastrointestinal distress, flushing, swollen lips, swollen tongue, reduced blood pressure, anaphylaxis, and organ dysfunction/failure.
  • An “allergic response,” “allergic reaction,” “allergic symptom,” etc. also includes immunological responses and reactions such as, e.g. , increased IgE production, increased allergen-specific immunoglobulin production and/or eosinophilia.
  • the methods herein are for the treatment of adults, adolescents or children.
  • An adult is >18 years of age
  • an adolescent is ⁇ 2 and ⁇ 18 years of age and children are ⁇ 12 years of age.
  • the methods herein may be used to treat EoE in children who are ⁇ 3 years old.
  • an inhibitor of the IL-4/IL-13 pathway is used to treat moderate-to severe EoE in subjects that are not adequately controlled with standard-of-care treatment (e.g. , oral corticosteroids, dilation, etc.)
  • the subject can be an adult, an adolescent or a child.
  • the present invention also includes methods to increase esophageal distensibility.
  • the methods according to this aspect of the invention comprise administering to the patient in need thereof one or more doses of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor, thereby increasing the distensibility of the esophagus in the patient.
  • the present invention also includes methods for reducing eosinophilic infiltration.
  • the methods according to this aspect of the invention comprise administering to the patient in need thereof one or more doses of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to reduce or eliminate the number of eosinophils, e.g. , in the esophageal mucosa.
  • eosinophilic infiltration refers to the presence of eosinophils in an organ or tissue including blood, esophagus, stomach, duodenum, and ileum of a subject.
  • the term "eosinophilic infiltration” refers to presence of eosinophils in the mucosal lining of a region of the gastro-intestinal tract including, but not limited to, esophagus and stomach. Eosinophilic infiltration is analyzed, for example, in an esophageal tissue biopsy of a subject suffering from EoE. According to particular embodiments, “eosinophilic infiltration” refers to the presence of > 15 eosinophils per high power field in the esophagus.
  • high power field refers to a standard total magnification of 400X by a microscope used to view eosinophils in a tissue, e.g., from the esophagus of a subject.
  • "eosinophilic infiltration” includes infiltration into a tissue by leucocytes, for example, lymphocytes, neutrophils and mast cells. The leucocyte infiltration into, e.g.
  • esophageal tissue can be detected by cell surface markers such as eosinophil-specific markers (e.g., CDl lc Low/Ne , SiglecF + , F4/80 + , EMR1 + , Siglec 8 + , and MBP2 + ), macrophage-specific markers (e.g., CDl lb + , F4/80 + , CD14 + , EMR1 + , and CD68 + ), neutrophil-specific markers (e.g., CDl lb + , Ly6G + , Ly6C + , CDl lb + , and CD66b + ), and T-cell-specific markers (e.g., CD3 + CD4 + CD8 + ).
  • eosinophil-specific markers e.g., CDl lc Low/Ne , SiglecF + , F4/80 + , EMR1 + , Siglec 8 + , and MBP2 +
  • a reduction in esophagus eosinophils means that the number of eosinophils and other leucocytes measured in the esophagus of a subject with EoE and who has been treated with an IL-4/IL-13 pathway inhibitor, is at least 5%, 10%, 20%, 50%, 70%, 80%, or 90% lower than the esophagus eosinophils measured in the same or an equivalent subject that has not been treated with the IL-4/IL-13 pathway inhibitor.
  • reducing eosinophilic infiltration means detecting less than 15 eosinophils per high power field, more preferably less than 10 eosinophils, less than 9 eosinophils, less than 8 eosinophils, less than 7 eosinophils, less than 6 eosinophils, or less than 5 eosinophils per high power field in a biopsy of the esophageal mucosa.
  • a reduction in esophagus eosinophils means that no eosinophils are detected in the esophageal mucosa of a subject.
  • the present invention includes methods for treating, preventing or reducing the severity of eosinophilic esophagitis comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to a subject in need thereof, wherein the pharmaceutical composition is administered to the subject in multiple doses, e.g., as part of a specific therapeutic dosing regimen.
  • the therapeutic dosing regimen may comprise administering multiple doses of the pharmaceutical composition to the subject at a frequency of about once a day, once every two days, once every three days, once every four days, once every five days, once every six days, once a week, once every two weeks, once every three weeks, once every four weeks, once a month, once every two months, once every three months, once every four months, or less frequently.
  • the methods of the present invention comprise administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor in combination with a second therapeutic agent.
  • the second therapeutic agent may be an agent selected from the group consisting of, e.g.
  • an IL-lbeta inhibitor an IL-5 inhibitor, an IL-9 inhibitor, an IL-13 inhibitor, an IL-17 inhibitor, an IL-25 inhibitor, a TNF alpha inhibitor, an eotaxin-3 inhibitor, an IgE inhibitor, a prostaglandin D2 inhibitor, an immunosuppressant, a topical corticosteroid, an oral corticosteroid (e.g., budesonide), a systemic corticosteroid, an inhaled corticosteroid, a glucocorticoid, a proton pump inhibitor, a decongestant, an antihistamine, and a non-steroidal anti-inflammatory drug (NSAID).
  • the IL-4/IL-13 pathway inhibitor of the invention may be administered in combination with therapy including esophagus dilation, allergen removal and diet management.
  • the phrase 'in combination with means that the phrase 'in combination with"
  • the present invention includes methods comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to a subject who is on a background anti-allergy therapeutic regimen.
  • the background anti- allergy therapeutic regimen may comprise a course of administration of, e.g., steroids, antihistamines, decongestants, anti-IgE agents, etc.
  • the IL-4/IL-13 pathway inhibitor e.g.
  • an anti-IL-4R antibody may be added on top of the background anti-allergy therapeutic regimen.
  • the IL-4/IL-13 pathway inhibitor is added as part of a "background step-down" scheme, wherein the background anti-allergy therapy is gradually withdrawn from the subject over time (e.g., in a stepwise fashion) while the IL- 4/IL-13 pathway inhibitor is administered the subject at a constant dose, or at an increasing dose, or at a decreasing dose, over time.
  • the IL-4/IL-13 pathway inhibitor is administered as a monotherapy.
  • the present invention also includes methods involving the use, quantification, and analysis of EoE-associated biomarkers.
  • EoE-associated biomarker means any biological response, cell type, parameter, protein, polypeptide, enzyme, enzyme activity, metabolite, nucleic acid, carbohydrate, or other biomolecule which is present or detectable in an EoE patient at a level or amount that is different from (e.g. , greater than or less than) the level or amount of the marker present or detectable in a non-EoE patient.
  • EoE-associated biomarkers include, but are not limited to, e.g., esophagus eosinophils, eotaxin-3 (CCL26), periostin, serum IgE (total and allergen- specific), serum IgG (total and allergen-specific), IL-13, IL-5, serum thymus and activation regulated chemokine (TARC; CCL17), thymic stromal lymphopoietin (TSLP), serum eosinophilic cationic protein (ECP), calpain 14, filaggrin (FLG), signal transducer and activator of transcription 6 (STAT6), interleukin 4 receptor (IL-4R), and eosinophil- derived neurotoxin (EDN).
  • TARC serum thymus and activation regulated chemokine
  • TSLP thymic stromal lymphopoietin
  • ECP serum eosinophilic cationic protein
  • EoE-associated biomarker also includes a gene or gene probe known in the art that is differentially expressed in a subject with EoE as compared to a subject without EoE.
  • genes which are significantly up- regulated in a subject with EoE include, but are not limited to, T-helper 2 (Th2)-associated chemokines such as CCL8, CCL23 and CCL26, periostin, cadherin-like-26, and TNFa- induced protein 6 (Blanchard et al 2006, J. Clin. Invest. 116: 536 - 547).
  • EoE-associated biomarker also includes genes that are down regulated due to EoE such as terminal differentiation proteins (e.g., filaggrin) (Blanchard et al 2006, J. Clin. Invest. 116: 536 - 547). Certain embodiments of the invention relate to use of these biomarkers for monitoring disease reversal with the administration of the IL-4/IL-13 pathway inhibitor. Methods for detecting and/or quantifying such EoE-associated biomarkers are known in the art; kits for measuring such EoE-associated biomarkers are available from various commercial sources; and various commercial diagnostic laboratories offer services which provide measurements of such biomarkers as well.
  • terminal differentiation proteins e.g., filaggrin
  • methods for treating EoE comprise: (a) selecting a subject who exhibits a level of at least one EoE- associated biomarker prior to or at the time of treatment which signifies the disease state; and (b) administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of an IL-4/IL-13 pathway inhibitor.
  • the subject is selected on the basis of an elevated level of IgE or eotaxin-3.
  • methods for treating EoE comprise administering to a subject a pharmaceutical composition comprising a therapeutically effective amount of an IL-4/IL-13 pathway inhibitor, wherein administration of the pharmaceutical composition to the subject results in a decrease in at least one EoE-associated biomarker (e.g., esophagus eosinophils, eotaxin-3, IgE, etc.) at a time after administration of the pharmaceutical composition, as compared to the level of the biomarker in the subject prior to the administration.
  • EoE-associated biomarker e.g., esophagus eosinophils, eotaxin-3, IgE, etc.
  • an increase or decrease in an EoE-associated biomarker can be determined by comparing (i) the level of the biomarker measured in a subject at a defined time point after administration of the pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to (ii) the level of the biomarker measured in the patient prior to the administration of the pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor (i.e. , the "baseline
  • the defined time point at which the biomarker is measured can be, e.g., at about 4 hours, 8 hours, 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 15 days, 20 days, 35 days, 40 days, 50 days, 55 days, 60 days, 65 days, 70 days, 75 days, 80 days, 85 days, or more after administration of the of the pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor.
  • a subject may exhibit a decrease in the level of one or more of IgE and/or eotaxin-3 following administration of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor (e.g. , an anti- IL-4R antibody).
  • a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor (e.g. , an anti- IL-4R antibody).
