US20170240636A1 - Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28 - Google Patents

Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28 Download PDF

Info

Publication number
US20170240636A1
US20170240636A1 US15/515,461 US201515515461A US2017240636A1 US 20170240636 A1 US20170240636 A1 US 20170240636A1 US 201515515461 A US201515515461 A US 201515515461A US 2017240636 A1 US2017240636 A1 US 2017240636A1
Authority
US
United States
Prior art keywords
domain antibody
dose
domain
bms
study
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
US15/515,461
Other languages
English (en)
Inventor
Xavier Valencia
John P. Throup
Steven G. Nadler
Suzamme J. Suchard
Dominique Duchesne
Xiaoni LIU
Rong Shi
Diane E. Shevell
Jenny H. Xie
Marek Honczarenko
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.)
Bristol Myers Squibb Co
Original Assignee
Bristol-Myers Squibb Company
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 Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to US15/515,461 priority Critical patent/US20170240636A1/en
Publication of US20170240636A1 publication Critical patent/US20170240636A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [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
    • 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
    • A61K47/48215
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • autoimmune diseases such as systemic lupus erythematosus using domain antibodies that specifically bind human CD28 are provided.
  • SLE Systemic lupus erythematosus
  • Autoantibodies such as anti-double-stranded deoxyribonucleic acid (anti-dsDNA) or anti-nuclear antibody (ANA), and have helped to define the autoimmune nature of SLE. These autoantibodies, however, are not pathognomonic. SLE, with its pleiotropic clinical manifestations and lack of specific autoantibodies is the archetype of the non-organ specific autoimmune diseases. American College of Rheumatology (ACR) has developed an 11 factor set of guidelines to diagnose SLE. This intrinsic complexity for diagnosis and monitoring disease progression has hampered the validation of new treatments.
  • ACR American College of Rheumatology
  • T cells have a pivotal role in SLE.
  • SLE is characterized by hyper-responsive T cells, excessive autoantibody production, and antigen presenting cell (APC) hyperactivation.
  • the autoantibodies in particular, anti-nuclear antibodies, and anti-dsDNA antibodies
  • T-cell help is provided by co-stimulatory molecules and cytokines.
  • T cells can directly infiltrate the joints, skin, kidney, and brain causing damage directly through cytotoxicity or indirectly through the recruitment and activation of macrophages and neutrophils.
  • Lymphocytes from patients with SLE show signs of increased activation; e.g., the percentage of CD4+ T cells expressing CD25 increase as does the expression of CD86 on CD19+ B cells.
  • the increased CD86 expression is thought to render (autoreactive) B cells more susceptible to T-cell help and thus facilitate autoantibody production. Consistent with this observation, the number of activated B cells and levels of anti-dsDNA antibodies increase with diseased activity.
  • peripheral blood dendritic cells (DCs) and DCs derived from peripheral blood monocytes of SLE patients show increased expression of CD86 and the ratio of CD86/CD80 is higher in SLE patients compared with healthy donors.
  • CD28 expression on CD4+ and CD8+ T cells in lupus patients appears to be more variable. Regardless of the levels of CD28 expression, T cells from SLE patients appear to be more responsive to anti-CD28-mediated activation and patients with active disease have increased gene expression of CD28 when compared to normal controls. CTLA4 (a co-inhibitory molecule) is also increased in T cells from SLE patients but this does not seem to control aberrant T-cell activation. Taken together, these data suggest that the CD28-CD86/CD80 pathway plays a central role in the defective immune response observed in SLE patients.
  • Inhibition of CD28 mediated T cell activation could inhibit undesired T cell responses occurring during autoimmunity, transplant rejection, or allergic responses.
  • inhibiting CD28 mediated T cell activation could delay graft rejection, prevent acute allograft rejection, induce donor specific tolerance, and prevent development and interrupt the progression of chronic allograft rejection, as well as prevent graft versus host disease (GVH), i.e., when transplanted T cells mount a vigorous immune response against host tissue alloantigens (Salama et al. (2001) J. Clin. Invest. 108: 943-48).
  • GVH graft versus host disease
  • CD28 mediated T cell activation dampen the immune response through negating activation signaling through CD28, it should not impact the interaction of CD86 and CD80 to CTLA4, thereby preserving CTLA4 mediated inhibition of the T cell response.
  • inhibiting CD28 mediated T cell activation could be used to prevent induction of autoimmunity and moderate the progression and/or severity of lupus as well as other autoimmune diseases. (Saloman et al. (2001) Ann. Rev. Immunol. 19: 225-252).
  • the present invention provides a method of treating an immune disease in a patient, comprising administering to the patient a therapeutically effective amount of an anti-CD28 domain antibody which comprises a variable domain, wherein the variable domain comprises the amino acid sequence of SEQ ID NO: 12 (1h-239-891(D70C)) or differs from SEQ ID NO: 12 by up to 5 amino acids, wherein at least one dose of the anti-CD28 domain antibody is administered at a dose from about 1.25 mg to about 12.5 mg.
  • the immune disease is systemic lupus erythematosus (SLE).
  • the patient is a human patient.
  • the anti-CD28 domain antibody is administered at a dose selected from about 1.25 mg, about 5 mg, and about 12.5 mg.
  • the dose is at least 1.25 mg or at least 5 mg.
  • the dose is about 1.25 mg, about 5 mg, or about 12.5 mg.
  • the dose is administered every week or every two weeks.
  • at least 2 doses are administered, wherein the at least 2 doses are the same or different.
  • at least 12 doses are administered.
  • at least 24 doses are administered.
  • variable domain of the anti-CD28 domain antibody comprises: (1) a CDR1 region having the amino acid sequence of SEQ ID NO: 1; (2) a CDR2 region having the amino acid sequence of SEQ ID NO: 2; and (3) a CDR3 region having the amino acid sequence of SEQ ID NO: 3.
  • the anti-CD28 domain antibody comprises the amino acid sequence of SEQ ID NO: 12.
  • the anti-CD28 domain antibody comprises a 40 kDa branched polyethylene glycol.
  • the anti-CD28 domain antibody is BMS-931699.
  • the anti-CD28 domain antibody is administered subcutaneously.
  • the anti-CD28 domain antibody is formulated in a pharmaceutical composition for subcutaneous administration.
  • the anti-CD28 domain antibody is administered intraveneously.
  • the anti-CD28 domain antibody is formulated in a pharmaceutical composition for intraveneous administration.
  • the method of the invention further comprises administering to the patient an immunosuppressive/immunomodulatory and/or anti-inflammatory agent.
  • the immunosuppressive/immunomodulatory and/or anti-inflammatory agent is administered before the anti-CD28 domain antibody.
  • the immunosuppressive/immunomodulatory and/or anti-inflammatory agent is administered after the anti-CD28 domain antibody.
  • the immunosuppressive/immunomodulatory and/or anti-inflammatory agent is administered concurrently with the anti-CD28 domain antibody.
  • method of antagonizing CD28 comprising administering an effective amount of an anti-CD28 dAb disclosed herein to an individual. Also included is a method of antagonizing the binding of CD28 comprising administering an effective amount of the anti-CD28 dAb disclosed herein to an individual, wherein the anti-CD28 dAb antagonizes the binding of CD28 to CD80 and/or CD86 in the individual.
  • a method of treating, alleviating, or preventing a symptom of an immune disease comprising administering an effective amount of an anti-CD28 dAb disclosed herein to an individual having or at risk of having an immune disease.
  • a method of treating, alleviating, or preventing an immune disease comprising administering an effective amount of an anti-CD28 dAb disclosed herein to an individual having or at risk of having an immune disease.
  • an anti-CD28 dAb disclosed herein for preparing a medicament for treating or preventing an immune disease in a patient in need thereof. Also included is the use of an anti-CD28 dAb disclosed herein for preparing a medicament for treating or preventing a symptom of an immune disease in a patient in need thereof. Further included herein is the use of an anti-CD28 dAb disclosed herein for preparing a medicament for alleviating at least one symptom of an immune disease in a patient in need thereof.
  • an anti-CD28 domain antibody formulated in a pharmaceutical composition for subcutaneous administration.
  • the pharmaceutical composition may further comprise a pharmaceutically acceptable carrier.
  • the domain antibody may be administered subcutaneously.
  • kits for treating an immune disease in a patient comprising: (a) a dose of domain antibody comprising an anti-CD28 domain antibody, and (b) instructions for using the domain antibody in the disclosed methods.
  • FIG. 1 shows Part 1 of an FDA Phase 2 parallel-arm, randomized, double-blinded, multicenter, international study, adaptive design schematic for BMS-931699.
  • FIG. 2 shows Part 2 of an FDA Phase 2 parallel-arm, randomized, double-blinded, multicenter, international study, adaptive design schematic for BMS-931699.
  • FIG. 3 shows the long term extension (LTE) design schematic.
  • FIG. 4 demonstrates the expected receptor occupancy for BMS-931699 at steady state for 12.5 mg Weekly (QW), 12.5 mg Every 2 Weeks (QOW), 5 mg Every 2 Weeks (QOW) and 1.25 mg Every 2 Weeks (QOW).
  • the present disclosure provides anti-CD28 domain antibodies to antagonize CD28 activity and methods of treating immune diseases, such as lupus, using said domain antibodies.
  • the domain antibodies may be linked to polymers to improve pharmacokinietic properties, such as stability and half-life.
  • polymers e.g., polyethylene glycol; PEG
  • PEG polyethylene glycol
  • PEG modification of proteins has been shown to alter the in vivo circulating half-life, antigenicity, solubility, and resistance to proteolysis of the protein (Abuchowski et al. (1977) J. Biol. Chem., 252: 3578; Nucci et al. (1991) Adv.
  • the disclosed domain antibodies including BMS-931699 (otherwise known as lulizumab or 1h-239-891(D70C) formatted with a 40 kDa branched polyethylene glycol), monovalently bind CD28, and inhibit the interaction of CD80 and CD86 with CD28, the key co-stimulatory receptor of T lymphocytes.
  • targeting CD28 with the domain antibodies can provide opportunity to inhibit autoimmune processes leading to systemic lupus erythematosus among other graft-related or autoimmune diseases.
  • Such monovalent domain antibodies can also avoid potential undesirable effects that can occur with antibodies capable of divalent or multivalent binding of CD28. Domain antibodies described herein also do not block the interaction of CD80 and CD86 to CTLA4.
  • the domain antibodies described herein do not cross-react with CTLA4, and thus do not bind the common motif on CTLA4 and CD28 that binds CD80/86.
  • the domain antibodies can thus provide improved therapeutic benefits and reduced side-effects for autoimmune disease patients.
  • the domain antibody thus offers a novel therapeutic modality, currently not available to patients. Given the lack of therapeutic options for systemic lupus erythematosus and autoimmune disease subjects who have failed all conventional therapies, the domain antibody with its distinct mechanism of action and known safety profile demonstrates a favorable Risk/Benefit profile.
  • a “fixed dose” is a dose administered regardless of the subjects' body weight.
  • the term “human” when applied to a domain antibody or to an immunoglobulin variable domain means that the polypeptide has a sequence derived from a human immunoglobulin.
