Modulating gamma-c-cytokine activity

Information

  • Patent Grant
  • 11400134
  • Patent Number
    11,400,134
  • Date Filed
    Thursday, October 6, 2016
    7 years ago
  • Date Issued
    Tuesday, August 2, 2022
    a year ago
  • Inventors
  • Original Assignees
  • Examiners
    • Popa; Ileana
    Agents
    • Knobbe, Martens, Olson & Bear LLP
Abstract
Embodiments relate to peptide antagonists of γc-family cytokines, which is associated with important human diseases, such as leukemia, autoimmune diseases, collagen diseases, diabetes mellitus, skin diseases, degenerative neuronal diseases and graft-versus-host disease (GvHD). Thus, inhibitors of γc-cytokine activity are valuable therapeutic and cosmetic agents as well as research tools. Traditional approaches to inhibiting yc-cytokine activity involve raising neutralizing antibodies against each individual γc-cytokine family member/′ receptor subunit. However, success has been limited and often multiple γc-cytokine family members co-operate to cause the disease state. Combinatorial use of neutralizing antibodies raised against each factor is impractical and poses an increased risk of adverse immune reactions. The present embodiments overcome these shortcomings by utilizing peptide antagonists based on the consensus γc-subunit binding site to inhibit γc-cytokine activity. Such approach allows for flexibility in antagonist design. In several embodiments, peptides exhibit Simul-Block activity, inhibiting the activity of multiple γc-cytokine family members.
Description
SEQUENCE LISTING IN ELECTRONIC FORMAT

The present application is being filed along with a Sequence Listing as an ASCII text file via EFS-Web. The Sequence Listing is provided as a file entitled BION006WOSEQLIST.txt, created and last saved on Oct. 6, 2016, which is 5,981 bytes in size. The information in the electronic format of the Sequence Listing is incorporated herein by reference in its entirety in accordance with 35 U.S.C. § 1.52(e).


FIELD

Some embodiments relate to peptide antagonists of γc-family cytokines, a group of mammalian cytokines that are mainly produced by epithelial, stromal and immune cells and control the normal and pathological activation of a diverse array of lymphocytes. Some embodiments also relate to the therapeutic uses of such peptides for the treatment of certain human diseases. The present embodiments also relate to the cosmeceutical applications of such peptides. Description of target diseases, cosmeceutical applications, as well as methods of administration, production, and commercialization of the peptides are disclosed.


BACKGROUND

Cytokines are a diverse group of soluble factors that mediate various cell functions, such as, growth, functional differentiation, and promotion or prevention of programmed cell death (apoptotic cell death). Cytokines, unlike hormones, are not produced by specialized glandular tissues, but can be produced by a wide variety of cell types, such as epithelial, stromal or immune cells.


The γc-family cytokines are a group of mammalian cytokines that are mainly produced by epithelial, stromal and immune cells and control the normal and pathological activation of a diverse array of lymphocytes. These cytokines are critically required for the early development of T cells in the thymus as well as their homeostasis in the periphery.


SUMMARY

In some embodiments, a composite peptide is provided. In some embodiments, the composite peptide comprises amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).


In some embodiments, the composite peptide of claim 1, wherein the composite peptide inhibits the activity of two or more γc-cytokines selected from the group consisting of IL 2, IL 4, IL 7, IL 9, IL 15, and IL 21. In some embodiments, the composite peptide inhibits the activity of at least IL-15 and IL-21.


In some embodiments, the composite peptide further comprises a signal peptide, in some embodiments, the composite peptide is further conjugated to one or more additional moieties at the N terminus, C terminus or a side residue of the composite peptide. In some embodiments of the composite peptide, the one or more additional moieties are selected from the group consisting of bovine serum albumin (BSA), albumin, Keyhold Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell-specific antigen, a receptor, a ligand, a metal ion, and Poly Ethylene Glycol (PEG).


In some embodiments, the composite peptide consists of an amino acid sequence having at least 90% sequence identity with SEQ ID NO: 3. In some embodiments, the composite peptide consists of an amino acid sequence having at least 95% sequence identity with SEQ ID NO: 3.


In some embodiments, a pharmaceutical composition is provided. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a peptide conjugate, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the peptide conjugate or the derivative thereof modulates the activity of two or more γc-cytokines selected from the group consisting of IL 2, IL 4, IL 7, IL 9, IL 15, and IL 21, wherein the peptide conjugate comprises the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).