  • an IL-4/IL-13 pathway inhibitor e.g. , an anti- IL-4R antibody
  • the subject may exhibit a decrease in eotaxin-3 of about 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more from baseline (wherein “baseline” is defined as the level of eotaxin-3 in the subject just prior to the first administration).
  • the subject following administration of a first, second, third or fourth dose of a pharmaceutical composition comprising about 75 mg to about 600 mg of an anti-IL-4R antibody (e.g., dupilumab), the subject, according to the present invention, may exhibit a decrease in IgE of about 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more from baseline (wherein "baseline” is defined as the level of IgE in the subject just prior to the first administration).
  • baseline is defined as the level of IgE in the subject just prior to the first administration).
  • the present invention also includes methods for determining whether a subject is a suitable subject for whom administration of a pharmaceutical composition comprising an IL-4/IL-13 pathway antagonist would be beneficial. For example, if an individual, prior to receiving a pharmaceutical composition comprising an IL-4/IL-13 pathway antagonist, exhibits a level of an EoE-associated biomarker that signifies the disease state, the individual is therefore identified as a suitable patient for whom administration of a pharmaceutical composition of the invention (a composition comprising an anti-IL-4R antibody) would be beneficial.
  • the present invention includes methods for treating suitable subjects, wherein a suitable subject may be more susceptible to EoE, for example, due to food allergy, or an atopic disease.
  • the present invention includes methods comprising administering an IL-4/IL-13 pathway antagonist to subjects who have food allergy, atopic dermatitis, asthma, allergic rhinitis or allergic conjunctivitis.
  • the present invention includes methods comprising administering an IL-4/IL-13 pathway antagonist to subjects who have, Mendelian- inherited connective tissue disorders, e.g. , Marfan syndrome, Loeys-Dietz syndrome, hypermobile Ehlers Danlos syndrome (EDS) or joint hypermobility syndrome (JHS).
  • Such subject populations may have an elevated level of an EoE-associated biomarker.
  • an individual may be identified as a good candidate for anti-IL-4/IL-13 therapy if the individual exhibits one or more of the following: (i) an eotaxin-3 level greater than about 30 pg/ml, greater than about 40 pg/ml, greater than about 50 pg/ml, greater than about 100 pg/ml, greater than about 1500 pg/ml, greater than about 200 pg/ml, greater than about 250 pg/ml, greater than about 300 pg/ml, greater than about 350 pg/ml, greater than about 400 pg/ml, greater than about 450 pg/ml, or greater than about 500 pg/ml; or (ii) a serum IgE level greater than about 114 kU/L, greater than about 150 kU/L, greater than about 500 kU/L, greater than about 1000 kU/L, greater than about 1500 kU/L, greater than about
  • Additional criteria such as other clinical indicators of EoE (e.g. , dysphagia, thickening of the esophageal wall, and food allergy indicative of EoE), may be used in combination with any of the foregoing EoE-associated biomarkers to identify an individual as a suitable candidate for anti-IL- 4/IL-13 therapy as described elsewhere herein.
  • EoE e.g., dysphagia, thickening of the esophageal wall, and food allergy indicative of EoE
  • the present invention includes methods for improving one or more eosinophilic esophagitis (EoE)-related parameters in a subject in need thereof, wherein the methods comprise administering a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to the subject.
  • EoE eosinophilic esophagitis
  • EoE-related parameters include: (a) Straumann Dysphagia Instrument (SDI); (b) Eosinophilic Esophagitis Activity Index (EEsAI); (c) Eosinophilic Esophagitis Edema Rings Exudates Furrows and Strictures (EoE-EREFS); (d)
  • Eosinophilic Esophagitis Histological Scoring System EoE-HSS
  • Esophageal intraepithelial eosinophils Esophageal intraepithelial eosinophils
  • Esophageal distensibility Esophageal distensibility
  • An "improvement in an EoE- related parameter” means a decrease from baseline of one or more of SDI, EEsAI, EoE- EREFS, EoE-HSS, or esophageal intraepithelial eosinophils.
  • An improvement in esophageal distensibility means an increase from the baseline.
  • the term "baseline,” with regard to an EoE-related parameter means the numerical value of the EoE-related parameter for a subject prior to or at the time of administration of a pharmaceutical composition of the present invention.
  • an EoE-related parameter is quantified at baseline and at one or more time-points after administration of the pharmaceutical composition of the present invention.
  • an EoE-related parameter may be measured at day 1, day 2, day 3, day 4, day 5, day 6, day 7, day 8, day 9, day 10, day 11, day 12, day 14, day 15, day 22, day 25, day 29, day 36, day 43, day 50, day 57, day 64, day 71, day 85; or at the end of week 1, week 2, week 3, week 4, week 5, week 6, week 7, week 8, week 9, week 10, week 11, week 12, week 13, week 14, week 15, week 16, week 17, week 18, week 19, week 20, week 21, week 22, week 23, week 24, or longer, after the initial treatment with a pharmaceutical composition of the present invention.
  • the difference between the value of the parameter at a particular time point following initiation of treatment and the value of the parameter at baseline is used to establish whether there has been an "improvement" (e.g., a decrease) in the EoE-related parameter.
  • SDI Straumann Dysphagia Instrument
  • the total SDI score ranges from 0 to 9.
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in a decrease in SDI score of 3 points from the baseline.
  • the present invention includes therapeutic methods that result in a decrease from baseline in SDI score of decrease of 1, 2, 3, 4, 5, 6 or more points from baseline in SDI.
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in a decrease at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, or more at day 4, 8, 15, 22, 25, 29, 36, 43, 50, 57, 64, 71, 85 or later following administration of the IL-4/IL-13 pathway inhibitor (e.g.
  • administering following subcutaneous administration of about 300 mg of an anti-IL-4R antibody or antigen-binding fragment thereof).
  • administration of an IL-4/IL-13 pathway inhibitor to a subject results in a decrease from baseline in SDI of at least 40%.
  • Eosinophilic Esophagitis Activity Index (EEsAI).
  • the EEsAI is a non-validated, multimodular index in development at University Hospital Dreispital (Berne, Switzerland) (Schoepfer 2014), a part of the international EEsAI study group.
  • the EEsAI PRO module (questionnaire) used in this study includes items related to the intensity and frequency of dysphagia, the influence of specific food groups on dysphagia symptoms, and other symptoms independent of eating or drinking (i.e., heartburn, acid regurgitation, and chest pain).
  • the total EEsAI PRO score ranges from 0 to 100 ( Figure 1), wherein higher scores indicate worse symptoms.
  • the score consists of 5 parts: frequency of trouble swallowing, duration of trouble swallowing, pain when swallowing, visual dysphagia question, and avoidance, modification and slow eating (AMS).
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in a decrease in EEsAI score.
  • the present invention includes therapeutic methods which result in a decrease from baseline in EEsAI score of at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, or more at day 4, 8, 15, 22, 25, 29, 36, 43, 50, 57, 64, 71, 85 or later following administration of the IL-4/IL-13 pathway inhibitor (e.g.
  • administering following subcutaneous administration of about 300 mg of an anti-IL-4R antibody or antigen-binding fragment thereof).
  • administration of an IL-4/IL-13 pathway inhibitor to a subject results in a decrease from baseline in EEsAI score of at least 30% after administration.
  • Eosinophilic Esophagitis Edema Rings Exudates Furrows and Strictures (EoE- EREFS).
  • the EoE-EREFS edema, rings, exudates, furrows, strictures
  • This instrument includes a total of 17 items related to the presence and severity of esophageal features.
  • the specific esophageal features include: rings (concentric rings around esophagus - absent, mild, moderate, severe, not applicable); strictures (narrowing of the esophagus - yes, no, not applicable); diameter of the stricture (if applicable);
  • exudates (refer to white plaques - absent, mild, severe), furrows (vertical lines down the esophagus - absent, present); edema (loss of vascular markings of the mucosa - absent, present); crepe paper esophagus (absent, present); overall general appearance
  • EoE findings i.e., fixed rings, strictures, whitish exudates, furrowing, edema, and crepe paper mucosa.
  • mucosal changes associated with gastroesophageal reflux disease are recorded using the Los Angeles classification system for erosions (No Erosions or LA Classification A, B, C, D).
  • the EoE esophageal characteristics are analyzed based on the EoE-EREFS, a validated scoring system for inflammatory and remodeling features of disease using both overall scores and scores for each individual characteristic (Hirano 2014).
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in a decrease in EoE-EREFS score.
  • the present invention includes therapeutic methods which result in a decrease from baseline in EREFS score of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or more at day 4, 8, 15, 22, 25, 29, 36, 43, 50, 57, 64, 71, 85 or later following administration of the IL-4/IL-13 pathway inhibitor (e.g. , following subcutaneous administration of about 300 mg of an anti-IL-4R antibody or antigen-binding fragment thereof).
  • Eosinophilic Esophagitis Histological Scoring System (EoE-HSS).
  • the EoE- HSS generate separate severity (grade) and extent (stage) disease scores.
  • the score is used to measure 8 histologic features (parameters) of EoE from 3 different regions (proximal, mid and distal) of the esophagus (Collins et al 2017).
  • the 8 parameters include: eosinophil density, basal zone hyperplasia, eosinophil abscesses, eosinophil surface layering, dilated intercellular spaces, surface epithelial alteration, dyskeratotic cells, and lamina limba fibrosis.
  • a scale of 0 - 3 is used for each parameter, both grade and stage (0 being least inflamed, normal).
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in a decrease in EoE- HSS score.
  • the present invention includes therapeutic methods which result in a decrease from baseline in EoE-HSS of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or more at day 4, 8, 15, 22, 25, 29, 36, 43, 50, 57, 64, 71, 85 or later following administration of the IL-4/IL-13 pathway inhibitor (e.g.
  • administering following subcutaneous administration of about 300 mg of an anti-IL-4R antibody or antigen-binding fragment thereof).