  • a sequence is “derived from” a human immunoglobulin coding sequence when the sequence is either: a) isolated from a human individual or from cells or a cell line from a human individual; b) isolated from a library of cloned human antibody gene sequences (or a library of human antibody V domain sequences); or c) when a cloned human antibody gene sequence (or a cloned human V region sequence (including, e.g., a germline V gene segment)) was used to generate one or more diversified sequences that were then selected for binding to a desired target antigen.
  • a human domain antibody has at least 70% identical, at least 75% identical, at least 80% identical, at least 85% amino acid identity (including, for example, 87%, 90%, 93%, 95%, 97%, 99%, or higher identity) to a naturally-occurring human immunoglobulin variable domain sequence, e.g., a naturally-occurring human immunoglobulin variable domain sequence disclosed in Kabat (“Sequences of Proteins of Immunological Interest”, US Department of Health and Human Services 1991).
  • domain refers to a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed, or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain.
  • domain antibody is meant a folded polypeptide domain which comprises a sequence characteristic of immunoglobulin variable domains and which specifically binds an antigen (e.g., dissociation constant of 500 nM or less).
  • a “domain antibody” therefore includes complete antibody variable domains as well as modified variable domains, for example in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain a dissociation constant of 500 nM or less (e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less) and the target antigen specificity of the full-length domain.
  • dissociation constant of 500 nM or less e.g., 450 nM or less, 400
  • a “dAb” is used interchangeably with “domain antibody” herein.
  • a “domain antibody” or “dAb” used in the present invention refers to an “anti-CD28 domain antibody”.
  • sequence characteristic of immunoglobulin variable domains refers to an amino acid sequence that is identical, over 20 or more, 25 or more, 30 or more, 35 or more, 40 or more, 45 or more, or even 50 or more contiguous amino acids, to a sequence comprised by an immunoglobulin variable domain sequence. Sequences similar or identical (e.g., at least about 70% sequence identity) to the sequences disclosed herein are also included herein.
  • identity refers to the degree with which two nucleotide or amino acid sequences structurally resemble each other.
  • sequence “similarity” is a measure of the degree to which amino acid sequences share similar amino acid residues at corresponding positions in an alignment of the sequences. Amino acids are similar to each other where their side chains are similar. Specifically, “similarity” encompasses amino acids that are conservative substitutes for each other. A “conservative” substitution is any substitution that has a positive score in the blosum62 substitution matrix (Hentikoff and Hentikoff (1992) Proc. Natl. Acad. Sci. USA 89: 10915-10919).
  • sequence A is n % similar to sequence B
  • sequence B consists of identical amino acids or conservative substitutions.
  • Typical conservative substitutions are exchanges among Met, Val, Leu, and Ile; among Ser and Thr; among the residues Asp, Glu, and Asn; among the residues Gln, Lys, and Arg; or aromatic residues Phe and Tyr.
  • epitope refers to a unit of structure conventionally bound by an immunoglobulin V H /V L pair. Epitopes define the minimum binding site for an antibody, and thus represent the target of specificity of an antibody. In the case of a domain antibody, an epitope represents the unit of structure bound by a domain antibody in isolation. That is, the binding site is provided by one, single immunoglobulin variable domain. Epitopes can be linear or conformational, and can be as small as three amino acids.
  • CD28 activity is an activity involving or resulting from the binding of CD80, CD86 and/or another ligand to CD28, and includes, but is not limited to, activation of CD28-mediated cell signaling. CD28 activity also includes the induction of T cell proliferation and the induction of cytokine secretion, e.g., interleukin 2 (IL-2), by T cells.
  • IL-2 interleukin 2
  • the term “does not substantially agonize” means that a given agent, e.g., a domain antibody, does not substantially activate one or more of the CD28 activities as the term “activate” is defined herein.
  • an agent that “does not substantially agonize” means that the agent does not activate more than 20% of the activity which is activated by CD80 and/or CD86 binding to CD28, and in an aspect, the agent does not activate more than about 10%, 8%, 5%, 3%, or 2% or less, including zero activation, of the activity which is activated by CD80 and/or CD86 binding to CD28.
  • a domain antibody set forth herein that does not substantially agonize CD28 activity does not agonize CD28 activity more than 5% of the activity obtained upon agonism of CD28 activity by anti-CD28 mAb 9.3 (Gibson, et al. (1996) JBC, 271: 7079-7083) under otherwise identical assay conditions.
  • the terms “inhibit,” “inhibits” and “inhibited” refer to a decrease in a given measurable activity (e.g., binding activity) by at least 10% relative to a reference. Where inhibition is desired, such inhibition is at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, up to and including 100%, i.e., complete inhibition or absence of the given activity. Inhibition of CD28 binding to CD80 or CD86 can be measured as described in the working examples herein. As used herein, the term “substantially inhibits” refers to a decrease in a given measurable activity (e.g., the binding of CD28 to CD80 or CD86) by at least 50% relative to a reference.
  • substantially inhibits refers to a decrease in a given measurable activity of at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and up to and including 100% relative to a reference.
  • “inhibits the binding”, with reference to the binding of a domain antibody binding to CD28, or CD80 binding to CD28, or CD86 binding to CD28 refers to a decrease in binding by at least 10% relative to a reference.
  • “Inhibits the binding” refers to a decrease in binding of at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, up to and including 100%.
  • the terms “activate,” “activates” and “activated” refer to an increase in a given measurable activity by at least 5% relative to a reference, for example, at least 10%, 25%, 50%, 75%, or even 100%, or more.
  • the term “monovalent” means that a given domain antibody can bind only a single molecule of its target.
  • Naturally-occurring antibodies are generally divalent, in that they have two functional antigen-binding loops, each comprising a VH and a VL domain. Where steric hindrance is not an issue, a divalent antibody can bind two separate molecules of the same antigen. In contrast, a “monovalent” antibody has the capacity to bind only one such antigen molecule.
  • a “monovalent” antibody can also comprise more than one antigen binding site, e.g., two antigen binding sites, but the binding sites must be for different antigens, such that the antibody can only bind one molecule of CD28 at a time.
  • the antigen-binding domain of a monovalent antibody can comprise a V H and a V L domain, but in an aspect, comprises only a single immunoglobulin variable domain, i.e., a V H or a V L domain, that has the capacity to bind CD28 without the need for a corresponding V L or V H domain, respectively.
  • a monovalent antibody lacks the capacity to cross link molecules of a single antigen.
  • V H domain and V L domain refer to immunoglobulin variable regions as defined by Kabat et al. (Kabat et al. (1991) Sequences of Immunological Interest, 5th ed. U.S. Dept. Health & Human Services, Washington, D.C.), which is incorporated herein by reference.
  • “linked” refers to the attachment of a polymer moiety, such as PEG to an amino acid residue of a domain antibody. Attachment of a PEG polymer to an amino acid residue of a domain antibody, e.g., a domain antibody, is referred to as “PEGylation” and may be achieved using several PEG attachment moieties including, but not limited to N-hydroxylsuccinimide (NHS) active ester, succinimidyl propionate (SPA), maleimide (MAL), vinyl sulfone (VS), or thiol.
  • a PEG polymer, or other polymer can be linked to a domain antibody at either a predetermined position, or may be randomly linked to the domain antibody molecule.
  • the PEG polymer may be linked to a domain antibody at a predetermined position.
  • a PEG polymer may be linked to any residue in a domain antibody, however, it is preferable that the polymer is linked to either a lysine or cysteine, which is either naturally occurring in the domain antibody or which has been engineered into the domain antibody, for example, by mutagenesis of a naturally occurring residue in the domain antibody to either a cysteine or lysine.
  • PEG-linkage can also be mediated through a peptide linker attached to a domain antibody.
  • the PEG moiety can be attached to a peptide linker fused to a domain antibody, where the linker provides the site, e.g., a free cysteine or lysine, for PEG attachment.
  • linker provides the site, e.g., a free cysteine or lysine, for PEG attachment.
  • “linked” can also refer to the association of two or more domain antibodies, e.g., dAb monomers, to form a dimer, trimer, tetramer, or other multimer.
  • Domain antibody monomers can be linked to form a multimer by several methods known in the art, including, but not limited to, expression of the domain antibody monomers as a fusion protein, linkage of two or more monomers via a peptide linker between monomers, or by chemically joining monomers after translation, either to each other directly, or through a linker by disulfide bonds, or by linkage to a di-, tri- or multivalent linking moiety (e.g., a multi-arm PEG).
  • a fusion protein linkage of two or more monomers via a peptide linker between monomers, or by chemically joining monomers after translation, either to each other directly, or through a linker by disulfide bonds, or by linkage to a di-, tri- or multivalent linking moiety (e.g., a multi-arm PEG).
  • dAb multimers are specifically contemplated herein, e.g., in the context of dual- or multi-specific domain antibody constructs, it is emphasized that for any given domain antibody construct, the construct should only be able to bind one molecule of CD28, i.e., the constructs should have only one CD28-binding element, and should not cross link CD28.
  • polymer refers to a macromolecule made up of repeating monomeric units, and can refer to a synthetic or naturally occurring polymer such as an optionally substituted straight or branched chain polyalkylene, polyalkenylene, or polyoxyalkylene polymer or a branched or unbranched polysaccharide.
  • PEG polyethylene glycol
  • PEG polymer refers to polyethylene glycol, and more specifically can refer to a derivatized form of PEG, including, but not limited to N-hydroxylsuccinimide (NHS) active esters of PEG such as succinimidyl propionate, benzotriazole active esters, PEG derivatized with maleimide, vinyl sulfones, or thiol groups.
  • NHS N-hydroxylsuccinimide
  • PEG formulations can include PEG-O—CH 2 CH 2 CH 2 —CO 2 —NHS; PEG-O—CH 2 —NHS; PEG-O—CH 2 CH 2 —CO 2 —NHS; PEG-S—CH 2 CH 2 —CO—NHS; PEG-O 2 CNH—CH(R)—CO 2 —NHS; PEG-NHCO—CH 2 CH 2 —CO—NHS; and PEG-O—CH 2 —CO 2 —NHS; where R is (CH 2 ) 4 )NHCO 2 (mPEG).
  • PEG polymers set forth herein may be linear molecules, or may be branched wherein multiple PEG moieties are present in a single polymer.
  • PEG or another agent e.g., HSA
  • a domain antibody as described herein in an exemplary embodiment, will not impair the ability of the polypeptide to specifically bind CD28. That is, the PEG-linked domain antibody will retain its binding activity relative to a non-PEG-linked counterpart.
  • “retains activity” refers to a level of activity of a PEG-linked domain antibody which is at least 10% of the level of activity of a non-PEG-linked domain antibody, including at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, and up to 90%, including up to about 95%, 98%, and up to 100% of the activity of a non-PEG-linked domain antibody comprising the same antigen-binding domain or domains. More specifically, the activity of a PEG-linked domain antibody compared to a non-PEG linked domain antibody should be determined on a molar basis; that is equivalent numbers of moles of each of the PEG-linked and non-PEG-linked domain antibody should be used in each trial. In determining whether a particular PEG-linked domain antibody “retains activity”, the activity of a PEG-linked domain antibody may be compared with the activity of the same domain antibody in the absence of PEG.