In some embodiments of the pharmaceutical composition, the peptide conjugate or the derivative thereof inhibits the activity of two or more γc-cytokines selected from the group consisting of IL 2, IL 4, IL 7, IL 9, IL 15, and IL 21.


In some embodiments of the pharmaceutical composition, the peptide conjugate or the derivative thereof further comprises an additional conjugate at the N termini, C termini or a side residues thereof. In some embodiments of the pharmaceutical composition, the peptide conjugate or the derivative thereof further comprises a signal peptide.


In some embodiments, the pharmaceutical composition further comprises a protein that stabilizes the structure of the peptide conjugate or the derivative thereof and improves its biological activity, wherein the protein is selected from the group consisting of bovine serum albumin (BSA), albumin, Fc region of immunoglobulin G (IgG), biological proteins that function as scaffold, Poly Ethylene Glycol (PEG), and derivatives thereof.


In some embodiments of the pharmaceutical composition, the derivative thereof comprises a peptide sequence sharing at least 90% identity with the amino acid sequence of SEQ ID NO: 3. In some embodiments of the pharmaceutical composition, the derivative thereof comprises a peptide sequence sharing at least 95% identity with the amino acid sequence of SEQ ID NO: 3.


In some embodiments, a method for blocking signaling by two or more γc cytokine family members is provided. In some embodiments, the method comprises contacting a cell with a composite peptide and/or a pharmaceutical composition provided herein.


In some embodiments, a method of inhibiting γc cytokine binding to a γc subunit is provided. In some embodiments, the method comprises contacting a γc subunit of a cell with a composite peptide and/or a pharmaceutical composition provided herein.


In some embodiments, a method of treating a γc cytokine-mediated disease is provided. In some embodiments, the method comprises administering a composite peptide and/or a pharmaceutical composition provided herein to a subject in need thereof, wherein the γc cytokine-mediated disease is selected from the group consisting of CD4 leukemia, CD8 leukemia, LGL leukemia, systemic lupus erythematosis, Sjögren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, and graft-versus-host disease (GvHD).


In some embodiments, a method of treating an HTLV-1-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease is provided. In some embodiments, the method comprising administering a composite peptide and/or a pharmaceutical composition provided herein to a subject in need thereof, wherein the HAM/TSP associated disease is selected from the group consisting of Adult T-cell Leukemia (ATL), HTLV-associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, and myositis.


In some embodiments, a method of treating an inflammatory respiratory disease is provided. In some embodiments, the method comprising administering a composite peptide and/or a pharmaceutical composition provided herein to a subject in need thereof, wherein the inflammatory respiratory disease is selected from the group consisting of asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis.


In some embodiments, a method of treating a cosmetic condition is provided. In some embodiments, the method comprising administering a composite peptide and/or a pharmaceutical composition provided herein to a subject in need thereof, wherein the cosmetic disease is selected from the group consisting of acne, hair loss, sunburn, nail maintenance, and appearance of aging.


In some embodiments, a pharmaceutical composition for use in treating a condition in a patient is provided. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a peptide conjugate, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the peptide conjugate or the derivative thereof modulates the activity of two or more γc-cytokines selected from the group consisting of IL 2, IL 4, IL 7, IL 9, IL 15, and IL 21, wherein the peptide conjugate comprises the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).


In some embodiments of the pharmaceutical composition, the derivative thereof comprises a peptide sequence sharing at least 90% identity with the amino acid sequence of SEQ ID NO: 3. In some embodiments of the pharmaceutical composition, the derivative thereof comprises a peptide sequence sharing at least 95% identity with the amino acid sequence of SEQ ID NO: 3.


In some embodiments, a pharmaceutical composition is provided for use in treating a γc cytokine-mediated disease, wherein the γc cytokine-mediated disease is selected from the group consisting of CD4 leukemia, CD8 leukemia, LGL leukemia, systemic lupus erythematosis, Sjögren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, and graft-versus-host disease (GvHD).


In some embodiments, a pharmaceutical composition is provided for use in treating an HTLV-1-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease, wherein the HAM/TSP associated disease is selected from the group consisting of Adult T-cell Leukemia (ATL), HTLV-associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, and myositis.


In some embodiments, a pharmaceutical composition is provided for use in treating an inflammatory respiratory disease, wherein the inflammatory respiratory disease is selected from the group consisting of asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis.


In some embodiments, a pharmaceutical composition is provided for use in treating a cosmetic condition, wherein the cosmetic disease is selected from the group consisting of acne, hair loss, sunburn, nail maintenance, and appearance of aging.