  • administration of an IL-4/IL-13 pathway inhibitor to a subject results in a decrease from baseline in EoE-HSS score of at least 50%.
  • Esophageal intraepithelial eosinophils It refers to >15 eosinophils per high powered field (hpf) in esophageal biopsies. Peak intraepithelial eosinophils refers to >15 eosinophils per high powered field in at least 2 of 3 esophageal regions sampled.
  • administration of an IL-4/IL- 13 pathway inhibitor to a patient results in a decrease in peak intraepithelial eosinophils.
  • the present invention includes therapeutic methods which result in a decrease from baseline in intraepithelial eosinophils of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85% or more at day 4, 8, 15, 22, 25, 29, 36, 43, 50, 57, 64, 71, 85 or later following administration of the IL-4/IL-13 pathway inhibitor (e.g. , following subcutaneous administration of about 300 mg of an anti- IL-4R antibody or antigen-binding fragment thereof).
  • administration of an IL-4/IL-13 pathway inhibitor to a subject results in a decrease from baseline in intraepithelial eosinophils of at least 85%.
  • Esophageal Distensibilitv Esophageal Distensibilitv. Esophageal distensibility is assessed by using the endoluminal functional lumen imaging probe (EndoFLIP, Crospon, Ireland) to measure the diameter of the esophageal lumen and pressure.
  • EndoFLIP device is a catheter based procedure that measures the cross sectional area at multiple sites along the esophagus with simultaneous intraluminal pressure recordings during volumetric distension of the esophagus.
  • the analyses of cross sectional area versus pressure relationships of the esophagus allow for determination of esophageal compliance as well as the distensibility plateau (DP).
  • the DP has been shown to be significantly reduced in patients with EoE compared to healthy controls (Kwiatek 2011).
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in an increase in esophageal distensibility.
  • the present invention includes therapeutic methods which result in an increase from baseline in esophageal distensibility of at least about 5%, 10%, 15%, 20%, 25% or more at the end of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or later following administration of the IL-4/IL-13 pathway inhibitor (e.g. , following subcutaneous administration of about 300 mg of an anti- IL-4R antibody or antigen-binding fragment thereof).
  • administration of an IL-4/IL-13 pathway inhibitor to a subject results in an increase from baseline in esophageal distensibility of at least 10%, as measured by impedance planimetry.
  • EoE-QoL-A Adult Eosinophilic Esophagitis Quality of Life
  • the EoE-QOL-A questionnaire is a validated disease-specific measure of health-related quality of life in EoE patients (Taft 2011).
  • the instrument used in this study, the EoE-QOL-A v.3.0 includes 30 items related to established domains such as social functioning, emotional functioning, and disease impact of daily life experiences.
  • the EoE-QOL-A has a 1-week recall period. The items are graded on a 5-point scale: 'Not at All,' 'Slightly,' 'Moderately,' 'Quite a bit,' and 'Extremely'.
  • administration of an IL-4/IL-13 pathway inhibitor to a patient results in an increase in quality of life parameters in the patient.
  • the methods of the present invention comprise administering to a subject in need thereof a therapeutic composition comprising an IL-4/IL-13 pathway inhibitor.
  • an "IL-4/IL-13 pathway inhibitor” (also referred to herein as an "IL-4/IL-13 pathway antagonist," an "IL-4/IL-13 pathway blocker,” etc.) is any agent that inhibits or attenuates at least one of: (i) the binding of IL-4 and/or IL-13 to their respective receptors; (ii) signaling and/or activity of IL-4 and/or IL-13; and/or (iii) the downstream signaling/activity that results from binding of IL-4 and/or IL-13 to their respective receptors.
  • Exemplary IL-4/IL-13 pathway inhibitors include, but are not limited to, anti- IL-4 antibodies (e.g., the antibodies disclosed in US Patent 7740843, and US Patent Application Publications 20100297110, 20160207995 ), anti-IL-13 antibodies (e.g. , the antibodies disclosed in US Patents 7501121, 7674459, 7807788, 7910708, 7915388, 7935343, 8088618, 8691233, 9605065, US Patent Application Publications 20060073148, 20080044420, and EP2627673B1), bispecific antibodies that bind to IL-4 and IL-13 (e.g. , the antibodies disclosed in US Patent 8388965, US Patent Application Publications 20110008345, 20130251718, 20160207995), and IL-4 receptor (IL-4R) inhibitors (described below).
  • anti- IL-4 antibodies e.g., the antibodies disclosed in US Patent 7740843, and US Patent Application Publications 20100297110, 2016
  • an "IL-4R inhibitor” (also referred to herein as an "IL-4/IL-13 pathway inhibitor,” an “IL-4Ra antagonist,” an “IL-4R blocker,” an “IL-4Ra blocker,” etc.) is any agent that binds to or interacts with IL-4Ra or an IL-4R ligand, and inhibits or attenuates the normal biological signaling function a type 1 and/or a type 2 IL-4 receptor.
  • Human IL-4Ra has the amino acid sequence of SEQ ID NO: 11.
  • a type 1 IL-4 receptor is a dimeric receptor comprising an IL-4Ra chain and a yc chain.
  • a type 2 IL-4 receptor is a dimeric receptor comprising an IL-4Ra chain and an IL-13Ral chain.
  • Type 1 IL-4 receptors interact with and are stimulated by IL-4
  • type 2 IL-4 receptors interact with and are stimulated by both IL-4 and IL-13.
  • the IL-4R inhibitors that can be used in the methods of the present invention may function by blocking IL-4-mediated signaling, IL-13 -mediated signaling, or both IL-4- and IL-13-mediated signaling.
  • the IL- 4R inhibitors of the present invention may thus prevent the interaction of IL-4 and/or IL- 13 with a type 1 or type 2 receptor.
  • Non-limiting examples of categories of IL-4R inhibitors include IL-4 muteins (e.g. , pitrakinra), small molecule IL-4R inhibitors, anti-IL-4R aptamers, peptide-based IL- 4R inhibitors (e.g. , "peptibody” molecules), “receptor-bodies” (e.g., engineered molecules comprising the ligand-binding domain of an IL-4R component), and antibodies or antigen- binding fragments of antibodies that specifically bind human IL-4Ra.
  • IL- 4R inhibitors also include antigen-binding proteins that specifically bind IL-4 and/or IL- 13.
  • the IL- 4/IL-13 pathway inhibitor is an anti-IL-4Ra antibody or antigen-binding fragment thereof.
  • antibody includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, as well as multimers thereof (e.g., IgM).
  • each heavy chain comprises a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region comprises three domains, CHT , CH2 and CH3.
  • Each light chain comprises a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region comprises one domain (CLI).
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the FRs of the anti-IL-4R antibody may be identical to the human germline sequences, or may be naturally or artificially modified.
  • An amino acid consensus sequence may be defined based on a side-by-side analysis of two or more CDRs.
  • antibody also includes antigen-binding fragments of full antibody molecules.
  • antigen-binding portion of an antibody, “antigen- binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • Antigen-binding fragments of an antibody may be derived, e.g. , from full antibody molecules using any suitable standard techniques such as proteolytic digestion or recombinant genetic engineering techniques involving the manipulation and expression of DNA encoding antibody variable and optionally constant domains. Such DNA is known and/or is readily available from, e.g.
  • DNA libraries including, e.g. , phage-antibody libraries
  • the DNA may be sequenced and manipulated chemically or by using molecular biology techniques, for example, to arrange one or more variable and/or constant domains into a suitable configuration, or to introduce codons, create cysteine residues, modify, add or delete amino acids, etc.
  • Non-limiting examples of antigen-binding fragments include: (i) Fab fragments; (ii) F(ab')2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide.
  • CDR complementarity determining region
  • engineered molecules such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR-grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g. monovalent nanobodies, bivalent nanobodies, etc.), small modular molecules
  • SMIPs immunopharmaceuticals
  • shark variable IgNAR domains are also encompassed within the expression "antigen-binding fragment,” as used herein.
  • An antigen-binding fragment of an antibody will typically comprise at least one variable domain.
  • the variable domain may be of any size or amino acid composition and will generally comprise at least one CDR that is adjacent to or in frame with one or more framework sequences.
  • the VH and VL domains may be situated relative to one another in any suitable arrangement.
  • the variable region may be dimeric and contain VH- VH, VH-VL or VL-VL dimers.
  • the antigen-binding fragment of an antibody may contain a monomeric VH or VL domain.
  • an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain.
  • variable and constant domains that may be found within an antigen-binding fragment of an antibody of the present invention include: (i) VH- CHI ; (ii) VH-CH2; (iii) VH-CH3; (iv) VH-CH1-CH2; (V) VH-CH1-CH2-CH3; (vi) VH-CH2- CH3; (vii) VH-CL; (viii) VL-CH1; (ix) VL-CH2; (X) VL-CH3; (xi) VL-CH1-CH2; (xii) VL-CH1- CH2-CH3; (xiii) VL-CH2-CH3; and (xiv) VL-CL.
  • variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region.
  • a hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide molecule.
  • an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric VH or VL domain (e.g. , by disulfide bond(s)).
  • antibody also includes multispecific (e.g. , bispecific) antibodies.
  • a multispecific antibody or antigen-binding fragment of an antibody will typically comprise at least two different variable domains, wherein each variable domain is capable of specifically binding to a separate antigen or to a different epitope on the same antigen.
  • Any multispecific antibody format may be adapted for use in the context of an antibody or antigen-binding fragment of an antibody of the present invention using routine techniques available in the art.
  • the present invention includes methods comprising the use of bispecific antibodies wherein one arm of an immunoglobulin is specific for IL-4Ra or a fragment thereof, and the other arm of the immunoglobulin is specific for a second therapeutic target or is conjugated to a therapeutic moiety.
  • Exemplary bispecific formats that can be used in the context of the present invention include, without limitation, e.g. , scFv-based or diabody bispecific formats, IgG-scFv fusions, dual variable domain (DVD)-Ig, Quadroma, knobs-into-holes, common light chain (e.g., common light chain with knobs-into-holes, etc.), CrossMab, CrossFab, (SEED) body, leucine zipper, Duobody, IgGl/IgG2, dual acting Fab (DAF)-IgG, and Mab 2 bispecific formats (see, e.g. , Klein et al.