  • IC 50 refers to the concentration of an inhibitor necessary to inhibit a given activity by about 50%. IC 50 is determined by assaying a given activity, e.g., binding of CD28 to CD80 or CD86, in the presence of varying amounts of the inhibitor (e.g., domain antibody), and plotting the inhibitor concentration versus the activity being targeted. Binding of CD28 to CD80 or CD86 is measured herein by the method described the working examples. Alternatively, surface plasmon resonance (SPR) can be used.
  • SPR surface plasmon resonance
  • EC 50 refers to the concentration of compound or domain antibody that provokes a response in a subject, wherein the response is halfway between the baseline and the maximum response.
  • the baseline and maximum responses of a subject, with respect to a compound or domain antibody, can be determined by any technique known in the art.
  • fused to a domain antibody generally means that a polypeptide is fused to a given antibody through use of recombinant DNA techniques, though fusion may occur chemically at the protein level.
  • an antibody “fused to” another polypeptide e.g., to another antibody of different binding specificity, does not exist in nature and is generated through recombinant means.
  • the term “fused to a domain antibody” also encompasses the linkage of a polypeptide to a given domain antibody through, for example, disulfide or other chemical linkages, where the fused polypeptide is not naturally found fused to the domain antibody. Recombinant and chemical methods of fusing a polypeptide to another polypeptide, e.g., to an antibody, are well known in the art.
  • Fc domain refers to the constant region antibody sequences comprising CH2 and CH3 constant domains as delimited according to Kabat et al., supra.
  • the Fc portion of the heavy chain polypeptide has the ability to self-associate, a function which facilitates the formation of divalent antibodies.
  • the term “lacks an Fc domain” means that a given domain antibody lacks at least the portion of an immunoglobulin Fc domain (as such domains are defined according to Kabat et al., 1991, Sequences of Immunological Interest, 5 th ed. U.S. Dept. Health & Human Services, Washington, D.C.) sufficient to mediate the dimerization of Fc-containing domain antibodies. Dimerization of Fc-containing domain antibodies is measured, for example, by chromatographic methods or by surface plasmon resonance. A domain antibody lacking an Fc domain avoids Fc-platelet interactions and therefore avoids induction of platelet aggregation.
  • universal framework refers to a single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat (Kabat et al. (1991) Sequences of Immunological Interest, 5 th ed. U.S. Dept. Health & Human Services, Washington, D.C.) or corresponding to the human germline immunoglobulin repertoire or structure as defined by Chothia and Lesk, (1987) J. Mol. Biol. 196: 910-917.
  • the present invention relates to domain antibodies that specifically bind and inhibit human CD28 (“anti-CD28 domain antibodies”) and that are useful in the treatment of diseases involving the CD28 pathway. Accordingly, a method of treating an immune disease in a patient in need of such treatment is provided comprising administering to the patient a therapeutically effective amount of the anti-CD28 domain antibody.
  • Domain antibodies are provided that are monovalent for binding to CD28. While not wishing to be bound by any particular theory, it is believed that monovalency for CD28 binding removes the possibility for cross-linking cell surface receptors that occurs with prior art antibodies. Thus, in one aspect, the domain antibodies disclosed herein not only inhibit or antagonize the binding of CD80 or CD86 to CD28, they do not substantially agonize CD28 activity.
  • the antibodies monovalent for CD28 binding are human domain antibodies.
  • Human domain antibodies can be administered to human patients while largely avoiding the anti-antibody immune response often provoked by the administration of antibodies from other species, e.g., mouse.
  • murine antibodies can be “humanized” by grafting human constant domains onto the murine antigen-binding domains, human antibodies as disclosed herein are produced without the need for laborious and time-consuming genetic manipulation of a murine antibody sequence.
  • domain antibodies may include one or more of the following CDRs:
  • the anti-CD28 domain antibody has a CDR1 sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 1.
  • the CDR1 differs from SEQ ID NO: 1 by up to 5 amino acids (e.g., by 5, 4, 3, 2, 1, or 0 amino acids).
  • the anti-CD28 domain antibody has a CDR2 sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 2.
  • the CDR2 differs from SEQ ID NO: 2 by up to 5 amino acids (e.g., by 5, 4, 3, 2, 1, or 0 amino acids).
  • the anti-CD28 domain antibody has a CDR3 sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 3.
  • the CDR3 differs from SEQ ID NO: 3 by up to 5 amino acids (e.g., by 5, 4, 3, 2, 1, or 0 amino acids).
  • the anti-CD28 domain antibody comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence selected from SEQ ID NOs: 4-15.
  • the anti-CD28 domain antibody differs from the selected amino acid sequence by up to 5 amino acids.
  • the domain antibody differs from SEQ ID NO: 12 by up to 5 amino acids (e.g., by 5, 4, 3, 2, 1, or 0 amino acids).
  • the anti-CD28 domain antibody comprises an amino acid sequence selected from SEQ ID NOs: 4-15. In a specific embodiment, the anti-CD28 domain antibody comprises the amino acid sequence of SEQ ID NO: 12.
  • the anti-CD28 domain antibody may comprise 1h-239-891(D70C) (SEQ ID NO: 543).
  • the anti-CD28 domain antibody may comprise a 40 kDa branched polyethylene glycol.
  • the anti-CD28 domain antibody can be BMS-931699.
  • BMS-931699 is a potent inhibitor of T cell proliferation and cytokine production, with an EC 50 of 35 ⁇ 14 ng/mL and 25 ⁇ 6 ng/mL, respectively.
  • BMS-908613-P40B (a macaque surrogate for BMS-931699) is equipotent at inhibiting both CD80- and CD86-driven T cell proliferation.
  • no agonist or co-agonist activity was observed with BMS-931699.
  • the binding of the domain antibody to CD28 does not substantially agonize CD28 activity.
  • the dAb does not agonize CD28 signaling in combination with T cell receptor signaling.
  • the domain antibody inhibits the binding of CD28 to CD80.
  • the domain antibody inhibits the binding of CD28 to CD80, and does not substantially agonize signaling by CD28.
  • the domain antibody inhibits the binding of CD28 to CD86.
  • the domain antibody inhibits the binding of CD28 to CD86, and does not substantially agonize signaling by CD28.
  • a cytokine is at least one cytokine selected from the group consisting of GM-CSF, IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, IL-12 IL-13, IL-15, IL-17, IL-21, IL-22, IL-24, TGF ⁇ , TNF- ⁇ , TNF- ⁇ , IFN- ⁇ , IFN- ⁇ , IFN- ⁇ .
  • the domain antibody is a human domain antibody that monovalently binds CD28, and in an exemplary embodiment, without substantially agonizing CD28 activity.
  • the domain antibody interacts with human CD28 with a K d in the range of 50 nM to 1 pM, inclusive, as measured by surface plasmon resonance.
  • the K d for human CD28 can be 25 nM to 20 pM, 10 nM to 20 pM, 5 nm to 20 pM, 1 nM to 20 pM, 0.5 nM to 20 pM, 0.1 nM to 20 pM, 0.1 nM to 50 pM, 75 pM to 20 pM, or even 50 pM to 20 pM.
  • the K d for human CD28 is about 50 pM.
  • the domain antibody inhibits binding of CD80 to CD28 with an EC 50 of 50 nM or less. In one embodiment, the domain antibody inhibits binding of CD86 to CD28 with an EC 50 of 50 nM or less. In a further embodiment, the domain antibody has binding specificity to CD28 with a K off rate constant of 1 ⁇ 10 ⁇ 3 s ⁇ 1 or less, 1 ⁇ 10 ⁇ 4 s ⁇ 1 or less, 1 ⁇ 10 ⁇ 5 s ⁇ 1 or less, or 1 ⁇ 10 ⁇ 6 s ⁇ 1 or less, as determined by surface plasmon resonance. In one embodiment, the domain antibody neutralizes CD28 in a standard assay with a EC 50 of 50 nM or less.
  • the domain antibody comprises a single immunoglobulin variable domain that binds CD28.
  • the single immunoglobulin variable domain is a V H or a V L domain.
  • the domain antibody comprises a homomultimer or heteromultimer of two variable domains, e.g., a V H and V L domain, but one of the variable domains has the capacity to bind CD28 without the need for a corresponding V L or V H domain. That is, the dAb binds an antigen independently of the additional V H or V L domains.
  • the variable domains in these embodiments may comprise three complementarity determining regions (CDRs).
  • the domain antibody is free of an Fc domain.
  • an Fc domain consists of the CH2—CH3 regions, optionally including a hinge region linked to the CH2.
  • the domain antibody comprises a universal framework.
  • a domain antibody may comprise one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a corresponding framework (FW) region encoded by a human germline antibody gene segment, or the amino acid sequence of one or more of said framework regions collectively comprising up to 5, e.g., 1, 2, 3, 4 or 5, amino acid differences relative to the amino acid sequence of said corresponding framework region encoded by a human germline antibody gene segment.
  • the dAb comprises amino acid sequences of FW1, FW2, FW3, and FW4 that correspond to the FW1, FW2, FW3, and FW4 of a human antibody, e.g., a human germline antibody.
  • some or all of the amino acid sequences of FW1, FW2, FW3, and FW4 of the domain antibody are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • FW2 may be identical to the FW2 of a human antibody.
  • the amino acid sequences of FW1, FW2, FW3, and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
  • the human germline antibody gene segment can be selected from the group consisting of DP47, DP45, DP48, and DPK9.
  • the universal framework comprises a V H framework selected from the group consisting of DP47, DP45, and DP38, and/or the V L framework is DPK9.
  • a domain antibody is formatted to increase its in vivo half-life.
  • the domain antibody has an increased in vivo t- ⁇ or t- ⁇ half-life relative to the same unformatted domain antibody.
  • the t ⁇ -half-life of the domain antibody composition is increased by 10% or more when compared to an unmodified protein assayed under otherwise identical conditions. In another embodiment, the t ⁇ -half-life of the domain antibody composition is increased by 50% or more. In another embodiment, the t ⁇ -half-life of the domain antibody composition is increased by 2 ⁇ or more. In another embodiment, the t ⁇ -half-life of the domain antibody composition is increased by 5 ⁇ or more, e.g., 10 ⁇ , 15 ⁇ , 20 ⁇ , 25 ⁇ , 30 ⁇ , 40 ⁇ , 50 ⁇ , or more. In another embodiment, the t ⁇ -half-life of the domain antibody composition is increased by 100 ⁇ , 200 ⁇ , 300 ⁇ , 400 ⁇ , 500 ⁇ , or more.
  • the domain antibody has a t ⁇ half-life of 0.25 to 6 hours, inclusive. In another embodiment, the t ⁇ half-life is in the range of 30 minutes to 12 hours, inclusive. In another embodiment, the t ⁇ -half-life of the domain antibody is in the range of 1 to 6 hours.
  • the t ⁇ -half-life of the domain antibody is increased by 10% or more when compared to an unmodified protein assayed under otherwise identical conditions. In another embodiment, the t ⁇ -half-life of the domain antibody is increased by 50% or more. In another embodiment, the t ⁇ -half-life of the antibody domain antibody is increased by 2 ⁇ or more. In another embodiment, the t ⁇ -half-life of the domain antibody is increased by 5 ⁇ or more, e.g., 10 ⁇ , 15 ⁇ , 20 ⁇ , 25 ⁇ , 30 ⁇ , 40 ⁇ , or more. In another embodiment, the t ⁇ -half-life of the domain antibody is increased by 50 ⁇ or more.
  • the domain antibody has a t ⁇ half-life of 1 hour to 744 hours, inclusive. In another embodiment, the t ⁇ -half-life is in the range of 12 to 48 hours, inclusive. In another embodiment, the t ⁇ half-life is in the range of 12 to 26 hours, inclusive. In yet another embodiment, the t ⁇ half-life is about 336 hours.