In some embodiments, a kit for treating a condition in a patient is provided. In some embodiments of the kit, the condition is a γc cytokine-mediated disease, an HTLV-1-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease, an inflammatory respiratory disease, a cosmetic condition, or a combination thereof, the kit comprising a pharmaceutical composition provided herein.


In some embodiments of the kit, the pharmaceutical composition comprises a therapeutically effective amount of a peptide conjugate, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the peptide conjugate or the derivative thereof modulates the activity of two or more γc-cytokines selected from the group consisting of IL 2, IL 4, IL 7, IL 9, IL 15, and IL 21, wherein the peptide conjugate comprises the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).


In some embodiments of the kit, the derivative thereof comprises a peptide sequence sharing at least 90% identity with the amino acid sequence of SEQ ID NO: 3. In some embodiments of the kit, the derivative thereof comprises a peptide sequence sharing at least 95% identity with the amino acid sequence of SEQ ID NO: 3.


In some embodiments of the kit, the condition is one or more of CD4 leukemia, CD8 leukemia, LGL leukemia, systemic lupus erythematosis, Sjögren's Sjoegren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, graft-versus-host disease (GvHD), Adult T-cell Leukemia (ATL), HTLV-associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, myositis, asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis, acne, hair loss, sunburn, nail maintenance, or appearance of aging.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A shows an alignment of the D-helix region of human γc-cytokine family members.



FIG. 1B depicts the γc-box (SEQ ID NO: 10) and IL-2/IL-15 box (SEQ ID NO: 11) motifs which give rise to the consensus sequence around the D-helix region of the γc-cytokines.



FIG. 2 depicts a diagramed representation of the biochemical properties of amino acids.



FIG. 3A shows inhibition of IL-15 and IL-9 activity by BNZ-γ in a PT-18 proliferation assay.



FIG. 3B shows a proliferation assay of CTTL2 cells grown in the presence of IL-2 or IL-15 and 0, 0.1, 1 or 10 uM BNZ-γ.



FIG. 3C shows inhibition of IL-15-mediated tyrosine-phosphorylation of STAT5 by BNZ-γ.



FIG. 4A shows an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood. T-cell proliferation is inhibited by addition of BNZ-γ.



FIG. 4B shows the population of CD4+CD25+ cells in an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood is diminished after adding BNZ-γ to the culture.



FIG. 4C shows the population of CD4+Ki67 cells in an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood is reduced after adding BNZ-γ to the culture.



FIG. 4D shows the percent of live cells by Guava staining in an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood is not impacted after adding BNZ-γ to the culture.



FIG. 5 shows the alignment of the sequence of SEQ ID NO: 3 to the D-helix regions of different human γc-cytokine family members. The shaded areas represent amino acid sequences of the human γc-cytokine family members that are identical to their corresponding amino acids in the sequence of SEQ ID NO: 3.





DETAILED DESCRIPTION

Overview


More than 100 cytokines have been identified so far and are considered to have developed by means of gene duplications from a pool of primordial genes (See Bazan. J. F. 1990, Immunol. Today 11:350-354). In support of this view, it is common for a group of cytokines to share a component in their multi-subunit receptor system. The most well-documented shared cytokine subunit in T cells is the common γ subunit (γc-subunit).


The γc-subunit is shared by 6 known cytokines (Interleukin-2 (IL-2), Interleukin-4 (IL-4), Interleukin-7 (IL-7), Interleukin-9 (IL-9), Interleukin-15 (IL-15), and Interleukin-21 (IL-21), collectively called the “γc-cytokines” or “γc-family cytokines”) and plays an indispensable role in transducing cell activation signals for all these cytokines. Additionally, for each of the γc-cytokines, there are one or two private cytokine-specific receptor subunits that when complexed with the γc-subunit, give rise to a fully functional receptor. (See Rochman et al., 2009, Nat Rev Immunol. 9: 480-90.)


The γc-family cytokines are a group of mammalian cytokines that are mainly produced by epithelial, stromal and immune cells and control the normal and pathological activation of a diverse array of lymphocytes. These cytokines are critically required for the early development of T cells in the thymus as well as their homeostasis in the periphery. For example, in the absence of the γc-subunit, T, B and NK cells do not develop in mice. (See Sugamura et al., 1996, Annu. Rev. Immunol. 14:179-205).


