US20140099306A1 - Ctla4 fusion proteins for the treatment of diabetes - Google Patents

Ctla4 fusion proteins for the treatment of diabetes Download PDF

Info

Publication number
US20140099306A1
US20140099306A1 US14/021,629 US201314021629A US2014099306A1 US 20140099306 A1 US20140099306 A1 US 20140099306A1 US 201314021629 A US201314021629 A US 201314021629A US 2014099306 A1 US2014099306 A1 US 2014099306A1
Authority
US
United States
Prior art keywords
composition
oil
subject
administered
peptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/021,629
Other languages
English (en)
Inventor
Tihamer Orban
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.)
Phaim Pharma Ltd
Original Assignee
Orban Biotech LLC
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 Orban Biotech LLC filed Critical Orban Biotech LLC
Priority to US14/021,629 priority Critical patent/US20140099306A1/en
Assigned to ORBAN BIOTECH LLC reassignment ORBAN BIOTECH LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ORBAN, TIHAMER
Publication of US20140099306A1 publication Critical patent/US20140099306A1/en
Priority to US14/576,990 priority patent/US10233242B2/en
Assigned to DMNOMORE reassignment DMNOMORE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ORBAN BIOTECH LLC
Priority to US16/356,445 priority patent/US11286303B2/en
Assigned to PHAIM PHARMA LTD reassignment PHAIM PHARMA LTD CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: DMNOMORE LIMITED
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • the present invention relates generally to the field of autoimmune disease and specifically to the treatment, prevention, or delayed progression of Type 1 diabetes mellitus.
  • Type 1 diabetes mellitus is an immune-mediated disease where insulin-secreting ⁇ -cells are destroyed by an autoimmune response.
  • T1DM Type 1 diabetes mellitus
  • Type 1 diabetes mellitus is immune-mediated, in which insulin-producing ⁇ cells are destroyed. Yet, at the time of diagnosis, most patients still have appreciable amounts of insulin production. Preservation of residual ⁇ -cell function is highly desirable because it can reduce short-term and long-term complications of the disease.
  • a method of treating diabetes mellitus in a subject comprising administering an effective amount of a fusion protein composition comprising a T-cell co-stimulation antagonist and a portion of an immunoglobulin molecule.
  • the T-cell co-stimulation antagonist comprises the extracellular domain of CTLA4, an effective fragment of the extracellular domain or immunologically active variant of the extracellular domain.
  • the T-cell co-stimulation antagonist may bind a B7 antigen expressed on B cells or other antigen presenting cells (APCs).
  • the B7 antigen is expressed on B cells and on APCs.
  • the fusion protein is Abatacept.
  • the composition further comprises an oil-based carrier such as a water-in-oil emulsion (e.g., IFA or Montamide ISA).
  • the composition may be administered by intravenous infusion, such as in about 50 to 200 ml of physiological saline or at a dose ranging from about 5 mg/kg to about 50 mg/kg or at a dose ranging from about 250 to 2000 mg, or at a dose of 500 mg, 750 mg, or 1000 mg.
  • the methods as described herein may also comprise determining levels of C-peptide in blood samples taken from the subject over time as an indicator of effectiveness of the treatment in inhibiting activation of auto-aggressive T-cells.
  • the effectiveness of the composition in inhibiting activation of auto-aggressive T-cells is indicated by maintenance of C-peptide production or a delay in reduction of C-peptide production as compared to a standard or by improved HbA1c or reduction in the use of insulin by said subject as compared to a standard.
  • Such measurements can be done ex vivo, such as by analyzing a blood sample.
  • the reduction of C-peptide production in said subject may be delayed for at least 3, 6, 9, 12, or 18 months, or 2, 3, 4, or more years.
  • a preferred patient population is treated.
  • a patient selected from a population having a statistically greater response rate, such as a white patient is treated.
  • Some embodiments provided herewith provide a method of preventing the onset of diabetes in a subject at risk for diabetes mellitus comprising administering an effective amount of a fusion protein composition comprising a T-cell co-stimulation antagonist and a portion of an immunoglobulin molecule. Some embodiments provided herewith provide a method delaying the onset of diabetes by at least 3, 6, 9, 12, or 18 months, or 2, 3, 4, or more years in a subject at risk for diabetes mellitus by administering an effective amount of a fusion protein composition comprising a T-cell co-stimulation antagonist and a portion of an immunoglobulin molecule.
  • FIG. 1 is the population mean of stimulated C-peptide 2-h AUC mean over time for each treatment group.
  • the estimates are from the ANCOVA model adjusting for age, sex, baseline value of C-peptide, and treatment assignment.
  • Y-axis is on a log(y+1) scale. Error bars show 95% confidence intervals (CIs).
  • AUC area under the curve.
  • FIG. 3 is the proportion of participants with 2-h peak C-peptide remaining at or above 0.2 nmol/L over time for each treatment group.
  • FIGS. 4A and 4B are the population mean of (A) HbA1c and (B) insulin use over time for each treatment group. Estimates are from the ANCOVA model adjusting for age, sex, baseline value of HbA1c, and treatment assignment. Insulin use is per kg of bodyweight, at 3-month intervals. Error bars show 95% CIs. HbA1c is glycated haemoglobin A1c.
  • CTLA4 molecule can be used for the treatment, prevention, or delayed progression of Type 1 diabetes mellitus (T1DM) in a subject.
  • C-peptide is a protein that is produced in the body along with insulin.
  • preproinsulin is secreted with an A-chain, C-peptide, a B-chain, and a signal sequence.
  • the signal sequence is cut off, leaving proinsulin. Then the C-peptide is cut out, leaving the A-chain and B-chain to form insulin. Since C-peptide and insulin are present in equimolar amounts, it is a highly reliable marker for insulin production and the health of pancreatic ⁇ cells.