  • Bispecific antibodies can also be constructed using peptide/nucleic acid conjugation, e.g., wherein unnatural amino acids with orthogonal chemical reactivity are used to generate site-specific antibody - oligonucleotide conjugates which then self-assemble into multimeric complexes with defined composition, valency and geometry. (See, e.g., Kazane et al., J. Am. Chem. Soc. [Epub: Dec. 4, 2012]).
  • the antibodies used in the methods of the present invention may be human antibodies.
  • the term "human antibody,” as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may nonetheless include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the term "human antibody,” as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • the antibodies used in the methods of the present invention may be recombinant human antibodies.
  • the term "recombinant human antibody,” as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further below), antibodies isolated from a recombinant, combinatorial human antibody library (described further below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g. , Taylor et al. (1992) Nucl. Acids Res.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • the antibodies used in the methods of the present invention specifically bind IL-4Ra.
  • the term "specifically binds,” or the like, means that an antibody or antigen-binding fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions. Methods for determining whether an antibody specifically binds to an antigen are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like.
  • an antibody that "specifically binds" IL-4Ra includes antibodies that bind IL-4Ra or portion thereof with a KD of less than about 500 nM, less than about 300 nM, less than about 200 nM, less than about 100 nM, less than about 90 nM, less than about 80 nM, less than about 70 nM, less than about 60 nM, less than about 50 nM, less than about 40 nM, less than about 30 nM, less than about 20 nM, less than about 10 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, less than about 1 nM or less than about 0.5 nM, as measured in a surface plasmon resonance assay.
  • An isolated antibody that specifically binds human IL-4Ra may, however, have cross-reactivity to other antigens, such as IL-4Ra molecules from other (n
  • the IL- 4/IL-13 pathway inhibitor is an anti-IL-4Ra antibody, or antigen-binding fragment thereof comprising a heavy chain variable region (HCVR), light chain variable region (LCVR), and/or complementarity determining regions (CDRs) comprising any of the amino acid sequences of the anti-IL-4R antibodies as set forth in US Patent No. 7,608,693.
  • HCVR heavy chain variable region
  • LCVR light chain variable region
  • CDRs complementarity determining regions
  • the anti-IL-4Ra antibody or antigen-binding fragment thereof that can be used in the context of the methods of the present invention comprises the heavy chain complementarity determining regions (HCDRs) of a heavy chain variable region (HCVR) comprising the amino acid sequence of SEQ ID NO: 1 and the light chain complementarity determining regions (LCDRs) of a light chain variable region (LCVR) comprising the amino acid sequence of SEQ ID NO: 2.
  • HCDRs heavy chain complementarity determining regions
  • LCDRs light chain complementarity determining regions of a light chain variable region
  • the anti-IL-4Ra antibody or antigen-binding fragment thereof comprises three HCDRs (HCDR1, HCDR2 and HCDR3) and three LCDRs (LCDR1, LCDR2 and LCDR3), wherein the HCDR1 comprises the amino acid sequence of SEQ ID NO: 3; the HCDR2 comprises the amino acid sequence of SEQ ID NO: 4; the HCDR3 comprises the amino acid sequence of SEQ ID NO: 5; the LCDRl comprises the amino acid sequence of SEQ ID NO: 6; the LCDR2 comprises the amino acid sequence of SEQ ID NO: 7; and the LCDR3 comprises the amino acid sequence of SEQ ID NO: 8.
  • the anti-IL-4R antibody or antigen-binding fragment thereof comprises an HCVR comprising SEQ ID NO: 1 and an LCVR comprising SEQ ID NO: 2.
  • the methods of the present invention comprise the use of an anti-IL-4R antibody, wherein the antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9.
  • the anti-IL-4R antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • An exemplary antibody comprising a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and a light chain comprising the amino acid sequence of SEQ ID NO: 10 is the fully human anti-IL-4R antibody known as dupilumab.
  • the methods of the present invention comprise the use of dupilumab, or a bioequivalent thereof.
  • bioequivalent refers to anti-IL-4R antibodies or IL-4R-binding proteins or fragments thereof that are pharmaceutical equivalents or pharmaceutical alternatives whose rate and/or extent of absorption do not show a significant difference with that of dupilumab when administered at the same molar dose under similar experimental conditions, either single dose or multiple dose.
  • the term refers to antigen-binding proteins that bind to IL-4R which do not have clinically meaningful differences with dupilumab in their safety, purity and/or potency.
  • AMG317 Corren et al, 2010, Am J Respir Crit Care Med., 7S7(8):788-796
  • MEDI 9314 or any of the anti-IL-4Ra antibodies as set forth in US Patent No. 7,186,809, US Patent No. 7,605,237, US Patent No. 7,638,606, US Patent No. 8,092,804, US Patent No. 8,6
  • the anti-IL-4Ra antibodies used in the context of the methods of the present invention may have pH-dependent binding characteristics.
  • an anti-IL-4Ra antibody for use in the methods of the present invention may exhibit reduced binding to IL-4Ra at acidic pH as compared to neutral pH.
  • an anti-IL-4Ra antibody of the invention may exhibit enhanced binding to its antigen at acidic pH as compared to neutral pH.
  • the expression “acidic pH” includes pH values less than about 6.2, e.g., about 6.0, 5.95, 5.9, 5.85, 5.8, 5.75, 5.7, 5.65, 5.6, 5.55, 5.5, 5.45, 5.4, 5.35, 5.3, 5.25, 5.2, 5.15, 5.1, 5.05, 5.0, or less.
  • neutral pH means a pH of about 7.0 to about 7.4.
  • neutral pH includes pH values of about 7.0, 7.05, 7.1, 7.15, 7.2, 7.25, 7.3, 7.35, and 7.4.
  • "reduced binding to IL-4Ra at acidic pH as compared to neutral pH” is expressed in terms of a ratio of the KD value of the antibody binding to IL- 4Ra at acidic pH to the KD value of the antibody binding to IL-4Ra at neutral pH (or vice versa).
  • an antibody or antigen-binding fragment thereof may be regarded as exhibiting "reduced binding to IL-4Ra at acidic pH as compared to neutral pH” for purposes of the present invention if the antibody or antigen-binding fragment thereof exhibits an acidic/neutral KD ratio of about 3.0 or greater.
  • the acidic/neutral KD ratio for an antibody or antigen-binding fragment of the present invention can be about 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 10.5, 11.0, 11.5, 12.0, 12.5, 13.0, 13.5, 14.0, 14.5, 15.0, 20.0, 25.0, 30.0, 40.0, 50.0, 60.0, 70.0, 100.0, or greater.
  • Antibodies with pH-dependent binding characteristics may be obtained, e.g. , by screening a population of antibodies for reduced (or enhanced) binding to a particular antigen at acidic pH as compared to neutral pH. Additionally, modifications of the antigen-binding domain at the amino acid level may yield antibodies with pH-dependent characteristics. For example, by substituting one or more amino acids of an antigen- binding domain (e.g., within a CDR) with a histidine residue, an antibody with reduced antigen-binding at acidic pH relative to neutral pH may be obtained.
  • the expression "acidic pH” means a pH of 6.0 or less.
  • the present invention includes methods that comprise administering an IL-4/IL- 13 pathway inhibitor to a subject wherein the IL-4/IL-13 pathway inhibitor is contained within a pharmaceutical composition.
  • the pharmaceutical compositions of the invention may be formulated with suitable carriers, excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. "Compendium of excipients for parenteral formulations" PDA (1998) J Pharm Sci Technol 52:238-311.
  • Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g. , encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al, 1987, J. Biol. Chem. 262: 4429-4432).
  • Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g. , oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
  • epithelial or mucocutaneous linings e.g. , oral mucosa, rectal and intestinal mucosa, etc.
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe.
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused.
  • a disposable pen delivery device there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used.
  • polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), 1974, CRC Pres., Boca Raton, Florida.
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g. , Goodson, 1984, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138).
  • Other controlled release systems are discussed in the review by Langer, 1990, Science 249: 1527-1533.
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by known methods.
  • the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • a sterile aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g.
  • ethanol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (poly oxy ethylene (50 mol) adduct of hydrogenated castor oil)
  • the oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • the injection thus prepared is preferably filled in an appropriate ampoule.
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • compositions comprising an anti-IL-4R antibody that can be used in the context of the present invention are disclosed, e.g., in US Patent 8,945,559.
  • the present invention includes methods comprising administering to a subject an IL-4/IL-13 pathway inhibitor at a dosing frequency of about four times a week, twice a week, once a week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, once every six weeks, once every eight weeks, once every twelve weeks, or less frequently so long as a therapeutic response is achieved.
  • an IL-4/IL-13 pathway inhibitor e.g., an anti-IL-4R antibody
  • once a week dosing at an amount of about 25 mg, 50 mg, 100 mg, 150 mg, 200 mg, or 300 mg is employed.
  • an anti-IL-4R antibody once in 2 weeks dosing at an amount of about 25 mg, 50 mg, 100 mg, 150 mg, 200 mg, or 300 mg, is employed.
  • multiple doses of an IL-4/IL-13 pathway inhibitor may be administered to a subject over a defined time course.
  • the methods according to this aspect of the invention comprise sequentially administering to a subject multiple doses of an IL-4/IL-13 pathway inhibitor.
  • “sequentially administering” means that each dose of IL-4/IL-13 pathway inhibitor is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months).
  • the present invention includes methods that comprise sequentially administering to the patient a single initial dose of an IL-4/IL-13 pathway inhibitor, followed by one or more secondary doses of the IL-4/IL-13 pathway inhibitor, and optionally followed by one or more tertiary doses of the IL-4/IL-13 pathway inhibitor.