  • a domain antibody-containing composition comprising a ligand having an AUC value (area under the curve) in the range of 1 mg ⁇ min/ml or more.
  • the lower end of the range is 5, 10, 15, 20, 30, 100, 200, or 300 mg ⁇ min/ml.
  • a ligand or composition has an AUC in the range of up to 600 mg ⁇ min/ml.
  • the upper end of the range is 500, 400, 300, 200, 150, 100, 75, or 50 mg ⁇ min/ml.
  • a ligand will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg ⁇ min/ml, 15 to 100 mg ⁇ min/ml, 15 to 75 mg ⁇ min/ml, and 15 to 50 mg ⁇ min/ml.
  • the formatting comprises PEGylation of the dAb.
  • the PEG is covalently linked.
  • the PEG is linked to the domain antibody at a cysteine or lysine residue.
  • the PEG-linked domain antibody has a hydrodynamic size of at least 24 kD.
  • the total PEG size is from 20 to 60 kD, inclusive.
  • the PEG-linked domain antibody has a hydrodynamic size of at least 200 kD.
  • the PEG-linked domain antibody has an increased in vivo half-life relative to the same polypeptide composition lacking linked polyethylene glycol.
  • the t ⁇ -half-life of the domain antibody composition is increased by 10% or more.
  • the t ⁇ -half-life of the domain antibody composition is increased by 50% or more.
  • the t ⁇ -half-life of the domain antibody composition is increased by 2 ⁇ or more.
  • the t ⁇ -half-life of the domain antibody composition is increased by 5 ⁇ or more, e.g., 10 ⁇ , 15 ⁇ , 20 ⁇ , 25 ⁇ , 30 ⁇ , 40 ⁇ , 50 ⁇ , or more.
  • the t ⁇ -half-life of the domain antibody composition is increased by 100 ⁇ , 200 ⁇ , 300 ⁇ , 400 ⁇ , 500 ⁇ , or more.
  • the PEG-linked domain antibody has a t ⁇ half-life of 0.25 to 6 hours, inclusive. In another embodiment, the t ⁇ half-life is in the range of 30 minutes to 12 hours, inclusive. In another embodiment, the t ⁇ -half-life of the domain antibody is in the range of 1 to 6 hours.
  • the t ⁇ -half-life of the PEG-linked domain antibody is increased by 10% or more. In another embodiment, the t ⁇ -half-life of the PEG-linked domain antibody is increased by 50% or more. In another embodiment, the t ⁇ -half-life of the PEG-linked domain antibody is increased by 2 ⁇ or more. In another embodiment, the t ⁇ -half-life of the PEG-linked domain antibody is increased by 5 ⁇ or more, e.g., 10 ⁇ , 15 ⁇ , 20 ⁇ , 25 ⁇ , 30 ⁇ , 40 ⁇ , or more. In another embodiment, the t ⁇ -half-life of the PEG-linked domain antibody is increased by 50 ⁇ or more.
  • the PEG-linked domain antibody has a t ⁇ half-life of 1 to 170 hours, inclusive. In another embodiment, the t ⁇ -half-life is in the range of 12 to 48 hours, inclusive. In another embodiment, the t ⁇ -half-life is in the range of 12 to 26 hours, inclusive.
  • the PEG-linked domain antibody has an AUC value (area under the curve) in the range of 1 mg ⁇ min/ml or more.
  • the lower end of the range is about 5, 10, 15, 20, 30, 100, 200, or 300 mg ⁇ min/ml.
  • a ligand or composition has an AUC in the range of up to about 600 mg ⁇ min/ml.
  • the upper end of the range is about 500, 400, 300, 200, 150, 100, 75, or 50 mg ⁇ min/ml.
  • a ligand will have an AUC in the range selected from the group consisting of the following: about 15 to 150 mg ⁇ min/ml, about 15 to 100 mg ⁇ min/ml, about 15 to 75 mg ⁇ min/ml, and about 15 to 50 mg ⁇ min/ml.
  • the dAb may inhibit binding of CD28 to CD80 and/or CD86 with an EC 50 of about 100 nM, about 50 nM, about 1 nM, about 500 pM, about 100 pM, about 50 pM, about 10 pM, about 5 pM, or about 1 pM.
  • the domain antibody inhibits binding of CD28 to CD80 with an EC 50 in the range of 1 pM to 1.5 ⁇ M, inclusive; EC 50 for inhibition of CD28 binding to CD80.
  • the EC 50 can be in the range of 1 pM to 1 ⁇ M, 1 pM to 900 nM, 1 pM to 800 nM, 1 pM to 700 nM, 1 pM to 600 nM, 1 pM to 500 nM, 1 pM to 400 nM, 1 pM to 300 nM, 1 pM to 200 nM, 1 pM to 100 nM, 1 pM to 50 nM, 1 pM to 10 nM, 1 pM to 1 nM, 1 pM to 500 pM, 1 pM to 100 pM, 1 pM to 50 pM, 1 pM to 10 pM, or 1 pM to 5 pM.
  • Further acceptable ranges include, for example, 50 pM to 1 ⁇ M, 100 pM to 500 nM, 125 pM to 250 nM, 150 pM to 200 nM, 150 pM to 100 nM, and 200 pM to 50 nM.
  • the domain antibody inhibits binding of CD28 to CD86 with an EC 50 in the range of 1 pM to 1.5 ⁇ M, inclusive; EC 50 for inhibition of CD28 binding to CD86.
  • the EC 50 can be in the range of 1 pM to 1 ⁇ M, 1 pM to 900 nM, 1 pM to 800 nM, 1 pM to 700 nM, 1 pM to 600 nM, 1 pM to 500 nM, 1 pM to 400 nM, 1 pM to 300 nM, 1 pM to 200 nM, 1 pM to 100 nM, 1 pM to 50 nM, 1 pM to 10 nM, 1 pM to 1 nM, 1 pM to 500 pM, 1 pM to 100 pM, 1 pM to 50 pM, 1 pM to 10 pM, or 1 pM to 5 pM.
  • Further acceptable ranges include, for example, 50 pM to 1 ⁇ M, 100 pM to 500 nM, 125 pM to 250 nM, 150 pM to 200 nM, 150 pM to 100 nM, and 200 pM to 50 nM.
  • the domain antibody may comprise one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a corresponding framework region encoded by a human germline antibody gene segment, or the amino acid sequence of one or more of said framework regions collectively comprises up to 5 amino acid differences relative to the amino acid sequence of said corresponding framework region encoded by a human germline antibody gene segment.
  • the amino acid sequences of FW1, FW2, FW3, and FW4 of the domain antibody are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the amino acid sequences of FW1, FW2, FW3, and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
  • the amino acid sequences of said FW1, FW2, and FW3 of the domain antibody are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • the human germline antibody gene segments may be selected from the group consisting of DP47, DP45, DP48, and DPK9.
  • Domain antibodies as described herein are useful for antagonizing the activity of CD28. Therefore, domain antibodies as described herein can be used to treat a patient having a condition, disease or disorder mediated in whole or in part by CD28 activity. For example, domain antibodies as described herein are useful for the treatment or prevention of diseases or disorders in which inappropriate activation of a CD28-mediated pathway is involved, such as systemic lupus erythematosus (SLE).
  • SLE systemic lupus erythematosus
  • symptom of disease refers to a decrease of a symptom by at least 10% based on a clinically measurable parameter, or by at least one point on a clinically-accepted scale of disease or symptom severity.
  • symptom(s) of systemic lupus erythematosus refers to any of the clinically relevant symptoms of SLE known to those of skill in the art, including, but not limited to, BICLA (BILAG-Based Composite Lupus Assessment), SRI (Systemic Lupus Erythematosus Responder Index).
  • Non-limiting examples include the accumulation of IgG autoantibodies (e.g., against nuclear antigens such as chromatin, snRNPs (especially U1, Sm, Ro/SSA and La/SSB), phospholipids and cell surface molecules), hemolytic anemia, thrombocytopenia, leukopenia, glomerulonephritis, vasculitis, arthritis, and serositis).
  • a reduction in such a symptom of a patient is a reduction by at least 10% in a clinically measurable parameter, or by at least one point on a clinically-accepted scale of disease severity, compared to a patient treated with a placebo.
  • autoimmune diseases frequently involve inappropriate regulation or activity of CD28 pathways.
  • Administration of a domain antibody as described herein to an individual suffering from such a disease, including an autoimmune disease can reduce one or more symptoms of the disease.
  • diseases for which the domain antibodies described herein can be therapeutically useful include, but are not limited to, Addison's disease, allergy, ankylosing spondylitis, asthma, atherosclerosis, autoimmune diseases of the ear, autoimmune diseases of the eye, autoimmune atrophic gastritis, autoimmune hepatitis, autoimmune hymolytic anemia, autoimmune parotitis, primary biliary cirrhosis, benign lymphocytic aniitis, colitis, coronary heart disease, Crohn's disease, diabetes (Type I), diabetes, including Type 1 and/or Type 2 diabetes, epididymitis, glomerulonephritis, Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease,
  • Autoimmune-mediated conditions include, but are not limited to, conditions in which the tissue affected is the primary target, and in some cases, the secondary target.
  • Such conditions include, but are not limited to, AIDS, atopic allergy, bronchial asthma, eczema, leprosy, schizophrenia, inherited depression, transplantation of tissues and organs, chronic fatigue syndrome, Alzheimer's disease, Parkinson's disease, myocardial infarction, stroke, autism, epilepsy, Arthus's phenomenon, anaphylaxis, and alcohol and drug addiction.
  • the domain antibodies described herein also can be therapeutically useful in graft-related diseases, such as graft versus host disease (GVHD), acute transplantation rejection, and chronic transplantation rejection.
  • graft-related diseases such as graft versus host disease (GVHD), acute transplantation rejection, and chronic transplantation rejection.
  • the present invention provides a method of treating an immune disease in a patient, comprising administering to the patient a therapeutically effective amount of an anti-CD28 domain antibody.
  • the immune disease may be systemic lupus erythematosus.
  • the patient receiving an anti-CD28 domain antibody may have decreased SLE symptoms compared to a patient receiving placebo.
  • the patient receiving an anti-CD28 domain antibody may have lower levels of C3, C4, anti-dsDNA, and/or anti-ANA compared to a patient receiving placebo.
  • the patient receiving an anti-CD28 domain antibody may have decreased arthritis symptoms compared to a patient receiving placebo.
  • the patient receiving an anti-CD28 domain antibody may have decreased inflammatory skin disease symptoms compared to a patient receiving placebo.
  • the treatment may further comprise administering an immunosuppressive/immunomodulatory and/or anti-inflammatory agent.
  • the treatment may be administered in combination with an immunosuppressive/immunomodulatory and/or anti-inflammatory agent.
  • additional immunosuppressive/immunomodulatory and/or anti-inflammatory agents or therapies may comprise calcineuirin inhibitor, cyclosporine, cytoxan, prednisone, azathioprine, methotrexate, corticosteroids, nonsteroidal antiinflammatory drugs/Cox-2 inhibitors, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, infliximab, anakinra, mizoribine, mycophenolic acid, mycophenolate mofetil, interferon beta-1a, interferon beta-1b, glatiramer acetate, mitoxantrone hydrochloride, and/or other biologics like anti-TNF.
  • the domain antibodies also may be administered in combination with one or more of the following agents to regulate an immune response: CTLA4, soluble gp39 (also known as CD40 ligand (CD40L), CD154, T-BAM, TRAP), soluble CD29, soluble CD40, soluble CD80, soluble CD86, soluble CD56, soluble Thy-1, soluble CD3, soluble TCR, soluble VLA-4, soluble VCAM-1, soluble LECAM-1, soluble ELAM-1, soluble CD44, antibodies reactive with gp39, antibodies reactive with CD40, antibodies reactive with B7, antibodies reactive with CD28, antibodies reactive with LFA-1, antibodies reactive with LFA-2, antibodies reactive with IL-2, antibodies reactive with IL-12, antibodies reactive with IFN-gamma, antibodies reactive with CD2, antibodies reactive with CD48, antibodies reactive with any ICAM (e.g., ICAM-2), antibodies reactive with CTLA4, antibodies reactive with Thy-1, antibodies reactive with CD56, antibodies reactive with CD3, antibodies reactive with CD29, antibodies reactive with TCR
  • domain antibodies of the invention are administered in combination with another immunosuppressive/immunomodulatory or anti-inflammatory agent or therapy, e.g., as specified above, the administration may be made concomitantly or in sequence.
  • the dAbs are administered concomitantly with another agent, such as an agent specified above, the dAb and agent may administered in the same pharmaceutical composition.
  • the treatment may produce at least one therapeutic effect measurable by a biomarker selected from the group consisting of: CD28 receptor occupancy on T cells, C3, C4, anti-dsDNA, anti-ANA, anti-Ro autoantibodies, anti-La autoantibodies, anti-RNP autoantibodies, anti-Sm autoantibodies, anti-APL autoantibodies, CRP, total IgG, total IgM, RNA transcripts in whole blood, NGAL in urine, TWEAK in urine, MCP-1 in urine, IL-18 in urine, IL-1 in urine, total soluble CD28, T cell activation, leukocyte surface CD4, leukocyte surface CD8, leukocyte surface CD28, leukocyte surface CD57 and leukocyte intracellular granzyme B, serum IL-6, serum IL-18, serum TNF- ⁇ , serum ⁇ -interferon, serum BLyS(BAFF), CD154, sCD28 and microvessicles.