  • T cells play a central part in autoimmunity associated with Type 1 diabetes. To become fully activated and autoaggressive, T cells are believed to need at least two crucial signals.
  • the first signal is an interaction between an antigen in the groove of the MHC molecule on antigen-presenting cells and the T-cell receptor (TCR).
  • TCR T-cell receptor
  • the most important second signal is the interaction between CD80 and CD86 on the antigen presenting cells (APCs) and CD28 on the T cells. This costimulatory second signal is needed for full activation of cells, and without it T cells do not become functional. Therefore, co-stimulation blockade has been proposed as a therapeutic modality for autoimmunity and transplantation. (Bluestone J A, St Clair E W, Turka L A. Immunity 2006; 24: 233-38.)
  • CTLA4 Cytotoxic T-lymphocyte-associated antigen 4
  • CD152 Cytotoxic T-lymphocyte-associated antigen 4
  • CTLA4 is a protein involved in the regulation of the immune system.
  • Naturally occurring CTLA4 is described in U.S. Pat. Nos. 5,434,131, 5,844,095, and 5,851,795.
  • Natural CTLA4 proteins are encoded by the CTLA4 gene.
  • CTLA4 is a cell surface protein, having an N-terminal extracellular domain, a transmembrane domain, and a C-terminal cytoplasmic domain. The extracellular domain binds to and/or interferes with target antigens, such as CD80 and CD86, serves as nature natural break of T cell stimulation.
  • the extracellular domain of the CTLA4 molecule begins with methionine at position +1 and ends at aspartic acid at position +124; in other embodiments, the extracellular domain begins with alanine at position ⁇ 1 and ends at aspartic acid at position +124.
  • a CTLA4 molecule is a molecule comprising a cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) extracellular domain.
  • CTLA4 cytotoxic T-lymphocyte-associated antigen 4
  • the extracellular domain of CTLA4 comprises a portion of the CTLA4 protein that recognizes and binds to at least one B7 (CD80/86) antigens such as a B7 antigen expressed on B cells and APCs.
  • the extracellular domain may also include fragments or derivatives of CTLA4 that bind a B7 antigen.
  • the CTLA4 extracellular domain can also recognize and bind CD80 (B7-1) and/or CD86 (B7-2).
  • the extracellular domain may also include fragments or derivatives of CTLA4 that bind a binds CD80 and/or CD86.
  • the CTLA4 molecule may be a fusion protein, where a fusion protein is defined as one or more amino acid sequences joined together using methods well known in the art. The joined amino acid sequences thereby form one fusion protein.
  • the CTLA4 molecule contains at least a portion of an immunoglobulin, such as the Fc portion of an immunoglobulin.
  • the CTLA4 molecule is an isolated and purified CTLA4 molecule.
  • the CTLA4 molecule is a protein containing at least a portion of an immunoglobulin, such as the Fc portion of an immunoglobulin. In some embodiments, the CTLA4 molecule is an isolated and purified CTLA4 molecule.
  • the CTLA4 molecule is abatacept.
  • Abatacept is a soluble fusion protein that consists of the extracellular domain of human CTLA-4 linked to the modified Fc (hinge, CH2, and CH3 domains) portion of human immunoglobulin G1 (IgG1).
  • Abatacept is produced by recombinant DNA technology in a mammalian cell expression system. The apparent molecular weight of abatacept is 92 kilodaltons.
  • Abatacept was developed for use in adult rheumatoid arthritis and juvenile idiopathic arthritis and is indicated for reducing signs and symptoms, inducing major clinical response, inhibiting the progression of structural damage, and improving physical function in adult patients with moderately to severely active rheumatoid arthritis.
  • Abatacept was developed by Bristol-Myers Squibb and is disclosed, for example, in U.S. Pat. 5,851,795, U.S. Pat. No. 7,455,835, and U.S. Pat. Pub. 20011/311529.
  • Abatacept under the trade name ORENCIA, may be used as monotherapy or concomitantly with disease-modifying antirheumatic drugs (DMARDs) other than tumor necrosis factor (TNF) antagonists.
  • DMARDs disease-modifying antirheumatic drugs
  • TNF tumor necrosis factor
  • Abatacept is also indicated for reducing signs and symptoms in pediatric patients 6 years of age and older with moderately to severely active polyarticular juvenile idiopathic arthritis.
  • Abatacept may be used as monotherapy or concomitantly with methotrexate (MTX). Since abatacept is a selective costimulation modulator and inhibits the costimulation of T cells, it should not be administered concomitantly with TNF antagonists.
  • Abatacept selectively binds to CD80 and CD86, thereby blocking the interaction with CD28 and interfering with T-cell activation. It inhibits naive T-cell activation, thus having the potential to selectively inhibit T-cell response to specific antigens instead of broad immunosuppression. Effector-memory T-cell responses are less dependent on CD28 co-stimulation and, presumably, are less inhibited by co-stimulation blockade.
  • Co-stimulation blockade with abatacept has been shown to have clinical effectiveness in psoriasis (Abrams J R, Lebwohl M G, Guzzo C A, et al. J Clin Invest 1999; 103: 1243-52) and psoriatic arthritis (Mease P, Genovese M C, Gladstein G, et al. Arthritis Rheum 2011; 63: 939-48) and is approved for treatment of rheumatoid arthritis, Genant H K, Peterfy C G, Westhovens R, et al.
  • T-cell co-stimulatory antagonists such as CTLA-4 compositions and in particular abatacept, halts or slows autoimmune ⁇ -cell destruction leading to preservation of C-peptide secretion in recently diagnosed patients with Type 1 diabetes by blocking the generation of autoaggressive T cells (Orban et al., Lancet 2011; 378 (9789): 412-9.)
  • a method of treating, preventing, or delaying the progression of diabetes mellitus by administering a CTLA4 molecule can prevent or delay the onset of diabetes mellitus, or prevent or delay loss of residual ⁇ -cell mass, providing a longer remission period and delaying the onset of diabetes-related complications at a later stage of the life.