The γc-cytokines are important players in the development of the lymphoid cells that constitute the immune system, particularly T, B, and NK cells. Further, γc-cytokines have been implicated in various human diseases. Thus, factors that inhibit γc-cytokine activity would provide useful tools to elucidate the developmental mechanism of subsets of lymphocytes and to treat immune disorders and γc-cytokine-mediated diseases.


Germ line depletion of the genes encoding the γc-subunit in mice or mutations of γc-subunit in humans are known to cause severe combined immunodeficiency (SCID) by disrupting the normal appearance or function of NK, T, and B cells. The importance of the γc-subunit in the signal transduction of the γc-cytokines, IL-2, -4, -7, -9, 15, -21, is indicated in studies demonstrating the a of response of lymphocytes from these mice and human patients to the γc-cytokines (reviewed in Sugamura et al., 1995 Adv. Immunol. 59:225-277). This indicates that disruption of the interaction between the γc-subunit and a γc-cytokine would efficiently block the intracellular signaling events by the γc-cytokine family members. Therefore antagonist peptides according to the present embodiments are expected to effectively block the pathogenic changes in humans suffering from the diseases mediated by misregulation of the γc-cytokine family members.


As an alternative to antibody-mediated approaches for modulating the activity of individual γc-cytokines, Applicants have devised novel, low molecular weight compounds herein referred to as “Simul-Block”, which suppress the activity of multiple γc-cytokines. These low molecular weight compounds, which include both chemicals and peptides, are less immunogenic than antibodies. These properties distinguish Simul-Block as a more efficient strategy for mediating γc-cytokine activity in clinical interventions.


Pathologies Associated with the γc-Cytokines


Recent studies have indicated that dysregulation of expression and dysfunction of the γc-cytokines could lead to a wide variety of human immunologic and hematopoietic diseases.


IL-2


While IL-2 was historically considered a prototype T cell growth factor, the generation of a knockout mouse lacking IL-2 expression revealed that IL-2 is not critical for the growth or developmental of conventional T cells in vivo. Over-expression of IL-2, however, leads to a preferential expansion of a subset of T-cells; the regulatory T cells (T-regs). (See Antony et al., 2006, J. Immunol. 176:5255-66.) T-regs suppress the immune responses of other cells and thus act to maintain peripheral tolerance (reviewed in Sakaguchi et al., 2008, Cell 133:775-87). Breakdown of peripheral tolerance is thought to cause autoimmune diseases in humans.


Thus, the immunosuppressive function of T-regs is thought to prevent the development of autoimmune diseases (See Sakaguchi et al., 2008, Cell 133:775-87). T-regs have also been implicated in cancer, where solid tumors and hematologic malignancies have been associated with elevated numbers of T-regs (See Dc Rezende et al., 2010, Arch. Immunol. Ther. Exp. 58:179-190).


IL-4


IL-4 is a non-redundant cytokine involved in the differentiation of T helper cells into the Th2 (T-helper type 2) subset, which promotes the differentiation of premature B cells into IgE producing plasma cells. IgE levels are elevated in allergic asthma. Thus, IL-4 is implicated in the development of allergic Asthma. Antibodies targeting IL-4 can be used to treat or even prevent the onset of allergic asthma. (See Le Buanec et al., 2007, Vaccine 25:7206-16.)


IL-7


IL-7 is essential for B cell development and the early development of T cells in the thymus. In mice, the abnormal expression of IL-7 causes T-cell-associated leukemia. (See Fisher et al., 1993, Leukemia 2:S66-68.) However, in humans, misregulation of IL-7 does not appear to cause T-cell-associated leukemia. In humans, up-regulation of IL-7 either alone or in combination with another γc-cytokine family member, IL-15, has been implicated in Large Granular Lymphocyte (LGL) leukemia.


IL-9


The role of IL-9 is still rather uncharacterized compared to other γc-cytokine family members. Mice depleted of the IL-9 gene appear normal and do not lack any subsets of cells in the lymphoid and hematopoietic compartments. Recent studies, however, reveal an in vivo role for IL-9 in the generation of Th17 (T-helper induced by interleukin-17) cells (See Littman et al., 2010, Cell 140(6):845-58; and Nowak et al., 2009, J. Exp. Med. 206: 1653-60).