  • the terms “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration of the IL-4/IL-13 pathway inhibitor.
  • the “initial dose” is the dose that is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”);
  • the “secondary doses” are the doses that are administered after the initial dose;
  • the “tertiary doses” are the doses that are administered after the secondary doses.
  • the initial, secondary, and tertiary doses may all contain the same amount of IL-4/IL-13 pathway inhibitor, but generally may differ from one another in terms of frequency of administration.
  • the amount of IL-4/IL-13 pathway inhibitor contained in the initial, secondary and/or tertiary doses varies from one another (e.g. , adjusted up or down as appropriate) during the course of treatment.
  • the initial dose comprises a first amount of the antibody or antigen-binding fragment thereof and the one or more secondary doses each comprise a second amount of the antibody or antigen-binding fragment thereof.
  • the first amount of antibody or fragment thereof (initial dose) is 1.5x, 2x, 2.5x, 3x, 3.5x, 4x, or 5x the second amount of the antibody or antigen-binding fragment thereof (secondary dose).
  • one or more (e.g., 1, 2, 3, 4, or 5) doses are administered at the beginning of the treatment regimen as "loading doses" followed by subsequent doses that are administered on a less frequent basis (e.g.,
  • an IL-4/IL-13 pathway inhibitor may be administered to a patient in need thereof at a loading dose of about 300mg to about 600mg followed by one or more maintenance doses of about 25mg to about 400mg.
  • the initial dose and the one or more secondary doses each include lOmg to 600mg of the IL- 4/IL-13 pathway inhibitor, e.g., lOOmg to 400mg of the IL-4/IL-13 pathway inhibitor, e.g. , lOmg, 25mg, 50mg, lOOmg, 150mg, 200mg, 250mg, 300mg, 400mg or 500mg of the IL- 4/IL-13 pathway inhibitor.
  • each secondary and/or tertiary dose is administered 1 to 14 (e.g. , 1, 1 1 ⁇ 2, 2, 21 ⁇ 2, 3, 31 ⁇ 2, 4, 41 ⁇ 2, 5, 51 ⁇ 2, 6, 61 ⁇ 2, 7, 71 ⁇ 2, 8, 81 ⁇ 2, 9, 91 ⁇ 2, 10, 101 ⁇ 2, 11, 11 1 ⁇ 2, 12, 121 ⁇ 2, 13, 131 ⁇ 2, 14, 141 ⁇ 2, or more) weeks after the immediately preceding dose.
  • the phrase "the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose of IL-4/IL-13 pathway inhibitor that is administered to a patient prior to the administration of the very next dose in the sequence with no intervening doses.
  • the methods according to this aspect of the invention may comprise
  • any number of secondary and/or tertiary doses of an IL-4/IL-13 pathway inhibitor for example, in certain embodiments, only a single secondary dose is administered to the patient. In other embodiments, two or more (e.g. , 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient. Likewise, in certain embodiments, only a single tertiary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient.
  • each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 6 weeks after the immediately preceding dose.
  • each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 2 to 4 weeks after the immediately preceding dose.
  • the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the regimen.
  • the methods of the present invention comprise administering to the subject a corticosteroid (CS) in combination with an IL- 4/IL-13 pathway inhibitor (e.g., an anti-IL-4R antibody).
  • a corticosteroid e.g., an anti-IL-4R antibody
  • the expression “in combination with” means that the CS is administered before, after, or concurrent with the IL-4/IL-13 pathway inhibitor.
  • the term “in combination with” also includes sequential or concomitant administration of IL-4/IL-13 pathway inhibitor and CS.
  • the CS when administered "before" the IL-4/IL-13 pathway inhibitor, the CS may be administered more than 72 hours, about 72 hours, about 60 hours, about 48 hours, about 36 hours, about 24 hours, about 12 hours, about 10 hours, about 8 hours, about 6 hours, about 4 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes or about 10 minutes prior to the administration of the IL-4/IL-13 pathway inhibitor.
  • the CS When administered “after” the IL-4/IL-13 pathway inhibitor, the CS may be administered about 10 minutes, about 15 minutes, about 30 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours, about 72 hours, or more than 72 hours after the administration of the IL-4/IL-13 pathway inhibitor.
  • Administration "concurrent" with the IL-4/IL-13 pathway inhibitor means that the CS is administered to the subject in a separate dosage form within less than 5 minutes (before, after, or at the same time) of administration of the IL-4/IL-13 pathway inhibitor, or administered to the subject as a single combined dosage formulation comprising both the CS and the IL-4/IL- 13 pathway inhibitor.
  • the amount of IL-4/IL-13 pathway inhibitor (e.g. , anti-IL-4Ra antibody) administered to a subject according to the methods of the present invention is, generally, a therapeutically effective amount.
  • therapeutically effective amount means an amount of IL-4/IL-13 pathway inhibitor that results in one or more of: (a) a reduction in the severity or duration of a symptom of eosinophilic esophagitis; (b) a reduction in the number of eosinophils in esophagus; (c) increase in esophagus distensibility; (d) reduction in episodes of dysphagia; (e) prevention or alleviation of an allergic reaction; and (f) a reduction in the use or need for conventional allergy therapy (e.g. , reduced or eliminated use of antihistamines, decongestants, nasal or inhaled steroids, anti-IgE treatment, epinephrine, etc.).
  • a therapeutically effective amount can be from about 0.05 mg to about 600 mg, e.g., about 0.05 mg, about 0.1 mg, about 1.0 mg, about 1.5 mg, about 2.0 mg, about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, about 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, about 400 mg, about 410 mg, about 420 mg, about 430 mg,
  • 300 mg of an anti-IL-4R antibody is administered.
  • the amount of IL-4/IL-13 pathway inhibitor contained within the individual doses may be expressed in terms of milligrams of antibody per kilogram of patient body weight (i.e. , mg/kg).
  • the IL-4/IL-13 pathway inhibitor e.g., anti-IL-4Ra antibody
  • the present invention includes a method of reducing dysphagia comprising: (a) selecting a patient with at least one of the following characteristics: (i) the patient exhibits >1 episodes of dysphagia per week; (ii) the patient has been treated previously with high-dose proton pump inhibitors (PPIs); and (iii) the patient has had at least one prior esophageal dilation; and (b) administering a therapeutically effective amount of a pharmaceutical composition comprising an interleukin-4/interleukin-13 (IL- 4/IL-13) pathway inhibitor to the patient in need thereof.
  • PPIs proton pump inhibitors
  • the present invention includes a method of increasing esophageal distensibility comprising: (a) selecting a patient with at least one of the following characteristics: (i) the patient exhibits >1 episodes of dysphagia per week; (ii) the patient has been treated previously with high-dose PPIs; and (iii) the patient has had at least one prior esophageal dilation; and (b) administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to the patient in need thereof.
  • the present invention includes the method of Embodiment 1 or 2, wherein the patient has moderate-to-severe EoE.
  • the present invention includes a method of treating, preventing or ameliorating at least one symptom or indication of active eosinophilic esophagitis (EoE) comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an IL-4/IL-13 pathway inhibitor to a patient in need thereof.
  • EoE active eosinophilic esophagitis
  • the present invention includes the method of any one of Embodiments 1 to 4, wherein the patient has one or more characteristics selected from the group consisting of: (1) the patient has > 15 eosinophils per high powered field (hpf) in the esophagus prior to or at the time of the treatment ("baseline”); (2) prior treatment with at least one of high dose PPIs, esophageal dilation, corticosteroids, allergen withdrawal and/or diet modification; (3) the patient exhibits >1 episodes of dysphagia per week; (4) the patient is unresponsive or resistant to prior treatment with high dose PPIs or esophageal dilation; (5) the patient has a Straumann Dysphagia Instrument (SDI) score >2; (6) the patient has an Eosinophilic Esophagitis Severity and Activity Index (EEsAI) score >30, >40, or >50; (7) the patient suffers from EoE for at least 3 years; (8) the patient, prior to
  • the present invention includes the method of any one of Embodiments 1 to 5, wherein administration of the IL-4/IL-13 pathway inhibitor results in improvement of an EoE-related parameter selected from the group consisting of: (a) reduction of at least 40% from baseline in dysphagia frequency and severity, as measured by Straumann Dysphagia Instrument (SDI) score; (b) reduction of 3 points from baseline in the SDI score; (c) reduction of more than 85% from baseline in peak intraepithelial eosinophil count in proximal, mid and/or distal regions of the esophagus; (d) increase of at least 10% from baseline in esophageal distensibility, as measured by impedance planimetry; (e) decrease of more than 50% from baseline in severity and extent of disease, as measured by EoE Histology Scoring System (HSS) score; and (f) reduction of more than 30% from baseline in dysphagia, as measured by Eosinophilic Esophagitis
  • an EoE-related parameter
  • the present invention includes the method of any one of Embodiments 1 to 6, wherein the IL-4/IL-13 pathway inhibitor is an antibody or antigen- binding fragment thereof that specifically binds IL-4 receptor (IL-4R).
  • the IL-4/IL-13 pathway inhibitor is an antibody or antigen- binding fragment thereof that specifically binds IL-4 receptor (IL-4R).
  • the present invention includes the method of any one of Embodiments 1 to 7, wherein the IL-4/IL-13 pathway inhibitor is administered at a dose of about 50 - 600 mg.
  • the present invention includes the method of any one of Embodiments 1 to 8, wherein the IL-4/IL-13 pathway inhibitor is administered at a dose of about 300 mg.
  • the present invention includes the method of any one of Embodiments 1 to 7, wherein the IL-4/IL-13 pathway inhibitor is administered at an initial dose followed by one or more secondary doses, wherein each secondary dose is administered 1 to 4 weeks after the immediately preceding dose.
  • the present invention includes the method of Embodiment 10, wherein the initial dose comprises 50 - 600mg of the IL-4/IL-13 pathway inhibitor.
  • the present invention includes the method of Embodiment 10 or 11, wherein each secondary dose comprises 25 - 400mg of the IL-4/IL-13 pathway inhibitor.
  • the present invention includes the method of any one of Embodiments 10 to 12, wherein the initial dose comprises 600mg of the IL-4/IL-13 pathway inhibitor and each secondary dose comprises 300mg of the IL-4/IL-13 pathway inhibitor.