  • T1DM may be treated by the methods as described herein.
  • the treatment can be for subjects with residual beta-cell function as well as for those no longer having any beta-cell function.
  • the treatment may also be suggested for subjects provided exogenous beta-cells through transplant or injection or other beta cell replacement modalities (like embryonic or other stem cell therapies or other replacement modalities).
  • T1DM may be prevented in a subject by first selecting a subject who is susceptible to developing diabetes and administering a CTLA4 molecule as described herein.
  • the subject who is susceptible to developing diabetes may be selected by the expression of one or more of: GAD65 autoantibodies (GAAs), ICA512 autoantibodies (ICA512AAs), or anti-insulin autoantibodies (IAAs).
  • GAD65 autoantibodies GAAs
  • ICA512AAs ICA512 autoantibodies
  • IAAs anti-insulin autoantibodies
  • T1DM The onset of T1DM may be delayed by the methods as described herein such that insulin is not needed by the subject for a longer length of time.
  • the present method may extend the “honeymoon phase” in an already diabetic subject.
  • the honeymoon phase is where insulin is secreted by the pancreas, causing high blood sugar levels to subside, and resulting in normal or near normal glucose levels due to responses to insulin injections and treatment.
  • the CTLA4 molecules as described herein may be administered in combination with a pharmaceutically acceptable carrier and administered as a pharmaceutical composition.
  • pharmaceutically acceptable carrier includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's Pharmaceutical-Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof.
  • any conventional carrier medium is incompatible with the compounds of provided herein, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention.
  • materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatine; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil, sesame oil; olive oil; corn oil and soybean oil; glycols; such as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium
  • Suitable routes of administration include, but are not limited to, inhalation, transdermal, oral, rectal, transmucosal, intestinal and parenteral administration, including intramuscular, subcutaneous and intravenous injections.
  • compositions as described herein may be administered with an adjuvant.
  • adjuvant can be a compound that lacks significant activity administered alone but can potentiate the activity of another therapeutic agent.
  • an adjuvant is selected from the group consisting of buffers, anti-microbial preserving agents, surfactants, antioxidants, tonic regulators, antiseptics, thickeners and viscosity improvers.
  • the adjuvant is IFA or other oil-based adjuvant is present between 30-70%, preferably between 40-60%, more preferably between 45-55% proportion weight by weight (w/w).
  • CTLA4and IFA or other oil based adjuvant are present in about a 50/50 weight by weight ratio.
  • the pharmaceutical composition is free of contaminants, e.g., pyrogens.
  • the compound can be formulated readily by combining the CTLA4 molecule with one or more pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired. to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethyl-cellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the compound may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Injection is a preferred method of administration for the compositions of the current invention.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the composition may be a water-in-oil emulsion.
  • the composition may be an oil-in-water emulsion.
  • oil-in-water emulsions may be particularly useful for controlling the release profile and providing a slow release of the active drug, which can potentially be absorbed unaltered from such an emulsion.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • composition comprising a CTLA4 molecule also includes an oil-based carrier.
  • the oil-based carrier is a composition that includes at least 10% by weight of a natural or synthetic oil suitable for administration to a human in conjunction with a therapeutic agent.
  • the carrier includes at least 20, 30, 50, 70, 80, 90, 95, 98, or 99% oil by weight.
  • the oil-based carrier can include less than 70, 60, 50, 40, 30 or 20% oil by weight.
  • the oil will be in the range of 10 to 95%, preferably 20 to 90% or 30 to 70% oil by weight.
  • the oil should be chosen such that it provides for sustained release of a substance dispersed within it when administered to a subject.
  • Suitable oils include mineral oil (e.g., Drakeol 6 VR light mineral oil), vegetable oil, squalene, or liquid paraffin.
  • the oil-based carrier can contain more than one type of oil.
  • the oil-based carrier can include an immune stimulator, e.g., an immunostimulating glucan, but it is much preferred that the oil-based carrier does not include an immune stimulator, e.g., an immunostimulating glucan, a bacterial component, e.g., a mycobacterial component.
  • the oil-based carrier does not include an alum component.
  • an oil based carrier is believed to work by triggering the immunocompetent cells, which are related to the inflammatory as well as protective ability.
  • An oil-based carrier can also act as an antigen vehicle and a slow release or long-term antigen presentation device.
  • an oil-based carrier and antigen composition When injected into a subject, an oil-based carrier and antigen composition can form a depot of antigen at the injection site, thereby protecting the antigen from degradation. From this depot the antigen can be released slowly into the system and provides a prolonged antigen presentation as well as expanded total contact surface area and the attraction of inflammatory cells. Macrophages can digest most of the incorporated materials and present the processed antigens on their surface. From this depot the antigen can be released slowly into the system and provides a prolonged antigen supply to act as costimulatory modulator.