IL-15


IL-15 is critically involved in the development of NK cells, NK-T cells, some subsets of intraepithelial lymphocytes (IELs), γδ-T cells, and memory-phenotype CD8 T-cells (See Waldmann, 2007, J. Clin. Immunol. 27:1-18; and Tagaya et al., 1996, EMBO J. 15:4928-39.) Over-expression of IL-15 in mice leads to the development of NK-T cell and CD8 cell type T cell leukemia (See Fehniger et al., 2001, J. Exp. Med. 193:219-31: Sato et al. 2011 Blood in press). These experimentally induced leukemias appear similar to LGL (large-granular lymphocyte) leukemia in humans, since in both instances the leukemic cells express CD8 antigen.


It is also suspected that IL-15-mediated autocrine mechanisms may be involved in the leukemic transformation of CD4 T lymphocytes. (See Azimi et al., 1998, Proc. Natl. Acad. Sci. 95:2452-7; Azimi et al., 1999, J. Immunol. 163:4064-72; Azimi et al., 2000, AIDS Res. Hum. Retroviruses 16:1717-22; and Azimi et al., 2001, Proc. Natl. Acad. Sci. 98:14559-64). For example, CD4-tropic HTLV-I, which causes Adult T cell leukemia in humans, induces autocrine growth of virus-transformed T cells through the production of IL-15 and IL-15Rα (Azimi et al., 1998, Proc. Natl. Acad. Sci. 95:2452-7).


In addition to leukemic transformation, recent studies implicate IL-15 in the pathological development of Celiac disease (CD), an autoimmune disease. IL-15 is known to stimulate the differentiation of NK, CD8 and intestinal intraepithelial lymphocyte (IEL) cells into lymphokine-activated killer (LAK) cells by inducing the expression of cytolytic enzymes (i.e., Granzyme and Perforin) as well as interferon-γ. Celiac Disease (denoted CD from herein) is an immune-mediated enteropathy that is triggered by the consumption of gluten-containing food in individuals that express specific HLA-DQ alleles.


The prevalence of this disease is 1% in the western population. The only current treatment for CD is the complete elimination of gluten from the patient's diet. The pathology of CD is mainly caused by extensive damage to the intestinal mucosa, which is caused by activated CD8 T cells that have infiltrated to the intestinal lamina propria. These CD8 T cells appear to be activated through mechanisms involving IL-15. One recent publication demonstrated in mice that ectopic over-expression of IL-15 by enterocytes leads to the development of enteropathy, which closely resembles the lesions in CD patients. Neutralization of IL-15 activity dramatically diminished the pathological changes. Thus, an intervention blocking the activation of CD8 T cells by IL-15 appears to provide an alternative strategy in managing CD to the conventional gluten-free diet.


IL-21


IL-21 is the most recently discovered member of the γc-family. Unlike other family members, IL-21 does not appear to have potent growth-promoting effects. Instead, IL-21 is thought to function more as a differentiation factor than a factor controlling cellular proliferation (See Tagaya, 2010, J. Leuk. Biol. 87:13-15).


Current Strategies for Treating γc-Cytokine-Mediated Disorders


Because the γc-cytokines are thought to be involved in numerous human diseases, several methods of treating γc-cytokine-implicated diseases by inhibiting γc-cytokine family activities have been proposed. These methods include the use of cytokine-specific monoclonal antibodies to neutralize the targeted cytokine's activity in vivo; use of monoclonal antibodies targeting the private cytokine-specific receptor subunits (subunits other than the shared γc-subunit) to selectively inhibit cytokine activity: and use of chemical inhibitors that block the downstream intracellular cytokine signal transduction pathway.


While cytokine-specific antibodies are often the first choice in designing therapeutics, cytokines that share receptor components display overlapping functions (See Paul, W. E., 1989, Cell 57:521-24) and more than one cytokine can co-operate to cause a disease (see example described below). Thus, approaches involving neutralization of a single cytokine may not be effective in the treatment of cytokine-implicated human diseases.


Strategies for designing therapeutics that inhibit the function of multiple cytokines via antibodies which recognize a shared receptor component have also been proposed. However, the multi-subunit nature of cytokine receptor systems and the fact that functional receptors for a single cytokine can assume different configurations makes this approach difficult.


For example, a functional IL-15 receptor can be either IL-15Rβ/γc or IL-15Rα/β/γc. (See Dubois et al., 2002, Immunity 17:537-47.) An antibody against the IL-15Rβ receptor (TMβ1), is an efficient inhibitor of the IL-15 function, but only when the IL-15Rα molecule is absent from the receptor complex. (See Tanaka et al., 1991, J. Immunol. 147:2222-28.) Thus, the effectiveness of a monoclonal anti-receptor antibody, whether raised against a shared or a private subunit, can be context-dependent and is unpredictable in vivo.