  • a biomarker selected from the group consisting of: CD
  • the anti-CD28 domain antibodies set forth herein can be incorporated into pharmaceutical compositions suitable for administration to a subject.
  • the pharmaceutical composition comprises the domain antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial, and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carrier excludes tissue culture medium comprising bovine or horse serum.
  • pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable substances include minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the domain antibody.
  • compositions as described herein may be in a variety of forms. These include, for example, liquid, semi-solid, and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, powders, liposomes, and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions e.g., dispersions or suspensions
  • powders e.g., liposomes, and suppositories.
  • Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies.
  • One mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions methods of preparation include vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the domain antibodies described herein can be administered by a variety of methods known in the art, although for many therapeutic applications.
  • the polypeptide can be administered by instravenous (IV), intramuscular (IM) or subcutaneous (SC) injection.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, and microencapsulated delivery systems.
  • Domain antibodies are well suited for formulation as extended release preparations due, in part, to their small size, the number of moles per dose can be significantly higher than the dosage of, e.g., full sized antibodies.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • agents that delays absorption for example, monostearate salts and gelatin.
  • Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. Additional methods applicable to the controlled or extended release of polypeptide agents such as the monovalent domain antibodies disclosed herein are described, for example, in U.S. Pat. Nos. 6,306,406 and 6,346,274, as well as, for example, in U.S. Patent Publication Nos.
  • the domain antibody can be formulated in a pharmaceutical composition for subcutaneous administration, which can include a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can further comprise 12.5 mg/mL BMS-931699 in 20 mM phosphate, pH 5.9, with 5% (w/v) sorbitol.
  • a domain antibody is co-formulated with and/or co-administered with one or more additional therapeutic agents.
  • a domain antibody can be co-formulated and/or co-administered with one or more additional antibodies that bind other targets (e.g., antibodies that bind other cytokines or that bind cell surface molecules), or, for example, one or more cytokines.
  • targets e.g., antibodies that bind other cytokines or that bind cell surface molecules
  • Such combination therapies may utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
  • compositions disclosed herein can include a “therapeutically effective amount” or a “prophylactically effective amount” of a domain antibody.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the domain antibody can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of domain antibody to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, because a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • a non-limiting range for a therapeutically or prophylactically effective amount of a domain antibody is 0.1-20 mg/kg, and in an aspect, 1-10 mg/kg.
  • the domain antibody can be administered at a dose of about 1.25 mg to about 12.5 mg; i.e. the dose can be at least 1.25 mg, at least 5 mg, at least 12.5 mg, about 1.25 mg, about 5 mg, or about 12.5 mg. 1.25 mg to about 12.5 mg mg mg of domain antibody can be administered subcutaneously.
  • the dose may be administered on a fixed or varied schedule. For example, the dose may be administered on a weekly or bi-weekly basis.
  • the dose can be administered over a set treatment regimen, such as once every two weeks for 24 weeks (12 doses total) or once a week for 24 weeks (24 doses total). It is to be noted that dosage values can vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the administering clinician.
  • the efficacy of treatment with a domain antibody as described herein is judged by the skilled clinician on the basis of improvement in one or more symptoms or indicators of the disease state or disorder being treated.
  • An improvement of at least 10% (increase or decrease, depending upon the indicator being measured) in one or more clinical indicators is considered “effective treatment,” although greater improvements are included, such as about 20%, 30%, 40%, 50%, 75%, 90%, or even 100%, or, depending upon the indicator being measured, more than 100% (e.g., two-fold, three-fold, ten-fold, etc., up to and including attainment of a disease-free state.
  • Indicators can be physical measurements, e.g., enzyme, cytokine, growth factor or metabolite levels, rate of cell growth or cell death, or the presence or amount of abnormal cells.
  • One can also measure, for example, differences in the amount of time between flare-ups of symptoms of the disease or disorder (e.g., for remitting/relapsing diseases, such as multiple sclerosis).
  • non-physical measurements such as a reported reduction in pain or discomfort or other indicator of disease status can be relied upon to gauge the effectiveness of treatment.
  • various clinically acceptable scales or indices can be used, for example, the Crohn's Disease Activity Index, or CDAI (Best et al.
  • Gastroenterology 70: 439 which combines both physical indicators, such as hematocrit and the number of liquid or very soft stools, among others, with patient-reported factors such as the severity of abdominal pain or cramping and general well-being, to assign a disease score.
  • prophylaxis performed using a composition as described herein is “effective” if the onset or severity of one or more symptoms is delayed or reduced by at least 10%, or abolished, relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
  • the domain antibodies described herein bind human CD28, where one is to evaluate its effect in an animal model system
  • the polypeptide must cross-react with one or more antigens in the animal model system, in an aspect, at high affinity.
  • One of skill in the art can readily determine if this condition is satisfied for a given animal model system and a given domain antibody. If this condition is satisfied, the efficacy of the domain antibody can be examined by administering it to an animal model under conditions which mimic a disease state and monitoring one or more indicators of that disease state for at least a 10% improvement.
  • Tables 1 and 2 summarize the IV and SC PK parameters of 1h-239-891(D70C)-P30L and -P40B, respectively.
  • Nonlinear pharmacokinetics were observed for both P30L and P40B PEG formats.
  • the terminal half-lives (T1 ⁇ 2) of P30L and P40B were 1.6 and 2.5 days, respectively.
  • the absorption of 1h-239-891(D70C) was nearly complete after SC dosing for P30L at 0.05 mg/kg and for P40B at 0.05 and 5 mg/kg (bioavailability F>90%), but reduced to 70% for P30L at 5 mg/kg.
  • the steady-state volume of distribution (Vss) was comparable between the two PEG formats.
  • CD28 dAbs that recognize mouse CD28 were used to evaluate the impact of direct inhibition of CD28 in murine efficacy models.
  • BMS1m-74-14982-P40B (a murine surrogate for BMS-931699) completely suppressed IgG titers at 0.1 mg/kg in mice, with an in vivo EC 50 of 18 ng/mL.
  • CD28 receptor occupancy (RO) of ⁇ 100% 24 hours post dose was required for maximal efficacy.
  • mCTLA4Ig completely suppressed the IgG response at 3 mg/kg, with an in vivo EC 50 of 1656 ng/mL.
  • the nonclinical safety assessment that supports the clinical development of BMS-931699 includes: 1) single-dose PK/PD studies conducted with BMS-908613-P40B in monkeys to support the minimal anticipated biological effect level (MABEL) dose rationale; 2) a single-dose exploratory toxicity study in mice with BMS1m-74-14982-S60C-P40B to assess potential toxicity of CD28 antagonism in a rodent model; 3) pivotal GLP 1- and 6-month repeat-dose toxicity studies of BMS-931699 in cynomolgus monkeys; 4) an exploratory in vitro study of potential BMS-931699-related effects (cytokine release, T-cell activation/proliferation) on human T cells; and 5) a Good Laboratory Practice (GLP) human tissue cross-reactivity study to demonstrate target distribution and inform of any potential unexpected epitope binding.
  • MABEL minimal anticipated biological effect level
  • the cynomolgus monkey was selected as the toxicology species because BMS-931699 binds comparably to macaque CD28, is pharmacologically active in monkeys, and does not cross-react with rodent CD28.
  • BMS-908613-P40B inhibits TDARs
  • BMS-931699 decreases in cortical lymphocytes in various lymph nodes which is reflective of decreased germinal center activity.
  • BMS-931699 and BMS-908613-P40B showed similar binding affinities for human CD28 and similar potency in in vitro mixed lymphocyte reaction (MLR) assays.
  • MLR mixed lymphocyte reaction
  • BMS1m-74-14982-S60C-P40B at SC doses up to 18 mg/kg was not associated with any adverse drug-related findings.
  • BMS-908613-P40B at SC doses up to 5 mg/kg was not associated with any adverse drug-related findings including any effects on plasma cytokines.
  • BMS-908613-P40B-related effects at doses ⁇ 0.5 mg/kg were limited to reversible dose-dependent suppression of primary TDARs to KLH (IgG), when KLH was administered 24-hours postdose, an expected pharmacologic effect.
  • IV doses of BMS-931699 up to 15 mg/kg (combined sex mean AUC from time zero to 168 hours [AUC(0-168 h)] ⁇ 16,700 ⁇ g ⁇ hr/mL) or a SC dose of 3.5 mg/kg (mean AUC ⁇ 3520 ⁇ g ⁇ hr/mL) administered once weekly for 5 weeks were clinically well tolerated.
  • BMS-931699-related effects at all doses included non-adverse reductions in peripheral blood regulatory T lymphocytes (Tregs), minimal to mild decreases in cortical lymphocytes in lymph nodes, and minimal to slight macrophage and/or epithelial cell vacuolation in various tissues that was not associated with inflammation or necrotic changes or altered organ function.
  • Tregs peripheral blood regulatory T lymphocytes
  • macrophage and/or epithelial cell vacuolation in various tissues was not associated with inflammation or necrotic changes or altered organ function.
  • Reductions in Tregs and cortical lymphocytes in lymph nodes were expected pharmacologic effects, while vacuolation in various tissues was attributed to the PEG moiety of BMS-931699.
  • no-observed-adverse-effect level was considered to be 15 mg/kg/week IV mean AUC[0-168 h] of 15,200 ⁇ g ⁇ h/mL on Day 22) and 3.5 mg/kg/week SC (mean AUC[0-168 h] of 3330 ⁇ g ⁇ h/mL on Day 22).
  • BMS-931699 was clinically well tolerated by cynomolgus monkeys for 6 months when administered as weekly SC doses of ⁇ 10 mg/kg (AUC[[0-168 h] 12,100 ⁇ g ⁇ h/mL).
  • the primary BMS-931699-related findings at all doses were pharmacologically-mediated decreases in peripheral blood Tregs, B lymphocytes, serum IgG, and cortical lymphocytes in lymph nodes or spleen. Given the changes in Tregs were minimal, reversible, and not associated with any other correlative adverse findings, they were not considered adverse.
  • Lymphoma was noted in 1 female at 1 mg/kg/week that was considered secondary to BMS-931699-induced immunosuppression in cynomolgus monkeys latently infected with LCV; based on this finding a NOAEL was not determined in this study.
  • the PK of BMS1m-74-14982-S60C-P40B r and BMS-908613-P40B r were evaluated in mice and cynomolgus monkeys, respectively. After intravenous (IV) administration (0.2 mg/kg in mice; 0.05, and 5 mg/kg in monkeys), circulating BMS-1m74-14982-S60C-P40B and BMS-908613-P40B concentrations exhibited bi-exponential declines.
  • the steady-state volumes of distribution (Vss) for BMS1m-74-14982-S60C-P40B and BMS-908613-P40B in mice were (0.13 L/kg) which is greater than the reported plasma volume in mice indicating that the drug largely resides in the extracellular space.