  • Oil based carriers optionally include an emulsifier or surfactant component.
  • the emulsifier or surfactant (and the amount of emulsifier or surfactant) is chosen such that it facilitates the mixture or dispersion of a substance, e.g., an antigen preparation, with the oil.
  • An oil-based carrier can include 0.1 to 50%, preferably 1 to 30%, more preferably 5 to 20% by weight of a surfactant or emulsifier.
  • emulsifiers or surfactants include Arlacel A, mannide oleate (e.g., Montamide 80-mannide monooleate), anhydrous mannitol/oleic acid ester, polyoxyethylene or polyoxypropylene.
  • An oil-based carrier or adjuvant typically consists of two components: (1) an oil, and (2) an emulsifier or surfactant, mixed with water.
  • Suitable oils and emulsifiers are known in the art.
  • the oil can be mineral oil, vegetable oil, squalene or liquid paraffin.
  • the emulsifier or surfactant can be, e.g., Arlacel A, mannide oleate, anhydrous mannitol/oleic acid ester, polyoxyethylene or polyoxypropylene.
  • Exemplary oil-based adjuvants include conventional IFA, Montamide ISA adjuvants, or Hunter's TiterMax adjuvant.
  • the adjuvant includes 20 to 95%, preferably 30 to 90%, more preferably 40 to 70% by weight of an oil phase, and 0.1 to 50%, preferably 1 to 30%, more preferably 5 to 20% by weight of a surfactant or emulsifier.
  • oil-based adjuvants are described, e.g., in U.S. Pat. No. 5,814,321, U.S. Pat. No. 6,299,884, U.S. Pat. No. 6,235,282, and U.S. Pat. No. 5,976,538.
  • IFA is typically a mixture of a non-metabolizable oil (e.g., mineral oil), water, and a surfactant (e.g., Arlacel A)
  • a surfactant e.g., Arlacel A
  • IFA does not contain a bacterial component, e.g., mycobacteria.
  • CFA Complete Freund's Adjuvant
  • Montanide ISA Adjuvants are a group of oil/surfactant based adjuvants in which different surfactants are combined with either a non-metabolizable mineral oil, a metabolizable oil, or a mixture of the two. They are prepared for use as an emulsion with aqueous Ag solution.
  • the surfactants of the Montanide group undergo strict quality control to guard against contamination by any substances that could cause excessive inflammation, as has been found for some lots of Arlacel A used in Freund's adjuvant.
  • the various Montanide ISA group of adjuvants are used as water-in-oil emulsions, oil-in-water emulsions, or water-in-oil-in-water emulsions.
  • the different adjuvants accommodate different aqueous phase/oil phase ratios, because of the variety of surfactant and oil combinations.
  • Hunter's TiterMax (CytRx Corp., Norcross, Ga.) is an oil/surfactant-based adjuvant prepared as a water-in-oil emulsion in a manner similar to that used for conventional Freund's adjuvants. However, it uses a metabolizable oil (squalene) and a nonionic surfactant that has good protein antigen-binding capacity as well as adjuvant activity.
  • the adjuvant activity may relate, in part, to the surfactant's ability to activate complement and bind complement components, as this helps target the Ag to follicular dendritic cells in the spleen and lymph nodes.
  • the surfactant used in the commercially available adjuvant is one of a number of synthetic nonionic block copolymers of polyoxyethylene and polyoxypropylene developed by Hunter (Hunter et al., 1991 Vaccine 9:250-256).
  • the utilization of copolymer-coated microparticles to stabilize the emulsion permits formation of stable emulsions with less than 20% oil, an important factor in minimizing total adjuvant injected.
  • An adjuvant can be used with antigens to elicit cell-mediated immunity and the production of antibodies of protective isotypes (IgG2a in mice and IgG1 in primates).
  • IgG2a in mice
  • IgG1 in primates
  • Different types of adjuvants share similar side effects, such as a reaction at the injection site and pyrogenicity.
  • Alum a commonly used adjuvant for human vaccine also produces an appreciable granulomatous response at the injection site (Allison & Byars (1991) Mol Immunol 28:279-284).
  • the mode of action of an incomplete Freund's adjuvant can involve non-specific as well as specific immune responses. IFA seems to work by triggering the immunocompetent cells, which are related to the inflammatory as well as protective ability.
  • IFA also acts as an antigen vehicle and a slow release or long-term antigen presentation device. Injecting a patient with an IFA and antigen compound, it forms a depot of antigen at the injection site, thereby protecting the antigen from degradation. From this depot the antigen is released slowly into the system and provides a prolonged antigen presentation as well as expanded total contact surface area and the attraction of inflammatory cells. Macrophages digest most of the incorporated materials and present the processed antigens on their surface. From this depot the antigen can be released slowly into the system and provides a prolonged antigen supply to act as costimulatory modulator.
  • the specific enhancing effect of the IFA on the antigen immunogenicity has been found to lead to increased humoral immunity (preferentially protective antibody production; IgG1 in humans and IgG2a in mice) and to elicit specific cell mediated immunity (preferentially Th2 type).
  • humoral immunity preferentially protective antibody production
  • IgG1 in humans
  • IgG2a in mice
  • Th2 type specific cell mediated immunity
  • human recombinant insulin B-chain in IFA results in Th2 cytokine pattern in NOD mice islets (Ramiya et al. (1996) J Autoimmun 9:349-356).
  • IFA is unique among adjuvants tried for diabetes prevention in animal models. Ramiya and coworkers (supra) concluded that both alum and DPT as adjuvants have ‘non-specific’ protective effects unrelated to the antigen used, while IFA is the only one with antigen specific protective effect for diabetes prevention in animals.
  • IFA preferably an IFA approved for human use, e.g., Montanide (e.g., Montanide ISA51, Seppic Inc., France) or an equivalent composition, is a preferred adjuvant for use in the methods and vaccines described herein.