Although clinical use of monoclonal antibodies against biologically active factors or receptors associated with the pathogenesis of diseases is an established practice, there are few demonstrations of successful outcomes. Moreover, establishment of a clinically-suited monoclonal antibody treatment is a long and difficult process, with the successful generation of a neutralizing antibody largely a matter of luck. For example, due to the critical importance of the γc-subunit in mediating signaling by γc-family cytokines, many attempts to generate polyclonal and monoclonal antibodies against the γc-subunit have been made and there exist many commercial antibodies recognizing the γc-subunit in mice and in humans. Curiously, however, none of these anti-γc-subunit antibodies block the function of the γc-cytokines.


Another problem with the therapeutic use of monoclonal antibodies is that monoclonal antibodies are usually generated by immunizing rodents with human proteins, so the generated antibody is a foreign protein and thus highly immunogenic. To circumvent this problem, the amino acid sequence of the monoclonal antibody is molecularly modified so that the antibody molecule is recognized as a human immunoglobulin (a process called humanization), but this process requires time and expense.


Targeting JAK3, as an Existing Alternative Example for the Inhibition of Multiple γc-Cytokines


The interaction between the γc-subunit and a γc-cytokine leads to the activation of an intracellular protein tyrosine kinase called Janus kinase 3 (Jak3). Jak3, in turn, phosphorylates multiple signaling molecules including STAT5, and PI3 kinase. The interaction of the γc-subunit and Jak3 is very specific. In fact, there is no other receptor molecule that recruits Jak3 for signal transduction. (See O'Shea, 2004, Ann. Rheum. Dis. 63:(suppl. 11):ii67-7.) Thus, the inhibition of cytokine signaling through the γc-subunit can be accomplished by blocking the activity of Jak3 kinase. Accordingly, multiple chemical inhibitors that target the kinase activity of Jak3 have been introduced to the market. (See Pesu et al., 2008, Immunol. Rev. 223:132-142.) One such example is CP690,550.


The major shortcoming of these protein kinase inhibitors is the lack of specificity to Jak3 kinase. These drugs intercept the binding of ATP (adenosine-triphosphate) molecules to Jak3 kinase, a common biochemical reaction for many protein kinases, and thus tend to block the action of multiple intracellular protein kinases that are unrelated to Jak3 kinase whose actions are critically needed for the well-being of normal cells in various tissues. Thus, more specific inhibitors of signaling through the γc-subunit are needed.


There is therefore a great need for an alternative strategy for treating γc-cytokine-implicated diseases.


Discovery of the γc-Box


The C-terminus (the D-helix) of the γc-cytokines contains the proposed site for interacting with the common γc-subunit of the multi-unit cytokine receptors. (Bernard et al., 2004 J. Biol. Chem. 279:24313-21.) Comparison of the biochemical properties of the amino acids of all γc-cytokines identified in mice and humans revealed that the chemical nature of the amino acids, for example, hydrophobicity, hydrophilicity, base/acidic nature, are conserved, if not identical, at many positions in the D-helix across the members of the γc-cytokine family.


In contrast, the sequence of IL-13, which is related to the γc-cytokine, IL-4, but does not bind to the γc-subunit, does not exhibit significant homology in the D-helix region to the γc-cytokines, suggesting that the sequence homology in the D-helix region is correlated with binding to the γc-subunit. As shown in FIG. 1A, alignment of the amino acid sequences of the D-helix region of γc-cytokine family members in humans reveals a motif of moderate sequence homology in these cytokines referred to herein as “the γc-box”.


The γc-box (SEQ ID NO: 10) comprises 19 amino acids where out of the 19 positions, positions 4, 5, and 13 are fully conserved as Phenylalanine, Leucine, and Glutamine, respectively. Less conservation is observed at positions 6, 7 and 11 of the γc-box where the amino acid is one of two or three related amino acids that share physico-chemical properties: position 6 may be occupied by the polar amino acids Glutamate. Asparagine or Glutamine; non-polar amino acids Serine or Arginine can occupy position 7; and position 11 is occupied by either of the non-polar aliphatic amino acids Leucine or Isoleucine. Positions 9 and 16 may be occupied by the either the non-polar amino acid Isoleucine or the polar amino acid Lysine. See FIG. 1B. Some differences in the amino acid composition of the γc-box are observed at positions 9 and 16 amongst subfamilies of the γc-cytokines. Comparison of the γc-cytokines across species indicates that Isoleucine is often present at the 9 and 16 positions in the IL-2/15 subfamily, whereas the other γc-family members often possess Lysine in these positions. Not wishing to be bound by a particular theory, Isoleucine and Lysine are biochemically different and thus may impart specific conformational differences between the IL-2/15 subfamily and other γc-cytokines.