  • the Vss (0.053 L/kg) for BMS-908613-P40B was similar to the reported plasma volume, indicating very limited extravascular distribution.
  • the serum clearance of BMS-1m74-14982-S60C-P40B in mice and plasma clearance of BMS-908613-P40B in monkeys were 3.9 mL/h/kg and 0.82 to 1.1 mL/h/kg, respectively.
  • the apparent elimination half-life (T-HALF) after IV administration was 27 hours in mice and 50 to 71 hours in monkeys.
  • BMS-1m74-14982-S60C-P40B and BMS-908613-P40B were well absorbed, with bioavailability of 78% in mice and 96 to 111% in monkeys, respectively.
  • BMS-908613-P40B exhibited a more than dose-proportional increase in exposure in female monkeys (at 0.05, 0.5, and 5 mg/kg, peak plasma concentrations [Cmax] were 0.335, 5.0, and 61.9 ⁇ g/mL, respectively, and the area under the concentration-time curve from time zero to infinity [AUC(INF)] values were 43.4, 642, and 6790 ⁇ g*h/mL, respectively).
  • BMS 1m74-14982-S60C-P40Br exhibited an approximately dose-proportional increase in exposure.
  • the Tmax was 4 to 9 hours.
  • the early clinical program for BMS-931699 consists of a First-in-Humans (FIH) single ascending dose (SAD) study (Part 1) along with a neo-antigen immunization SAD study (Part 2), followed by a multiple ascending dose (MAD) study in healthy subjects.
  • FIH First-in-Humans
  • SAD single ascending dose
  • MAD multiple ascending dose
  • a total of 156 subjects were enrolled in the FIH SAD study IM128001, including 108 subjects in Part 1 (single-ascending dose study) and 48 subjects in SAD Part 2 (KLH immunization).
  • BMS-931699 was safe and well tolerated after a single dose of up to 100 mg IV. There were no deaths.
  • the MAD study IM128003 was performed in 24 subjects (3 cohorts of 8 subjects) treated for 5 weeks with either SC doses of BMS-931699 (6.25 mg every other week [EOW], 12.5 mg weekly [QW], or 37.5 mg QW) or placebo (3:1 randomization).
  • BMS-931699 6.25 mg every other week [EOW], 12.5 mg weekly [QW], or 37.5 mg QW
  • placebo 3:1 randomization
  • preliminary results from the MAD study show no evidence of CD28 agonistic activity, as defined by clinically significant cytokine release and/or lymphocyte changes. Infections were observed in the BMS-931699-treated healthy subject cohorts (5/18; 27.8%) but no correlation was observed between exposure and infection rate.
  • the SAD study IM128001 evaluated BMS-931699 in the dose range of 0.01 mg to 100 mg (0.01, 0.05, 0.25, 3, 9, 25, 50, and 100 mg) following 30-minute IV infusion, and doses of 9, 25, and 50 mg following SC administration.
  • BMS-931699 exhibited linear PK after single IV and SC administration.
  • the Cmax and AUC(INF) of BMS-931699 administered IV and SC increased approximately in proportion to dose over the range of 3 mg to 100 mg.
  • AUC and Cmax values of BMS-931699 increased in a dose-proportional manner following single doses of 3 mg to 100 mg IV and 9 mg to 50 mg SC in healthy subjects.
  • the mean total body clearance (CLT), V Z and V SS were in the range of 0.42-0.59 L/min, 3.4-5.1 L, and 3.2-4.5 L respectively, and relatively consistent among all the dose groups following single IV administration.
  • the mean apparent total body clearance (CLT)/F and V Z /F were in the range of 0.59-0.70 L/min and 6.0-7.3 L, respectively, and relatively consistent among all the dose groups following single SC administration.
  • the T-HALF of BMS-931699 was similar following a single dose or multiple doses in healthy subjects (4 to 5.5 days for single dose and 6 to 7 days for multiple doses). Bioavailability of BMS-931699 following SC administration on AUC(INF) was 68.2%.
  • the MAD study IM128003 evaluated BMS-931699 in three treatment groups: 6.25 mg every other week (3 doses), 12.5 mg weekly (5 doses), and 37.5 mg weekly (5 doses). Following every other week and weekly SC administration, the pharmacokinetics of BMS-931699 is linear over the range of 6.25 mg every other week to 37.5 mg weekly.
  • the geometric mean of accumulation index of AUC were 1.3, 2.4 and 3 for 6.25 mg EOW, 12.5 mg QW and 37.5 mg QW, respectively.
  • BMS-931699 Four treatments of BMS-931699 are selected for initial evaluation in humans in a Phase 2 dose ranging study, including 1.25 mg every other week, 5 mg every other week, 12.5 mg every other week, and 12.5 mg weekly. These dosing regimens were selected based on the PK/PD relationship using receptor occupancy (RO) and IgG suppression following Keyhole Limpet Hemocyacin (KLH)-antigen challenge as predictive markers for immunosuppressive activity and clinical efficacy in SLE patients.
  • RO receptor occupancy
  • KLH Keyhole Limpet Hemocyacin
  • the exposures associated with the lowest proposed dose of 1.25 mg every other week dosing regimen are expected to be very low (5 times lower than the lowest dose tested in the MAD study in normal healthy volunteers) with a large projected safety margin (50 ⁇ ) from the lowest dose tested in 6-month monkey study.
  • the predicted receptor occupancy distribution for the 1.25 mg every other week is expected to fall below 80% for the entire treatment population, while this dose is expected to maintain receptor occupancy above 40% for majority of the human subjects.
  • the 5 mg every other week regimen is expected to elicit >80% receptor occupancy in approximately 50% of the patients, thereby providing some immunosuppressive activity, which is expected to induce some clinical response, albeit suboptimal.
  • AUC(TAU) exposure associated with this dosing regimen
  • the higher doses of 12.5 mg every other week and every week are expected to provide >80% and >90% receptor occupancy for the entire treatment population, and expected to elicit near maximal immunosuppressive activity potentially leading to near maximal efficacious response.
  • the exposures associated with these 2 regimens are considerably lower (5.1 ⁇ and 2.5 ⁇ ), than that of the lowest tested dose in 6-month monkey study.
  • the projected exposures of the four doses are within the range of exposures tested in the MAD study in healthy human subjects. The highest dose in this POC study will still be one third of the highest dose in the MAD study.
  • a Phase 2, parallel-arm, randomized, double-blinded, multicenter, international study, with an adaptive design is conducted.
  • the study comprises a short-term period which has two parts: Part 1 ( FIG. 1 ) and Part 2 ( FIG. 2 ), and a long-term extension (LTE) period ( FIG. 3 ).
  • Part 1 focuses on assessing safety and receptor occupancy.
  • Part 1 is limited to 30 to 50 patients (approximately 6-10 patients/arm) are included in the Safety/RO interim analysis (IA).
  • the 6 to 10 patients per arm complete 28 days (4 weeks) of treatment. These patients continue study treatment beyond the IA for Safety/RO and be treated for up to 24 weeks and be followed for 6 additional weeks after treatment is completed.
  • a total of up to 50 randomized subjects are treated for up to 6 months. Treatment could be shorter if IA for Safety/RO analysis indicates that one or more arms should be dropped.
  • Part 2 is opened once the cumulative safety profile is considered acceptable and receptor occupancy data for 6-10 subjects per cohort dosed for >28 days is available. Approximately 300 subjects are randomized into this part of the study (number of patient to be randomized into Part 2 may increase based on the results of the futility interim analysis). All subjects undergo screening evaluations to determine eligibility and allow down titration of prednisone (or prednisone equivalent) within 28 days prior to administration of their first dose of study medication.
  • the dose may be adapted as follows:
  • the long-term extension (LTE) period is optional and includes eligible subjects who have completed Day 169 (week 24) of treatment and consent to participate. This period of the study remains blinded but no longer has a placebo arm. Eligible subjects remain on their originally-assigned dose arm, unless they were on the placebo arm during the short-term period. Placebo-arm subjects are re-randomized into one of the existing active arms at Day 169 (24 weeks). Re-randomization of the placebo subjects is done by IVRS and only the unblinded pharmacist/drug preparer knows the new randomization arm. The LTE remains blinded to the study team and study personnel.
  • the approximate duration of the study is 28 days (4 weeks) of screening, 168 days (24 weeks) of treatment, and 42 days (6 weeks) of safety follow-up, for a total of approximately 238 days (34 weeks) in the short-term period. If the subject is eligible and opts to continue into the LTE, the 42 day follow-up visit is performed after the short-term period is completed and the subject enters LTE directly. If the subject opts not to enter LTE then a follow-up visit is completed 42 days after end of treatment. At the time of writing there is no defined end date to the long term extension period, however, the LTE provision may be further adjusted based on results from the ongoing lulizumab development program. Subjects discontinuing treatment during the LTE period complete the follow-up visit approximately 6 weeks after receiving their last dose of study medication.
  • Part 1 or Part 2 Subjects randomized in either Part 1 or Part 2 are treated for up to 24 weeks and have the same procedures performed and follow the same visit schedule.
  • This Example summarizes potential risks of treatment with BMS-931699 and the precautions that are required in clinical studies. This assessment is based on nonclinical data and the clinical experience to date with BMS-931699 set forth in the previous Examples.
  • Blocking the function of CD28 is expected to modulate the immune response. Modulation of immune response may predispose to infection.
  • BMS-913699 treatment resulted in bacterial or viral infection.
  • single doses and repeat doses of BMS-931699 have been administered in healthy subjects. Based on results from both single- and repeat-dose IV and SC nonclinical toxicology studies in cynomolgus monkeys and the clinical data from studies IM128001 and IM128003, BMS-931699 has demonstrated an acceptable safety profile, supporting continued development.
  • the expected exposure [AUC(TAU)] at steady state for the highest proposed dose 12.5 mg weekly is approximately 2.5 ⁇ lower than the lowest tested dose of 1 mg/kg/week in the 6 month toxicity study in monkeys where minimal to moderate vacuolation of macrophages were observed.
  • the highest dose in this Phase 2 study is one third of the highest dose in the MAD study. Intense safety monitoring is put in place during Part 1 of Phase 2, allowing early detection of any safety signals.
  • Phase 2 study initiates with the 4 regimens of 1.25 mg every other week, 5 mg every other week, 12.5 mg every other week, and 12.5 mg weekly.
  • Safety/receptor occupancy interim analysis is performed to ensure the exposures and receptor occupancy observations approximate the original predictions.
  • IA interim analysis for safety and receptor occupancy is performed in Part 1 of the study, when at least 6 patients per treatment arm have reached Study Day 29. That receptor occupancy observations approximate the original predictions and BMS-931699 is well tolerated in SLE patients is confirmed. The rest of the patients are enrolled in the study.
  • dosing regimens originally included in Part 1 may be discontinued and/or new dosing regimens may be added according to the criteria outlined below. Enrollment in Part 2 of the study is opened after the IA has resulted in a decision regarding the dosing regimens to be carried forward:
  • the safety analysis focuses on incidence and severity of all adverse events (AEs), serious AEs and pre-established Events of Special Interest such as infection AEs and any other safety analysis requested by DMC.
  • AEs adverse events
  • serious AEs and pre-established Events of Special Interest
  • the DMC in conjunction with an unblinded internal safety monitoring team may require one or more doses to be discontinued if stopping criteria are met or other safety signals arise that the Medical Monitor and/or DMC consider of sufficient concern.
  • the unblinded safety monitoring team is not involved in the regular study activities.