  • Montanide ISA51 has shown no systemic or significant local side effects in our animal and in our human studies.
  • the compounds can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired. to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Injection is a preferred method of administration for the compositions of the current invention.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the amount of the combination of a CTLA4 molecule provided to the subject will depend on both the size and weight of the subject as well as the progression of the disease.
  • the therapeutically effective amount can be initially determined from in vitro assays. Since the compounds of the present invention may have a low absorption and low bioavailability, the therapeutically effective amount may be determined from, for example, blood level of the compounds or metabolites thereof or fecal concentration of the compounds or metabolites thereof. As is well known in the art, therapeutically effective amounts for use in humans can also be determined from animal models. A therapeutically effective dose can also be determined from human data for compounds which are known to exhibit similar pharmacological activities. The applied dose can be adjusted based on the relative potency of the administered compound as compared with the known compound.
  • Patient doses for parenteral administration of the compounds described herein typically range from about 1 mg/day to about 10,000 mg/day, more typically from about 10 mg/day to about 1,000 mg/day, and most typically from about 50 mg/day to about 500 mg/day. Stated in terms of patient body weight, typical dosages range from about 0.01 to about 150 mg/kg/day, more typically from about 0.1 to about 15 mg/kg/day, and most typically from about 1 to about 10 mg/kg/day, for example 5 mg/kg/day or 3 mg/kg/day.
  • the CTLA4 molecule may be administered in a single daily dose or it may be administered multiple times per day. Alternatively, it may be administered less than once a day. The dosing may be over a period of time, such as once a month, or every 28 days. In some embodiments, additional doses (e.g., bolus dosing) may be given at the beginning of treatment. In some embodiments, a dose containing approximately 5, 10, 20, 30, 50, 100 mg/kg of the CTLA4 molecule.
  • administering or “administration” are intended to encompass all means for directly and indirectly delivering a compound to its intended site of action.
  • delaying the progression as used herein in the context of delaying the progression of diabetes mellitus means that the loss of functional residual ⁇ -cell mass, after the clinical onset of Type 1 diabetes is delayed.
  • the delayed progression of T1DM can be measured, for example, by measuring C-peptide production.
  • pharmaceutically acceptable refers to additives or compositions that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to an animal, such as a mammal (e.g., a human).
  • pharmaceutically acceptable carrier includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's, The Science and Practice of Pharmacy, (Gennaro, A.
  • materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatine; talc.
  • Excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil, sesame oil; olive oil; corn oil and soybean oil; glycols; such as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
  • oils such as peanut oil, cottonseed oil; safflower oil, sesame oil; olive oil
  • composition refers to a composition described herein, or pharmaceutically acceptable salts thereof, with other agents such as carriers and/or excipients.
  • a pharmaceutical composition will have the active agent present at least 95% purity, or 98% purity, or 99% purity, or more.
  • the term “subject” is a human or other animal, having a diabetes, pre-diabetes, or a predisposition to diabetes.
  • the subject will be in need of the therapeutic treatment as provided herein.
  • Preferred patients are mammals. Examples of patients include but are not limited to, humans, horses, monkeys, dogs, cats, mice, rates, cows, pigs, goats and sheep.
  • “subjects” are generally human patients having diabetes.
  • “subjects” are human patients who have been diagnosed with T1DM within the last 200, 100, or 50 days.
  • “subjects” are human patients who have been recently diagnosed with diabetes mellitus but still have residual beta-cell function. In some such embodiments the residual beta-cell function is detectable or at least 10%, 20%, 30%, 40%, 50%, 60%, or more of the beta cells in a fully functioning pancreas.
  • terapéuticaally effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired biological or medicinal response in a cell culture, tissue system, animal, or human (e.g, the desired therapeutic result).
  • a therapeutically effective amount of the composition may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the CTLA4 molecule to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the pharmacological agent are outweighed by the therapeutically beneficial effects.
  • the response includes alleviation and/or delay of onset of one or more symptoms of the disease, condition, or disorder being treated.
  • treatment is defined as the application or administration of the therapeutic agents to a subject, or application or administration of the therapeutic agents to an isolated tissue or cell line from a subject who has diabetes, a symptom of disease or a predisposition toward a disease. Treatment is intended to encompass preventing the onset, slowing the progression, reversing or otherwise ameliorating, improve, or affect the disease, the symptoms or of disease or the predisposition toward disease.
  • treatment of a subject e.g., a human subject, with a composition described herein, can slow, improve, or stop the ongoing autoimmunity, e.g., a reaction against pancreatic ⁇ -cells, in a subject before, during, or after the clinical onset of Type 1 diabetes.
  • a subject e.g., a human subject
  • a composition described herein can slow, improve, or stop the ongoing autoimmunity, e.g., a reaction against pancreatic ⁇ -cells, in a subject before, during, or after the clinical onset of Type 1 diabetes.