Conservation of the γc-box motif between γc-cytokines is supported by findings that an Glutamine (Gin, Q) residue located in the D-helix region is critical for the binding of the γc-cytokines to the γc-subunit. (Bernard et al., 2004 J. Biol. Chem. 279: 24313-21.)


Peptide Inhibitors of γc-Cytokine Activity


The activity of γc-family cytokines may be blocked by disrupting the interaction between the γc-cytokine and the γc-subunit, for example by introducing a competitive inhibitor which can interact with the γc-subunit without stimulating signaling through the multi-subunit cytokine receptors. Not to be bound by a particular theory, the conserved γc-box motif, which participates in binding of the γc-family cytokines to the γc-subunit, presents a core base amino acid sequence which can be utilized to design peptide inhibitors of γc-cytokine signaling.


The core γc-box amino acid sequence comprises: D/E-F-L-E/Q/N-S/R-X-I/K-X-L/I-X-Q (SEQ ID NO: 2) (where X denotes any amino acid). At least some embodiments described herein relate to custom peptide derivatives of the core γc-box amino acid sequence which can inhibit the activity of one or more γc-cytokines. Custom peptide derivatives include any peptide whose partial amino acid sequence shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to the core γc-box amino acid sequence. Custom peptide derivatives further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of the core γc-box. For example, amino acids with similar physico-chemical properties would include Phenylalanine, Tyrosine, Tryptophan, and Histidine, which are aromatic amino acids. FIG. 2 shows a diagrammed representation of amino acids with similar physico-chemical properties which may be may be substituted for the amino acids comprising the core γc-box. Peptide derivatives of the core γc-box may be 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 24, 25-30, 30-35, 35-40, 40-45, 45-50, or more than 50 amino acids in length. In some embodiments, the custom peptide derivatives may be conjugated to the N-termini, C-termini and/or to the side residues of existing biological proteins/peptides.


Based on the identification of the conserved γc-box motif in cytokines which bind to the γc-subunit, Applicants have devised a novel, 19-mer custom derivative peptide which is an artificial composite peptide combining the amino acid sequence of the human IL-2 and IL-15 γc-box. The 19-mer peptide, herein referred to as BNZ-γ, consists of the amino acid sequence: I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), where the amino acids depicted by bold characters are conserved between IL-2 and IL-15 and the underlined amino acids represent positions where the physico-chemical properties of the amino acids are conserved.


Applicants discovered that the 19-mer BNZ-γ, suppresses IL-15 and IL-9 induced cellular proliferation, but not IL-3 or IL-4 induced cellular proliferation. See FIG. 3A and EXAMPLE 2. Applicants further demonstrated that BNZ-γ inhibits IL-15 mediated phosphorylation of the intracellular cytokine signal transduction molecule, STAT-5. See FIG. 3C and EXAMPLE 5. These results demonstrate that custom peptide derivatives of the conserved γc-box motif can inhibit the activity of multiple γc-cytokines.


Several embodiments relate to custom derivative peptides of the 19-mer BNZ-γ amino acid sequence, I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), which can inhibit the activity of one or more γc-cytokines. Custom peptide derivatives of the 19-mer BNZ-γ amino acid sequence include any peptide whose partial amino acid sequence shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to amino acid sequence: I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S(SEQ ID NO: 1). Custom peptide derivatives further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of sequence: I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S(SEQ ID NO: 1). In several embodiments, the amino acid residues of the custom derivative peptides retain similar physico-chemical properties with the amino acid residues of BNZ-γ, but exhibit different biological inhibition specificity to the 6 γc-cytokine family members from that of the original 19-mer peptide. Peptide derivatives of BNZ-γ may be 19, 20, 21, 22, 24, 25-30, 30-35, 35-40, 40-45, 45-50, or more than 50 amino acids in length. In some embodiments, the custom peptide derivatives may be conjugated to the N-termini, C-termini and/or to the side residues of existing biological proteins/peptides.