  • the median receptor occupancy at Day 29 for each treatment arm is calculated.
  • Dose decrease and/or reduction of frequency of administration is considered if receptor occupancy results fall outside the parameters indicated above. This adjustment occurs for safety reasons or in case unforeseen receptor occupancy profiles are observed in SLE patients.
  • the decision to adjust dose and/or frequency is taken after review of the data by the clinical team. Subjects do not change dose. If an arm is modified or removed, subjects randomized to that arm and discontinued.
  • an interim analysis for futility and dose adaption occurs after 30 patients per treatment arm (including patients from Part 1) have completed at least 85 days of treatment period or discontinued.
  • One or more treatment arms may be dropped for lack of efficacy, or a treatment arm may be added to explore a suboptimal dose.
  • Exposure-response analysis for efficacy and safety may be conducted in parallel with this interim analysis to facilitate the dose selection for this additional lower treatment arm.
  • the long-term extension period has interim analyses for dose adaptation. If a significant safety concern is identified, one or more dose arms may be dropped. If one or more dose arms are dropped for safety reasons, all subjects currently receiving that or those dose(s) are discontinued from receiving study medication.
  • subjects Unless intolerant, subjects must be currently receiving at least one of the following steroid-sparing agents for a minimum of 12 weeks, and a stable dose for at least 56 days (8 weeks) prior to signing consent: azathioprine (AZA), chloroquine, hydroxychloroquine, methotrexate (MTX), leflunomide, mycophenolate mofetil/mycophenolic acid. Subjects must remain on stable dose throughout the study.
  • azathioprine AZA
  • chloroquine chloroquine
  • hydroxychloroquine hydroxychloroquine
  • methotrexate (MTX) methotrexate
  • leflunomide mycophenolate mofetil/mycophenolic acid
  • Prednisone (or prednisone-equivalent) is not required; however, if subject is taking prednisone (or prednisone equivalent), the dose cannot exceed 30 mg/day at screening for a subject to be eligible and must be stable at a maximum of 10 mg/day for at least 5 days prior to Day 1 (randomization). Any other immunossuppressive or biologic drug requires washout periods prior to study entry. If subjects are receiving non-steroidal anti-inflammatory drugs (NSAIDs) (including marketed COX-2 inhibitors), doses must be stable for 14 days prior to first dose of study medication on Day 1 (randomization) and subject must remain on the same dose throughout the study. Note: NSAIDS should be withheld for at least 12 hours prior to visits where BILAG, SLEDAI 2K, joint counts, Cutaneous Lupus Erthematosus Disease Area and Severity Index (CLASI) and MDGA is assessed.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • Female patients of childbearing potential have a negative serum or urine pregnancy test (minimum sensitivity 25 IU/L or equivalent units of human chorionic gonadotropin) within 24 hours prior to the start of study drug administration.
  • Female patients are not breastfeeding.
  • Female patients of childbearing potential must use contraception for the duration of treatment with study drug plus 5 half-lives of study drug (7 days) plus 30 days (duration of ovulatory cycle) for a total of 65 days post-treatment completion.
  • Males who are sexually active with female patients of childbearing potential must use contraception for the duration of treatment with study drug plus 5 half-lives of the study drug (7 days) plus 90 days (duration of sperm turnover) for a total of 125 days post-treatment completion.
  • Azoospermic males and women of child bearing potential who are continuously not heterosexually active are exempt from contraceptive requirements. However they must still undergo pregnancy testing.
  • RA rheumatoid arthritis
  • MS multiple sclerosis
  • Subjects with type 1 diabetes mellitus, thyroid autoimmune disease and secondary Sjögren syndrome are eligible.
  • Subjects with primary anti-phospholipid antibody syndrome as the sole or primary feature of their SLE or SLE-like syndrome are excluded.
  • subjects with secondary anti-phospholipid syndrome are included in the study, unless they have had a serious thrombotic event (e.g. pulmonary embolism, stroke, or deep vein thrombosis) within one year prior to signing consent.
  • Subjects on chronic coumadin or enoxaparin can be enrolled in the study.
  • Subjects with active, severe, lupus nephritis (WHO class III, IV) which requires or may require treatment with cytotoxic agents or high dose corticosteroids.
  • Subjects with prior, controlled renal disease with residual proteinuria up to 3 g/day or a urine protein/creatinine ratio of 3 mg/mg or 339 mg/mmol are allowed.
  • HIV Human Immunodeficiency Virus
  • WBC White blood cells
  • BMS-931699 or a look alike placebo is administered weekly as a solution subcutaneously (SC) single injection, dependent on the dosage panel.
  • the clinical label reflects the product name as “BMS-931699-01” to be linked with the product description on the vial.
  • the composition of the BMS-931699-01 injection is 12.5 mg/mL BMS-931699 in 20 mM phosphate, pH 5.9, with 5% (w/v) sorbitol.
  • the BMS-931699-01 injection is packaged in a 3 cc vial with a 13 mm opening, 1-panel, open label.
  • the BMS-931699-1 injection appears clear to slightly opalescent, colorless to pale yellow solution.
  • the BMS-931699-01 injection is stored refrigerated 2-8° C. (36-46° F.).
  • Table 4 below indicates the total dose and number of vials per dose for each dosage panel.
  • SC subcutaneous
  • no dilution of the drug product solution (12.5 mg/mL) is required for doses of 12.5 mg.
  • doses of 5 mg and 1.25 mg require dilution.
  • a 21-gauge, 1.5 inch (3.8 cm) sterile needle is used for withdrawal of this product from the vial, and a 27 gauge, 0.5 inch (1.3 cm) sterile needle is used for SC dosing.
  • a conventional, commercially available polycarbonate syringe of appropriate size is used for withdrawal and administration. After withdrawal into an appropriate sized syringe, the product is administered within 4 hours. If not dosed immediately, filled syringes are kept at 2°-8° C. (36°-46° F.) with protection from light prior to use.
  • the placebo for BMS-931699 injection is a normal saline solution, which is administered in a similar fashion as described for the BMS-931699 injection. Study personnel administer the dose to the subject.
  • Every randomized subject is required to come to the clinic/research center weekly to be dosed. This ensures double-blind is maintained despite the variability of regimens.
  • Subjects randomized to weekly subcutaneous injections of either placebo or BMS-931699 are dosed weekly as per schedule and subjects randomized to one of the every other week arm are alternating between receiving a subcutaneous injection of BMS-931699 one week and one of placebo the following week.
  • Pharmacodynamic, target engagement and disease related biomarker assays are incorporated into the study inform dose selection, monitor efficacy and potentially predict treatment response.
  • Blood and urine are drawn for the measurement of markers of target engagement and phamacodynamic effects of BMS-931699 including CD28 receptor occupancy, C3, C4, and auto-antibodies.
  • Target engagement as assessed by CD28 receptor occupancy on T cells, is incorporated into the study to inform dose selection for the Phase 3 study, monitor efficacy and potentially predict treatment response.
  • the relationship between the concentration levels of BMS-931699 and the CD28 receptor occupancy is characterized.
  • biomarkers include: other cytokines and chemokines, anti-double-stranded deoxyribonucleic acid (anti-dsDNA), anti-nuclear antibody (anti-ANA), anti-Ro (otherwise known as anti-SSA, anti-SSA/RO, or anti-Ro/SSA) autoantibodies, anti-Lupus (anti-La) (otherwise known as anti-Sjögren syndrome type B antigen (anti-SS-B)) autoantibodies, anti-ribonuclear protein (anti-RNP) autoantibodies, anti-Sm nuclear antigen autoantibodies, anti-APL autoantibodies, and other autoantibodies.
  • anti-dsDNA anti-double-stranded deoxyribonucleic acid
  • anti-ANA anti-nuclear antibody
  • anti-Ro anti-Ro
  • anti-La anti-Sjögren syndrome type B antigen
  • anti-RNP anti-ribonuclear protein
  • C-reactive protein C-reactive protein
  • IgG total immunoglobulin G
  • IgM total immunoglobulin M
  • RNA transcripts in whole blood proteins in urine (NGAL, TWEAK, MCP-1, IL-18, IL-1), total soluble CD28, T cell activation, leukocyte phenotypes in peripheral blood mononuclear cells (PMBCs) and whole blood (surface CD4, surface CD8, surface CD28, surface CD57 and intracellular granzyme B), soluble inflammatory mediators (serum IL-6, IL-18, TNF- ⁇ , ⁇ -interferon, BLyS(BAFF), CD154, sCD28 and other soluble receptors, microvessicles) can also serve as biomarkers.
  • PMBCs peripheral blood mononuclear cells
  • soluble inflammatory mediators serum IL-6, IL-18, TNF- ⁇ , ⁇ -interferon,
  • RNA sample is collected in PAX gene tubes at times indicated. These samples provide broad genomic profiling to search for novel pharmacodynamic and efficacy biomarkers related to inflammatory and/or autoimmune pathways. Furthermore, these samples are used to search for gene expressions at baseline that may be predictive of efficacy for BMS-931699 treated subjects.
  • T cells may be characterized for activation and for subpopulations. Markers may include combinations of, but not limited to, surface CD4, CD8, CD28, CD57, and intracellular granzyme B. Other peripheral cells, including B cells, monocytes/macrophags, dendritic cells, and NK cells may be analyzed.
  • Urine is collected and analyzed for markers of SLE and other inflammatory disorders.
  • the samples are analyzed for proteomic profiles of inflammatory markers (including but not limited to IL-18, IL-1, NGAL, uTWEAK, MCP-1). Exploratory analysis is carried out to identify biomarkers to monitor PD and response to therapy in patients with kidney involvement.
  • Serum and plasma is collected for the measurement of soluble inflammatory mediators associated with inflammation, SLE or co-stimulation blockade (including, but not limited to, serum IL-6, IL-18, TNF- ⁇ , ⁇ -interferon, BLyS(BAFF), CD154, sCD28 and other soluble receptors, microvessicles).
  • SLE soluble inflammatory mediators associated with inflammation
  • co-stimulation blockade including, but not limited to, serum IL-6, IL-18, TNF- ⁇ , ⁇ -interferon, BLyS(BAFF), CD154, sCD28 and other soluble receptors, microvessicles.
  • the SF-36 is used to measure health-related quality of life. Individual subscale scores and two summary scores are calculated: (1) physical component summary (PCS) which includes physical functioning, role-physical, bodily pain, and general health; (2) mental component summary (MCS) which includes vitality, social functioning, roleemotional, and mental health.
  • PCS physical component summary
  • MCS mental component summary
  • the SF-36 is a widely recognized tool that is recognized by the FDA as a validated instrument to measure health-related quality of life across multiple disease states.
  • FACIT-F is a health related quality of life questionnaire focused on Fatigue.
  • FACIT-F includes the following components; physical well-being, social/family well-being, emotional well-being, functional well-being and additional concerns.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Transplantation (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
US15/515,461 2014-09-30 2015-09-30 Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28 Abandoned US20170240636A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/515,461 US20170240636A1 (en) 2014-09-30 2015-09-30 Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462057981P 2014-09-30 2014-09-30
US15/515,461 US20170240636A1 (en) 2014-09-30 2015-09-30 Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28
PCT/US2015/053233 WO2016054218A1 (en) 2014-09-30 2015-09-30 Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/053233 A-371-Of-International WO2016054218A1 (en) 2014-09-30 2015-09-30 Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/842,885 Continuation US11267888B2 (en) 2014-09-30 2020-04-08 Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28