  • Patients (aged 6-45 years) diagnosed with Type 1 diabetes within the past 100 days were parallel-screened for this study. Patients were eligible to participate in the study if they had at least one diabetes-related autoantibody (microassayed insulin antibodies [if duration of insulin therapy was less than 7 days]; glutamic acid decarboxylase-65 [GAD-65] antibodies; islet-cell antigen-512 [ICA-512] antibodies; or islet-cell autoantibodies) and had stimulated C-peptide concentrations of 0.2 nmol/L or higher measured during a mixed-meal tolerance test (MMTT) done at least 21 days after diagnosis of diabetes and within 37 days of randomization.
  • MMTT mixed-meal tolerance test
  • AUC area under the curve.
  • HbA1c glycated haemoglobin A1c. *Excludes participants with data missing for indicated variable (number missing: race, 1; HbA1c, 2; insulin use, 1; HLA allele status, 4). ⁇ Islet-cell autoantibodies by immunofluorescence not tested on 16 patients (considered negative for count). ⁇ Range 51-108 for abatacept group and 38-107 for placebo.
  • Abatacept (Orencia, Bristol-Myers Squibb, Princeton, N.J., USA) was given on days 1, 14, and 28, and then every 28 days with the last dose on day 700 (total 27 doses) as a 30-min intravenous infusion at a dose of 10 mg/kg (maximum 1000 mg per dose) in a 100 mL 0.9% sodium chloride infusion. Normal saline infusion was used as placebo. Patients did not receive any premedication.
  • a routine chemistry panel was done (Roche Diagnostics [Indianapolis, Ind., USA] Hitachi 917 Analyzer and reagents). HLA class II alleles were measured with PCR amplification and sequence-specific hybridization. ⁇ -cell function was assessed by stimulated C-peptide secretion. The prespecified primary outcome of this trial was a comparison of the area under the curve (AUC) of stimulated C-peptide response over the first 2 h of a 4-h MMTT2, done at the 24-month visit. The 4-h MMTTs were done at baseline and at 24 months; 2-h MMTTs were obtained at 3, 6, 12, and 18 months. Patients who had completed their 2-year visit MMTT were included in the primary outcome assessment.
  • Prespecified secondary outcomes included: slope of C-peptide over time, difference between groups in incidence of loss of peak C-peptide to less than 0.2 nmol/L, differences in HbA1c and insulin dose over time, and safety.
  • Prespecified subgroup factors included age, sex, race, baseline C-peptide, baseline insulin use, baseline HbA1c, and HLA type.
  • Spotfire S+ 8.1 a statistical analysis software, was used for all analyses.
  • a sample size of 108 participants was planned to provide 85% power to detect a 50% increase in geometric mean C-peptide relative to the placebo group using a test at the 0.05 level (one-sided), with 10% loss to follow-up and a 2:1 allocation to treatment versus control (based on an estimated mean of 0.248 and SD of 0.179, on the transformed scale). All analyses were based on the prespecified intention to treat cohort with known measurements. Missing values were assumed to be missing at random. The p values associated with the intention-to-treat treatment comparisons of the primary and secondary endpoints are two-sided, although the design of the trial was based on a one-sided hypothesis test.
  • C-peptide AUC equals the AUC divided by the 2-h interval (i.e., AUC/120).
  • the AUC was computed using the trapezoidal rule from the timed measurements of C-peptide during the MMTT.
  • the time to first stimulated peak C-peptide of less than 0.2 nmol/L was analyzed with standard survival methods (Cox model and Kaplan-Meier method).
  • Adverse event grades were analyzed with the Wilcoxon rank sum test. (Agresti A. Categorical data analysis.
  • FIG. 5 shows the results of a homogeneity test of treatment effect on age, sex, race, baseline C-peptide, baseline insulin use, baseline HbA1c, and HLA type.
  • the apparent adverse effect of abatacept in non-white participants might be hypothesis-generating, however the groups size was small.
  • results show that over 2 years co-stimulation modulation with abatacept slows the reduction in ⁇ -cell function in recent-onset Type 1 diabetes by 9.6 months.
  • the early beneficial effect suggests that T-cell activation still occurs around the time of clinical diagnosis of Type 1 diabetes, even though the disease course has presumably been in progress for several years.
  • the fall in ⁇ -cell function in the abatacept group parallels that in the placebo group on the basis of the mixed-model results that included the time interval from 6 to 24 months.
  • This subsequent reduction in ⁇ -cell function causes us to speculate that continuing T-cell activation subsides as the clinical course of the disease progresses. Nevertheless, the difference from the placebo group is maintained during drug administration. Further observation will establish whether the beneficial effect continues after cessation of monthly abatacept infusions. Follow up of these patient shows that the drug beneficial effect lasts beyond the drug administration for at least one year.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Epidemiology (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Dermatology (AREA)
  • Dispersion Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
US14/021,629 2012-06-27 2013-09-09 Ctla4 fusion proteins for the treatment of diabetes Abandoned US20140099306A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/021,629 US20140099306A1 (en) 2012-06-27 2013-09-09 Ctla4 fusion proteins for the treatment of diabetes
US14/576,990 US10233242B2 (en) 2012-06-27 2014-12-19 CTLA4 fusion proteins for the treatment of diabetes
US16/356,445 US11286303B2 (en) 2012-06-27 2019-03-18 CTLA4 fusion proteins for the treatment of diabetes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201213534560A 2012-06-27 2012-06-27
US14/021,629 US20140099306A1 (en) 2012-06-27 2013-09-09 Ctla4 fusion proteins for the treatment of diabetes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US201213534560A Continuation 2012-06-27 2012-06-27