  • Embodiment 14 includes the method of Embodiment 13, wherein each secondary dose is administered one week after the immediately preceding dose.
  • the present invention includes the method of Embodiment 13, wherein each secondary dose is administered 2 weeks after the immediately preceding dose.
  • the present invention includes the method of any one of Embodiments 4 to 15, wherein the symptom or indication of EoE is selected from the group consisting of eosinophilic infiltration of the esophagus, thickening of the esophageal wall, food refusal, vomiting, abdominal pain, heartburn, regurgitation, dysphagia and food impaction.
  • the present invention includes the method of any one of Embodiments 1 to 16, wherein the patient exhibits an allergic reaction to a food allergen contained in a food item selected from the group consisting of a dairy product, egg, wheat, soy, corn, fish, shellfish, peanut, a tree nut, beef, chicken, oat, barley, pork, green beans, apple and pineapple.
  • a dairy product egg, wheat, soy, corn, fish, shellfish, peanut, a tree nut, beef, chicken, oat, barley, pork, green beans, apple and pineapple.
  • the present invention includes the method of any one of
  • the present invention includes the method of any one of
  • the present invention includes the method of Embodiment 19, wherein the EoE-associated biomarker is selected from the group consisting of eotaxin-3, periostin, serum IgE (total and allergen-specific), IL-13, IL-5, serum TARC, TSLP, serum ECP, and EDN.
  • the EoE-associated biomarker is selected from the group consisting of eotaxin-3, periostin, serum IgE (total and allergen-specific), IL-13, IL-5, serum TARC, TSLP, serum ECP, and EDN.
  • the present invention includes the method of any one of Embodiments 1 to 20, wherein the IL-4/IL-13 pathway inhibitor is administered in combination with a second therapeutic agent or therapy, wherein the second therapeutic agent or therapy is selected from the group consisting of an IL-lbeta inhibitor, an IL-5 inhibitor, an IL-9 inhibitor, an IL-13 inhibitor, an IL-17 inhibitor, an IL-25 inhibitor, a TNF alpha inhibitor, an eotaxin-3 inhibitor, an IgE inhibitor, a prostaglandin D2 inhibitor, an immunosuppressant, a topical corticosteroid, an oral corticosteroid, a systemic corticosteroid, an inhaled corticosteroid, a glucocorticoid, a proton pump inhibitor, a NSAID, esophagus dilation, allergen removal and diet management.
  • the second therapeutic agent or therapy is selected from the group consisting of an IL-lbeta inhibitor, an IL-5 inhibitor, an IL-9 inhibitor, an
  • the present invention includes the method of any one of Embodiments 1 to 21 , wherein the IL-4/IL-13 pathway inhibitor is selected from the group consisting of an anti-IL-4 antibody, an anti-IL-13 antibody, an anti-IL-4/IL-13 bispecific antibody, an IL-4 receptor (IL-4R) inhibitor, and an anti-IL-4R antibody.
  • the IL-4/IL-13 pathway inhibitor is selected from the group consisting of an anti-IL-4 antibody, an anti-IL-13 antibody, an anti-IL-4/IL-13 bispecific antibody, an IL-4 receptor (IL-4R) inhibitor, and an anti-IL-4R antibody.
  • Embodiment 23 includes the method of Embodiment 22, wherein the IL-4/IL-13 pathway inhibitor is an IL-4R inhibitor.
  • the present invention includes the method of Embodiment 22, wherein the IL-4/IL-13 pathway inhibitor is an anti-IL-4 antibody.
  • the present invention includes the method of Embodiment 22, wherein the IL-4/IL-13 pathway inhibitor is an anti-IL-13 antibody.
  • the present invention includes the method of Embodiment 22, wherein the IL-4/IL-13 pathway inhibitor is a bispecific antibody that specifically binds to IL-4 and IL-13.
  • the present invention includes the method of any one of Embodiments 1 to 23, wherein the IL-4/IL-13 pathway inhibitor is an antibody or antigen- binding fragment thereof that binds IL-4Ra and prevents the interaction of IL-4 and/or IL- 13 with a type 1 or type 2 IL-4 receptor.
  • the IL-4/IL-13 pathway inhibitor is an antibody or antigen- binding fragment thereof that binds IL-4Ra and prevents the interaction of IL-4 and/or IL- 13 with a type 1 or type 2 IL-4 receptor.
  • Embodiment 28 includes the method of Embodiment 27, wherein the antibody or antigen-binding fragment thereof prevents the interaction of IL-4 with both type 1 and type 2 IL-4 receptors.
  • the present invention includes the method of Embodiment 28, wherein the antibody or antigen-binding fragment thereof comprises the heavy chain complementarity determining regions (HCDRs) of a heavy chain variable region (HCVR) comprising the amino acid sequence of SEQ ID NO: 1 and the light chain
  • HCDRs heavy chain complementarity determining regions
  • HCVR heavy chain variable region
  • LCDRs complementarity determining regions
  • LCVR light chain variable region
  • the present invention includes the method of Embodiment 28, wherein the antibody or antigen-binding fragment thereof comprises three HCDRs (HCDR1, HCDR2 and HCDR3) and three LCDRs (LCDR1, LCDR2 and LCDR3), wherein the HCDR1 comprises the amino acid sequence of SEQ ID NO: 3; the HCDR2 comprises the amino acid sequence of SEQ ID NO: 4; the HCDR3 comprises the amino acid sequence of SEQ ID NO: 5; the LCDR1 comprises the amino acid sequence of SEQ ID NO: 6; the LCDR2 comprises the amino acid sequence of SEQ ID NO: 7; and the LCDR3 comprises the amino acid sequence of SEQ ID NO: 8.
  • the antibody or antigen-binding fragment thereof comprises three HCDRs (HCDR1, HCDR2 and HCDR3) and three LCDRs (LCDR1, LCDR2 and LCDR3)
  • the HCDR1 comprises the amino acid sequence of SEQ ID NO: 3
  • the HCDR2 comprises the amino acid sequence of SEQ ID NO: 4
  • the present invention includes the method of Embodiment 30, wherein the HCVR comprises the amino acid sequence of SEQ ID NO: 1 and the LCVR comprises the amino acid sequence of SEQ ID NO: 2.
  • the present invention includes the method of any one of Embodiments 29 to 31, wherein the antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • Embodiment 33 the present invention includes the method of any one of Embodiments 1 to 31, wherein the IL-4/IL-13 pathway inhibitor is dupilumab or a bioequivalent thereof.
  • Embodiment 34 includes the method of Embodiment 23, wherein the IL-4R inhibitor is AMG317 or MEDI9314.
  • Example 1 Clinical trial of subcutaneously administered dupilumab in adult patients with active, moderate-to-severe eosinophilic esophagitis (EoE)
  • Dupilumab is a fully human anti-IL-4R antibody comprising a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and a light chain comprising the amino acid sequence of SEQ ID NO: 10; an HCVR/LCVR amino acid sequence pair comprising SEQ ID NOs: 1 12; and heavy and light chain CDR sequences comprising SEQ ID NOs: 3 - 8.
  • SC subcutaneous
  • the secondary objectives of the study were: (1) to assess the safety, tolerability, and immunogenicity of SC doses of dupilumab in adult patients with active, moderate to severe EoE; (2) to assess the effect of dupilumab on esophageal eosinophilic infiltration; and (3) to evaluate the pharmacokinetics (PK) of dupilumab in adult patients with EoE.
  • PK pharmacokinetics
  • patient eligibility was assessed at the screening visit (to occur between day -35 and day -1). Patients who met eligibility criteria underwent day 1 baseline assessments. Patients were randomized in a 1 : 1 ratio to receive dupilumab or placebo during the 12-week double-blind treatment phase. After the 12- week double-blind treatment phase, patients were followed off study drug for an additional 16 weeks.
  • the target population included adult (18 to 65 years old) male or female patients with active EoE.
  • dysphagia is defined as trouble swallowing solid food, or having solid food stick, by patient report;
  • stable diet is defined as no initiation of single or multiple elimination diets or reintroduction of previously eliminated food groups
  • Exclusion Criteria A patient who met any of the following criteria was ineligible to participate in this study: (1) Prior participation in a dupilumab (anti-IL-4R) clinical trial; (2) Other causes of esophageal eosinophilia or the following diseases: hypereosinophilic syndromes, Churg-Strauss vasculitis, and eosinophilic gastroenteritis; (3) History of achalasia, active Helicobacter pylori infection, Crohn's disease, ulcerative colitis, celiac disease, and prior esophageal surgery prior to screening; (4) Any esophageal stricture unable to be passed with a standard, diagnostic, adult (9 tolO mm) upper endoscope or any critical esophageal stricture that requires dilation at screening; (5) History of bleeding disorders or esophageal varices; (6) Use of chronic aspirin, nonsteroidal agents, or anticoagulants within 2 weeks prior to screening.
  • Investigational drug Dupilumab SC, a loading dose of 600 mg on day 1 followed by weekly doses of 300 mg from week 1 to week 11
  • Placebo Placebo (same formulation as dupilumab without the active substance, anti-IL-4R monoclonal antibody) SC, a volume matching the dupilumab loading dose on day 1 followed by weekly doses matching the volume of dupilumab weekly dose from week 1 to week 11.
  • PPIs proton pump inhibitors
  • EOT end of treatment
  • stable dose for at least 3 months prior to screening
  • systemic leukotriene inhibitors topical, nasal, and/or inhaled corticosteroids
  • oral antihistamines for any duration
  • oral antibiotics for up to 2 weeks.
  • Medications used for the treatment of EoE (these were considered rescue medications): Swallowed topical corticosteroids, Systemic corticosteroids, Start or dose change of systemic leukotriene inhibitors, topical, nasal, and/or inhaled corticosteroids, and Systemic treatment for EoE with an immunosuppressive/immunomodulating substance (including, but not limited to, omalizumab, cyclosporine, mycophenolate-mofetil, azathioprine, methotrexate, IFN- ⁇ , or other biologies); (2) Allergen immunotherapy (SCIT and SLIT were allowed if dose was stable for 1 year or more; however, OIT was prohibited); (3) Patients who were not using PPI in the 8 weeks prior to screening were prohibited from starting PPI therapy prior to EOT visit; (4) Treatment with a live (attenuated) vaccine (Chickenpox (varicella), FluMist-
  • the primary efficacy endpoint was: Change in the Straumann Dysphagia Instrument (SDI) patient-reported outcome (PRO) score from baseline to week 10.
  • SDI Straumann Dysphagia Instrument
  • PRO patient-reported outcome
  • the secondary endpoints were: Percent change in weekly Eosinophilic
  • Esophagitis-Endoscopic Reference Score (EoE-EREFs) (endoscopy visual anatomical score) from baseline to week 12; Percentage of patients with use of rescue medication or procedure (e.g., esophageal dilation) through week 12; and Incidence of treatment- emergent adverse events (TEAEs)