Publications (1)

Publication Number Publication Date
US20170240636A1 true US20170240636A1 (en) 2017-08-24

Family

ID=54291723

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/515,461 Abandoned US20170240636A1 (en) 2014-09-30 2015-09-30 Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28
US16/842,885 Active US11267888B2 (en) 2014-09-30 2020-04-08 Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28
US17/584,207 Active US11912769B2 (en) 2014-09-30 2022-01-25 Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/842,885 Active US11267888B2 (en) 2014-09-30 2020-04-08 Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28
US17/584,207 Active US11912769B2 (en) 2014-09-30 2022-01-25 Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28

Country Status (14)

Country Link
US (3) US20170240636A1 (es)
EP (1) EP3201231B1 (es)
JP (1) JP2017535524A (es)
KR (1) KR20170062505A (es)
CN (1) CN106794255A (es)
AU (1) AU2015325015A1 (es)
BR (1) BR112017006520A2 (es)
CA (1) CA2963125A1 (es)
EA (1) EA201790475A1 (es)
ES (1) ES2783448T3 (es)
IL (1) IL251364A0 (es)
MX (1) MX2017003691A (es)
SG (1) SG11201702383SA (es)
WO (1) WO2016054218A1 (es)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150299321A1 (en) * 2008-07-18 2015-10-22 Bristol-Myers Squibb Company Compositions monovalent for cd28 binding and methods of use
WO2018183929A1 (en) 2017-03-30 2018-10-04 Progenity Inc. Treatment of a disease of the gastrointestinal tract with an immune modulatory agent released using an ingestible device
WO2019246312A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an immunomodulator
WO2019246317A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease or condition in a tissue originating from the endoderm
WO2020106757A1 (en) 2018-11-19 2020-05-28 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
US11267888B2 (en) 2014-09-30 2022-03-08 Bristol-Myers Squibb Company Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28
EP4252629A2 (en) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190086405A1 (en) * 2016-03-16 2019-03-21 Bristol-Myers Squibb Company Methods of diagnosing and treating lupus
JP2022525332A (ja) * 2019-03-14 2022-05-12 ビオンド バイオロジクス リミテッド 免疫抑制の方法
US20230322996A1 (en) * 2021-11-08 2023-10-12 Life Technologies As Polymer-interaction molecule conjugates and methods of use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100028354A1 (en) * 2008-07-18 2010-02-04 Bristol-Myers Squibb Company Compositions monovalent for cd28 binding and methods of use

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014120916A1 (en) * 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
US20170051059A1 (en) * 2014-03-19 2017-02-23 Bristol-Myers Squibb Company Methods of treating transplant rejection using a domain antibody directed against cd40
SG11201702383SA (en) 2014-09-30 2017-04-27 Bristol Myers Squibb Co Methods of treating systemic lupus erythematosus using a domain antibody directed against cd28

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100028354A1 (en) * 2008-07-18 2010-02-04 Bristol-Myers Squibb Company Compositions monovalent for cd28 binding and methods of use

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10919965B2 (en) 2008-07-18 2021-02-16 Bristol-Myers Squibb Company Compositions monovalent for CD28 binding and methods of use
US9908937B2 (en) * 2008-07-18 2018-03-06 Bristol-Myers Squibb Company Compositions monovalent for CD28 binding and methods of use
US20150299321A1 (en) * 2008-07-18 2015-10-22 Bristol-Myers Squibb Company Compositions monovalent for cd28 binding and methods of use
US11912769B2 (en) 2014-09-30 2024-02-27 Bristol-Myers Squibb Company Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28
US11267888B2 (en) 2014-09-30 2022-03-08 Bristol-Myers Squibb Company Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28
EP4252629A2 (en) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems
WO2018183929A1 (en) 2017-03-30 2018-10-04 Progenity Inc. Treatment of a disease of the gastrointestinal tract with an immune modulatory agent released using an ingestible device
EP4108183A1 (en) 2017-03-30 2022-12-28 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with an immune modulatory agent released using an ingestible device
WO2019246317A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease or condition in a tissue originating from the endoderm
WO2019246312A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an immunomodulator
WO2020106704A2 (en) 2018-11-19 2020-05-28 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
WO2020106750A1 (en) 2018-11-19 2020-05-28 Progenity, Inc. Methods and devices for treating a disease with biotherapeutics
WO2020106754A1 (en) 2018-11-19 2020-05-28 Progenity, Inc. Methods and devices for treating a disease with biotherapeutics
WO2020106757A1 (en) 2018-11-19 2020-05-28 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
EP4309722A2 (en) 2019-12-13 2024-01-24 Biora Therapeutics, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract

Also Published As

Publication number Publication date
KR20170062505A (ko) 2017-06-07
SG11201702383SA (en) 2017-04-27
EP3201231B1 (en) 2020-03-04
ES2783448T3 (es) 2020-09-17
US11912769B2 (en) 2024-02-27
IL251364A0 (en) 2017-05-29
MX2017003691A (es) 2017-06-26
AU2015325015A1 (en) 2017-05-18
EA201790475A1 (ru) 2017-08-31
US11267888B2 (en) 2022-03-08
US20220144951A1 (en) 2022-05-12
BR112017006520A2 (pt) 2017-12-19
WO2016054218A1 (en) 2016-04-07
EP3201231A1 (en) 2017-08-09
CN106794255A (zh) 2017-05-31
CA2963125A1 (en) 2016-04-07
US20200247890A1 (en) 2020-08-06
JP2017535524A (ja) 2017-11-30

Similar Documents

Publication Publication Date Title
US11912769B2 (en) Methods of treating systemic lupus erythematosus using a domain antibody directed against CD28
EP3348569B1 (en) Pan elr+ cxc chemokine antibodies
JP2021113190A (ja) 移植片拒絶の予防に使用するための抗cd40抗体
CN115996748A (zh) 用APRIL结合抗体治疗IgA肾病的方法
US20220259308A1 (en) Fcrn antibodies and methods of use thereof
US20230020999A1 (en) Anti-cd30 antibody-drug conjugates and their use for the treatment of hiv infection
US20240076394A1 (en) Modulating the immune response using anti-cd30 antibody-drug conjugates
CA3148495A1 (en) Use of calcineurin inhibitor free ctla4-ig + anti-il6/il6r for long term immunosuppression in solid organ transplant recipients
TW202302146A (zh) 使用抗baffr抗體治療全身性紅斑狼瘡

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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