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/576,990 Continuation US10233242B2 (en) 2012-06-27 2014-12-19 CTLA4 fusion proteins for the treatment of diabetes

Publications (1)

Publication Number Publication Date
US20140099306A1 true US20140099306A1 (en) 2014-04-10

Family

ID=49783866

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/021,629 Abandoned US20140099306A1 (en) 2012-06-27 2013-09-09 Ctla4 fusion proteins for the treatment of diabetes
US14/576,990 Active 2033-03-28 US10233242B2 (en) 2012-06-27 2014-12-19 CTLA4 fusion proteins for the treatment of diabetes
US16/356,445 Active 2033-04-04 US11286303B2 (en) 2012-06-27 2019-03-18 CTLA4 fusion proteins for the treatment of diabetes

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/576,990 Active 2033-03-28 US10233242B2 (en) 2012-06-27 2014-12-19 CTLA4 fusion proteins for the treatment of diabetes
US16/356,445 Active 2033-04-04 US11286303B2 (en) 2012-06-27 2019-03-18 CTLA4 fusion proteins for the treatment of diabetes

Country Status (11)

Country Link
US (3) US20140099306A1 (es)
EP (2) EP2866823A4 (es)
JP (1) JP2015522026A (es)
KR (1) KR20150033703A (es)
CN (2) CN113138279A (es)
AU (2) AU2013284460A1 (es)
CA (1) CA2877986A1 (es)
IN (1) IN2015DN00214A (es)
MX (1) MX2015000237A (es)
WO (1) WO2014004857A1 (es)
ZA (1) ZA201500228B (es)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10233242B2 (en) 2012-06-27 2019-03-19 Dmnomore CTLA4 fusion proteins for the treatment of diabetes
US11040093B2 (en) 2014-02-25 2021-06-22 Phaim Pharma Ltd Immunomodulatory therapy for type 1 diabetes mellitus autoimmunity

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8735359B2 (en) * 2012-05-24 2014-05-27 Orban Biotech Llc Combinations of modalities for the treatment of diabetes
US11319359B2 (en) 2015-04-17 2022-05-03 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities
CA3077509A1 (en) 2017-10-10 2019-04-18 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
JP7282760B2 (ja) 2017-10-18 2023-05-29 アルパイン イミューン サイエンシズ インコーポレイテッド バリアントicosリガンド免疫調節タンパク質ならびに関連する組成物および方法
AU2018368790A1 (en) 2017-11-20 2020-06-25 Ichan School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use
JP2021510153A (ja) 2018-01-05 2021-04-15 アイカーン スクール オブ メディシン アット マウント サイナイ 膵臓ベータ細胞の増殖を増加させる方法、治療方法、および組成物
WO2019183245A1 (en) 2018-03-20 2019-09-26 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use
US20220125880A1 (en) 2019-02-04 2022-04-28 Assistance Publique - Hôpitaux De Paris Method for treating checkpoint inhibitors induced adverse events
JOP20210298A1 (ar) * 2019-05-14 2023-01-30 Provention Bio Inc طرق وتركيبات للوقاية من مرض السكري من النوع الأول

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020114814A1 (en) * 1996-02-02 2002-08-22 Gray Gary S. CTLA4-Cgamma4 fusion proteins
US20040136998A1 (en) * 2002-10-30 2004-07-15 Bander Neil H. Methods and compositions for treating or preventing insulin-related disorders using binding agents specific for prostate specific membrane antigen
US7041634B2 (en) * 1995-09-27 2006-05-09 Emory University Method of inhibiting immune system destruction of transplanted viable cells
US7304033B2 (en) * 2001-05-23 2007-12-04 Bristol-Myers Squibb Company Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU212924B (en) 1989-05-25 1996-12-30 Chiron Corp Adjuvant formulation comprising a submicron oil droplet emulsion
US5851795A (en) 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
JP3722375B2 (ja) 1991-06-27 2005-11-30 ブリストル−マイヤーズ スクイブ カンパニー Ctla4レセプター、それを含有する融合タンパク質およびそれらの使用
US5844095A (en) 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
CA2085827C (en) 1991-12-23 2003-10-14 Lucas A. T. Hilgers Adjuvant composition containing synthetic hydrophobic lipopolysaccharide
FR2702373B1 (fr) 1993-03-08 1996-06-07 Rhone Merieux Emulsions vaccinales fluides eau-dans-l'huile contenant une huile métabolisable.
US6719972B1 (en) 1994-06-03 2004-04-13 Repligen Corporation Methods of inhibiting T cell proliferation or IL-2 accumulation with CTLA4- specific antibodies
US6110746A (en) 1995-06-30 2000-08-29 Yeda Research And Development Co. Ltd. Peptides derived from human heat shock protein 60 for treatment of diabetes, compositions, methods and kits
US20030190323A1 (en) 1995-07-05 2003-10-09 Yeda Research And Development Co. Ltd. Preparations and methods for the treatment of T cell mediated diseases
ES2277339T3 (es) 1995-11-30 2007-07-01 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Vacuna con adyuvante de aceite y metodo para su preparacion.
CA2326838C (en) 1998-04-03 2008-12-23 Osiris Therapeutics, Inc. Mesenchymal stem cells as immunosuppressants
ATE271066T1 (de) 2000-05-26 2004-07-15 Bristol Myers Squibb Co Lösliche mutante ctla4 moleküle und deren verwendung
AU2001273174B2 (en) 2000-07-03 2006-05-18 Bristol-Myers Squibb Company Methods for treating rheumatic diseases using a soluble CTLA4 molecule
US20040022787A1 (en) 2000-07-03 2004-02-05 Robert Cohen Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
WO2002053106A2 (en) * 2001-01-05 2002-07-11 Joslin Diabetes Center, Inc. Autoantigen composition
EP1440084B1 (en) 2001-10-24 2010-01-06 University College Cardiff Consultants Ltd. Sperm factor sequences
CN101277722A (zh) 2005-08-06 2008-10-01 王庆华 用于预防和治疗ⅰ型糖尿病的组合物及方法
US20090246212A1 (en) 2005-09-30 2009-10-01 Kyoto University Development of method for screening for drug capable of improving production of regulatory t cells and method for producing regulatory t cells using immunosuppressive macrolide antibiotic
WO2007127787A2 (en) 2006-04-25 2007-11-08 Joslin Diabetes Center, Inc. Insulin autoantigen-specific regulatory cd4+ t cells
US7528111B2 (en) * 2006-05-12 2009-05-05 Bristol-Myers Squibb Company Method of vaccinating subjects receiving immune modulating therapy
NZ599755A (en) 2007-08-30 2013-08-30 Curedm Group Holdings Llc Compositions and methods of using proislet peptides and analogs thereof
US9176122B2 (en) 2008-03-24 2015-11-03 University Of South Florida Biomarkers for predicting response to immunosuppressive therapy
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US20130209463A1 (en) 2010-06-30 2013-08-15 Compugen Ltd. Polypeptides and uses thereof as a drug for treatment of multiple sclerosis, rheumatoid arthritis and other autoimmune disorders
WO2012015903A1 (en) 2010-07-27 2012-02-02 Bayhill Therapeutics, Inc. Treatment of type i diabetes mellitus (t1dm) in young newly diagnosed patients
EP2704738A1 (en) * 2011-05-06 2014-03-12 Diamedica Inc. Compositions and methods for treating diabetes
US20130316375A1 (en) 2012-05-24 2013-11-28 Orban Biotech Llc Diabetes biomarkers
US8735359B2 (en) * 2012-05-24 2014-05-27 Orban Biotech Llc Combinations of modalities for the treatment of diabetes
KR20150033703A (ko) 2012-06-27 2015-04-01 오르반 바이오테크 엘엘씨 당뇨병 치료를 위한 ctla4 융합 단백질
GB2523399B (en) 2014-02-25 2019-03-13 Orban Tihamer A composition comprising ten overlapping peptide fragments of the entire preproinsulin sequence