  • the exploratory efficacy endpoints were: Change in mean esophageal intraepithelial eosinophil counts (eos/hpf) [calculated using peak count from each esophageal site] from baseline to week 12; Proportion of patients who achieve esophageal intraepithelial eosinophil count ⁇ 1 eos/hpf at week 12; Change in Collins Histology Score from baseline to week 12; and Change in esophageal distensibility plateau as measured by functional lumen imaging from baseline to week 12.
  • Screening/Baseline procedures The following procedures were performed only at the screening and/or baseline visit for the sole purpose of determining study eligibility or characterizing the baseline population: serum FSH (for confirmation of menopausal status), serum total IgE, HBsAg, and hepatitis C antibody.
  • Efficacy procedures Efficacy was assessed during the study at specified clinic visits using patient-reported outcomes (PROs), including Straumann Dysphagia
  • EoE-EREFS Eosinophilic Esophagitis Activity Index
  • HSS EoE Histological Scoring System
  • the SDI is a non-validated PRO that has been used in clinical studies to determine the frequency and intensity of dysphagia (Straumann 2010).
  • the total SDI score ranges from 0 to 9. In the Straumann study, a clinical response (improvement) was defined as a decrease in SDI score of at least 3 points from baseline.
  • the EEsAI is a non-validated, multimodular index in development at University Hospital Glassspital (Berne, Switzerland) (Schoepfer 2014), a part of the international EEsAI study group.
  • the EEsAI PRO module (questionnaire) used in this study includes items related to the intensity and frequency of dysphagia, the influence of specific food groups on dysphagia symptoms, and other symptoms independent of eating or drinking (i.e., heartburn, acid regurgitation, and chest pain).
  • the total EEsAI PRO score ranges from 0 to 100 ( Figure 1), wherein higher scores indicate worse symptoms. The score consists of 5 parts: frequency of trouble swallowing, duration of trouble swallowing, pain when swallowing, visual dysphagia question, and avoidance, modification and slow eating (AMS).
  • AMS modification and slow eating
  • the EoE-QOL-A questionnaire is a validated disease-specific measure of health- related quality of life in EoE patients (Taft 2011).
  • the instrument used in this study, the EoE-QOL-A v.3.0 includes 30 items related to established domains such as social functioning, emotional functioning, and disease impact of daily life experiences.
  • the EoE-QOL-A has a 1-week recall period. The items are graded on a 5-point scale: 'Not at All,' 'Slightly,' 'Moderately,' 'Quite a bit,' and 'Extremely'.
  • Esophageal biopsies were obtained by endoscopy at the screening and week 12 visits. The screening endoscopy was performed during the screening period to allow results to be available prior to day -1 for assessment of eligibility. A total of 9 mucosal pinch biopsies were collected at each time point from 3 esophageal regions: 3 proximal, 3 mid, and 3 distal. Two samples from each region were used for the histology (needed for study inclusion criteria, as well as secondary endpoint). To participate in the study, patients had to have a peak intraepithelial eosinophil count >15 eos/hpf (400X) in at least 2 of the 3 esophageal regions sampled.
  • Change in peak esophageal eos/hpf (400X) from baseline to week 12 was a secondary endpoint; this was determined by counting eosinophils in the most inflamed areas of each esophageal region sampled at each time point and calculating the change in the peak count at each site obtained at baseline compared to the count obtained at week 12.
  • the change in the mean of all 3 peak counts i.e., the peak count at each of the 3 esophageal regions for each patient at each time point (screening and week 12) was calculated. Tissue blocks remaining after the histological assessment were banked for exploratory research.
  • EoE-EREFS edema, rings, exudates, furrows, strictures
  • This instrument includes a total of 17 items related to the presence and severity of esophageal features.
  • the specific esophageal features include: rings (concentric rings around esophagus - absent, mild, moderate, severe, not applicable); strictures (narrowing of the esophagus - yes, no, not applicable); diameter of the stricture (if applicable); exudates (refer to white plaques - absent, mild, severe), furrows (vertical lines down the esophagus - absent, present); edema (loss of vascular markings of the mucosa - absent, present); crepe paper esophagus (absent, present); overall general appearance incorporating all endoscopically identified EoE findings (i.e., fixed rings, strictures, whitish exudates, furrowing, edema, and crepe paper mucosa).
  • EoE esophageal characteristics were analyzed based on the EoE-EREFS, a validated scoring system for inflammatory and remodeling features of disease using both overall scores and scores for each individual characteristic (Hirano 2014).
  • the modified EREFS score in this study was based on a total score range of 0 - 8; wherein higher scores indicate greater impairment.
  • Each score comprised: Edema: 0 - 1 ; Rings: 0 - 3; Exudates: 0 - 2; Furrows: 0 - 1 ; and Strictures: 0 - 1 to give a total possible score of 8.
  • EndoFLIP endolumenal functional lumen imaging probe
  • the EndoFLIP device is a catheter based procedure that measures the cross sectional area at multiple sites along the esophagus with simultaneous intraluminal pressure recordings during volumetric distension of the esophagus.
  • the analyses of cross sectional area versus pressure relationships of the esophagus allow for determination of esophageal compliance as well as the distensibility plateau (DP).
  • the DP has been shown to be significantly reduced in patients with EoE compared to healthy controls (Kwiatek 2011). Moreover, the esophageal distensibility has been associated with outcomes of both food impaction and need for esophageal dilation (Nicodeme 2013).
  • the EoE-HSS generated separate severity (grade) and extent (stage) disease scores.
  • the score was used to measure 8 histologic features (parameters) of EoE from 3 different regions (proximal, mid and distal) of the esophagus (Collins, et al. 2017).
  • the 8 parameters include: eosinophil density, basal zone hyperplasia, eosinophil abscesses, eosinophil surface layering, dilated intercellular spaces, surface epithelial alteration, dyskeratotic cells, and lamina limba fibrosis.
  • a scale of 0 - 3 was used for each parameter, both grade and stage (0 being least inflamed, normal).
  • Total score range for each region for this study was 0 - 21 (excluding lamina propria parameter). The lamina basement assessment was excluded given that 50% of pinch biopsies would not be deep enough for lamina propria assessment.
  • An AE is any untoward medical occurrence in a patient administered a study drug, which may or may not have a causal relationship with the study drug. Therefore, an AE is any unfavorable and unintended sign (including abnormal laboratory finding), symptom, or disease that is temporally associated with the use of a study drug, whether or not considered related to the study drug. An AE also includes any worsening (i.e., any clinically significant change in frequency and/or intensity) of a preexisting condition that is temporally associated with the use of the study drug.
  • a serious adverse event is any untoward medical occurrence that at any dose results in death, is life-threatening, requires in-patient hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly /birth defect and/or is an important medical event (e.g. , such as an event may jeopardize the patient or may require intervention to prevent 1 of the other serious outcomes listed above.
  • Baseline Characteristics Patients were randomized in a 1 : 1 ratio to receive a subcutaneous (SC) 600 mg dupilumab or SC placebo loading dose followed by weekly SC 300 mg dupilumab or SC placebo during the 12-week double-blind treatment phase. The randomization was stratified by baseline Straumann Dysphagia Instrument (SDI) PRO score (>5 and ⁇ 7 versus >7). Baseline demographic and disease characteristics were generally balanced between the two groups (Tables 1 - 2), except for mean total IgE (dupilumab 217.8 kU/L; placebo 468.2 kU/L).
  • SDI Straumann Dysphagia Instrument
  • n # of patients with both baseline and post-baseline actual observed values * For continuous variables, multiple imputation/ ANCOVA was used, LS (SE) was presented; for binary variables, patients with missing data were treated as non-responders and Fisher exact test was used for comparison, number and % of responder was presented.
  • Table 6 summarizes the effect of dupilumab on subjective patient-reported outcomes on dysphagia and on clinician-assessed objective measures.
  • Distal LS mean % change from 2.7 (5.3) -57.0 (5.3) -59.7 (-74.3,
  • Proximal LS mean % change 66.1 -50.2 (24.5) -116.3 (-188.0, from BL ( ⁇ SE) (27.4) —44.5)***
  • EoE Histology scoring system measures eosinophil density, basal zone hyperplasia, eosinophil abscesses, eosinophil surface layering, surface epithelial alteration, dyskeratotic epithelial cells and dilated intercellular spaces. Lamina basement was excluded from the analysis, as -50% of pinch biopsies were not deep enough for assessment.
  • Esophageal distensibility plateau is measured using the Functional Lumen Imaging Probe (EndoFLIP,® Crospon), a probe using impedance planimetry.
  • the continuous efficacy endpoints were analyzed in the full analysis set (FAS) using multiple imputation (MI), followed by an analysis of covariance (ANCOVA) model with treatment group as fixed effect, and baseline SDI and the relevant baseline value as covariates.
  • LS least-squares; qw, every week
  • dupilumab demonstrated a statistically significant reduction in the primary endpoint, change from baseline in Straumann Dysphagia
  • Eosinophilic Esophagitis Edema, Rings, Exudates, Furrows and Stricture (EoE-EREFS) score at week 12.
  • Dupilumab treatment was generally safe and well tolerated.
  • the most common TEAEs were mild ISRs and viral upper respiratory tract infections and nasopharyngitis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des méthodes de traitement, de prévention ou de réduction de la gravité d'une œsophagite à éosinophiles active. Dans certains modes de réalisation, la présente invention concerne des procédés d'augmentation de la distension oesophagienne. Les méthodes de la présente invention comprennent l'administration à un sujet en ayant besoin d'une composition pharmaceutique comprenant un inhibiteur de la voie de l'interleukine 4-l'interleukine 13 (IL-4/IL-13) tel qu'un anticorps anti-IL-4R.
EP18750578.9A 2017-08-04 2018-08-03 Méthodes de traitement de l'oesophagite à éosinophiles active Pending EP3661551A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762541242P 2017-08-04 2017-08-04
US201762561593P 2017-09-21 2017-09-21
EP18305252 2018-03-08
PCT/US2018/045195 WO2019028367A1 (fr) 2017-08-04 2018-08-03 Méthodes de traitement de l'oesophagite à éosinophiles active