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7041634B2 (en) * 1995-09-27 2006-05-09 Emory University Method of inhibiting immune system destruction of transplanted viable cells
US20020114814A1 (en) * 1996-02-02 2002-08-22 Gray Gary S. CTLA4-Cgamma4 fusion proteins
US7304033B2 (en) * 2001-05-23 2007-12-04 Bristol-Myers Squibb Company Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules
US20040136998A1 (en) * 2002-10-30 2004-07-15 Bander Neil H. Methods and compositions for treating or preventing insulin-related disorders using binding agents specific for prostate specific membrane antigen

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
Aly et al., Am. J. Therapeutics (2005) 12: 481-490. *
Blazar et al., J. Immunol., 1996, 157:3250-3259. *
Chittasupho et al., Ther. Deliv. 2011, 2: 873-889. *
Driessens et al., Immunol. Rev. 2009, 229: 126-144. *
Fukai et al., Graefe's Arch. Clin. Exp. Ophthalmol., 1999, 237:928-933. *
Mayo Foundation for Medical Education and Research, 2012, accessed at mayoclinic.com/health/type-1-diabetes/DS00329 on 09/13/2012. *
NCT00505375, ClinicalTrials.gov, updated 01/31/2011. *
Sanderson et al., J. Clin. Oncol. (2005); 23: 741-750. *
Sturmhoefel et al., Cancer research, 1999, 59:4964-4972. *
Swiniarski et al., Clinical Immunology, 1999, 92:235-245. *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10233242B2 (en) 2012-06-27 2019-03-19 Dmnomore CTLA4 fusion proteins for the treatment of diabetes
US11286303B2 (en) 2012-06-27 2022-03-29 Phaim Pharma Ltd CTLA4 fusion proteins for the treatment of diabetes
US11040093B2 (en) 2014-02-25 2021-06-22 Phaim Pharma Ltd Immunomodulatory therapy for type 1 diabetes mellitus autoimmunity

Also Published As

Publication number Publication date
CA2877986A1 (en) 2014-01-03
IN2015DN00214A (es) 2015-06-12
CN104519900A (zh) 2015-04-15
EP2866823A4 (en) 2016-03-02
EP2866823A1 (en) 2015-05-06
MX2015000237A (es) 2015-08-14
EP3811966A1 (en) 2021-04-28
CN113138279A (zh) 2021-07-20
US20190211105A1 (en) 2019-07-11
US20150104451A1 (en) 2015-04-16
ZA201500228B (en) 2016-01-27
WO2014004857A1 (en) 2014-01-03
US10233242B2 (en) 2019-03-19
AU2018201965A1 (en) 2018-04-12
AU2013284460A1 (en) 2015-01-29
AU2018201965B2 (en) 2019-11-21
JP2015522026A (ja) 2015-08-03
US11286303B2 (en) 2022-03-29
KR20150033703A (ko) 2015-04-01

Similar Documents

Publication Publication Date Title
US11286303B2 (en) CTLA4 fusion proteins for the treatment of diabetes
US20240058442A1 (en) Combinations of Modalities for the Treatment of Diabetes
Bach et al. Tolerance to islet autoantigens in type 1 diabetes
JP2000506130A (ja) 抑制性のt細胞反応を誘導するためのdnaワクチン接種
US20210177967A1 (en) Apoptotic cell-mediated induction of antigen specific regulatory t-cells for the therapy of autoimmune diseases in animals and humans
US20230114037A1 (en) Compositions and methods for prevention and treatment of immune deficiency and inflammation
TW202315884A (zh) 使用衍生自患者之抗原特異性調節t細胞的個體化細胞療法
Marshall Cytokine regulation of CD4 (+) T cells from allergic individuals
Van Belle et al. 9 Biologic Therapies for Type 1 Diabetes
Lambert Autoimmune Diabetes and Transplantation Tolerance Induced by Costimulation Blockade in NOD Mice: a Dissertation

Legal Events

Date Code Title Description
AS Assignment

Owner name: ORBAN BIOTECH LLC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ORBAN, TIHAMER;REEL/FRAME:031839/0093

Effective date: 20131220

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION

AS Assignment

Owner name: DMNOMORE, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ORBAN BIOTECH LLC;REEL/FRAME:036880/0640

Effective date: 20151015

AS Assignment

Owner name: PHAIM PHARMA LTD, UNITED KINGDOM

Free format text: CHANGE OF NAME;ASSIGNOR:DMNOMORE LIMITED;REEL/FRAME:060781/0230

Effective date: 20200313