Publications (1)

Publication Number Publication Date
EP3661551A1 true EP3661551A1 (fr) 2020-06-10

Family

ID=63113646

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18750578.9A Pending EP3661551A1 (fr) 2017-08-04 2018-08-03 Méthodes de traitement de l'oesophagite à éosinophiles active

Country Status (9)

Country Link
EP (1) EP3661551A1 (fr)
JP (2) JP7417515B2 (fr)
KR (1) KR20200035442A (fr)
CN (1) CN111032084A (fr)
AU (1) AU2018311981A1 (fr)
CA (1) CA3071528A1 (fr)
IL (1) IL272245A (fr)
MA (1) MA49744A (fr)
MX (1) MX2020001305A (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117085122A (zh) * 2022-04-29 2023-11-21 中山康方生物医药有限公司 抗人il-4ra的抗体及其用途

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI682781B (zh) * 2013-07-11 2020-01-21 美商再生元醫藥公司 藉由投與il-4r抑制劑治療嗜酸性食道炎的方法
CA2924873A1 (fr) * 2013-10-23 2015-04-30 Genentech, Inc. Methodes de diagnostic et de traitement de troubles eosinophiliques
EP3268032A1 (fr) * 2015-03-11 2018-01-17 GlaxoSmithKline Intellectual Property Development Limited Protéines de liaison à la tslp

Also Published As

Publication number Publication date
MX2020001305A (es) 2020-03-09
RU2020109331A3 (fr) 2021-12-28
KR20200035442A (ko) 2020-04-03
JP2024016237A (ja) 2024-02-06
RU2020109331A (ru) 2021-09-06
AU2018311981A1 (en) 2020-02-20
CN111032084A (zh) 2020-04-17
JP7417515B2 (ja) 2024-01-18
JP2020529434A (ja) 2020-10-08
MA49744A (fr) 2020-06-10
CA3071528A1 (fr) 2019-02-07
IL272245A (en) 2020-03-31

Similar Documents

Publication Publication Date Title
US11053309B2 (en) Methods for treating active eosinophilic esophagitis
US11421036B2 (en) Methods of treating eosinophilic esophagitis by administering an antibody which inhibits interleukin-4 receptor (IL-4R)
US20210363264A1 (en) Methods for treating eosinophilic esophagitis by administering an il-4r inhibitor
JP7434456B2 (ja) 抗il-13抗体を使用する、il-13活性が有害である疾患の治療方法
JP2024016237A (ja) 活動性好酸球性食道炎を治療する方法
RU2776651C2 (ru) Способы лечения активного эозинофильного эзофагита

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200116

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
RAV Requested validation state of the european patent: fee paid

Extension state: MA

Effective date: 20200116

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40031029

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20230216