Several embodiments relate to custom peptide derivatives of the γc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7, which are depicted in FIG. 1A. Other embodiments relate to custom derivative peptides which are artificial composite peptides combining the amino acid sequence of two or more of the human IL-15, IL-2, IL-21, IL-4, IL-9, and IL-7 γc-box motifs. Several embodiments relate to custom peptide derivatives of the of the γc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7 having a partial amino acid sequence that shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%. 90%, 95%, 97%, 98%, 99% or 99.8% identity to amino acid sequences of the of the γc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7. Custom peptide derivatives of the of the γc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7 further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of sequence of the γc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7.


Several embodiments relate to custom peptide derivatives that would inhibit the function of one, all, or selective members of the γc-cytokines. In some embodiments, the custom peptide derivatives selectively target individual γc-cytokine family members. For example, a custom peptide derivative can selectively inhibit the function of IL-2, IL-4, IL-7, IL-9, IL-15, or IL-21. In other embodiments, a custom peptide derivative can inhibit 2 or more γc-cytokine family members.


For example, the custom peptide derivatives of the present embodiments can selectively inhibit the function of IL-2 in combination with one or more of IL-4, IL-7, IL-9, IL-15, and IL-21; IL-4 in combination with one or more of IL-7, IL-9, IL-15, and IL-21; IL-7 in combination with one or more of IL-9, IL-15, and IL-21; IL-9 in combination with one or more of IL-2, IL-4, IL-7, IL-15, and IL-21; IL-15 in combination with one or more of IL-2, IL-4, IL-7, IL-9, and IL-21: or IL-21 in combination with one or more of IL-2, IL-4, IL-7, IL-9, and IL-15. In other embodiments, custom peptide derivatives can comprehensively target all γc-cytokine family members.


Not wishing to be bound by a particular theory, the custom peptide derivatives can inhibit the function of all or selective members of the γc-cytokines by diminishing the binding of γc-cytokines to the γc-subunit, for example, as a competitive inhibitor. Such custom peptide derivatives may be used in diverse applications, including as a clinical drug.


Several embodiments relate to custom peptide derivatives that would modulate (including enhance or reduce) the function of one, two, or more of selective members of the γc-cytokines. In some embodiments, the custom peptide derivatives selectively target individual γc-cytokine family members. For example, a custom peptide derivative can selectively enhance or inhibit the function of IL-2, IL-4, IL-7, IL-9, IL-15, or IL-21. In other embodiments, a custom peptide derivative can enhance or inhibit two or more γc-cytokine family members. In certain embodiments, custom peptide derivatives may comprise P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), which can enhance or inhibit the activity of one, two or more of γc-cytokines. In certain embodiments, custom peptide derivatives may comprise P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), which can inhibit the activity of at least IL-15 and IL-21.


In some embodiments, custom peptide derivatives may include any peptide whose partial amino acid sequence shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to amino acid sequence: P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3). Custom peptide derivatives further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of sequence: P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).


In several embodiments, the amino acid residues of the custom derivative peptides retain similar physico-chemical properties with the amino acid residues of P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), but exhibit different biological inhibition specificity to the 6 γc-cytokine family members (i.e. IL-2, IL-4, IL-7, IL-9, IL-15, or IL-21) from that of the original peptide of P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3). Peptide derivatives of the sequence of P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3) may be 19, 20, 21, 22, 24, 25-30, 30-35, 35-40, 40-45, 45-50, or more than 50 amino acids in length.


In some embodiments, the custom peptide derivatives may be conjugated to the N-termini, C-termini and/or to the side residues of existing biological proteins/peptides. In some embodiments, the composite peptide of SEQ ID NO: 3 may be conjugated to other moieties through the N-terminus, C-terminus or side chains of the composite peptide. The other moieties may include proteins or peptides that stabilize the composite peptide, or other moieties, including without limitation, bovine serum albumin (BSA), albumin, Keyhold Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell-specific antigen, a receptor, a ligand, a metal ion and Poly Ethylene Glycol (PEG).


The terms “oligopeptide,” “polypeptide,” “peptide,” and “protein” can be used interchangeably when referring to the custom peptide derivatives provided in accordance with the present embodiments and can be used to designate a series of amino acid residues of any length. Peptides according to the present embodiments may also contain non-natural amino acids. Linker elements can be joined to the peptides of the present embodiments through peptide bonds or via chemical bonds. The peptides of the present embodiments may be linear or cyclic, and may include (D) as well as (L) amino acids.


Peptides of the present embodiments may also contain one or more rare amino acids (such as 4-hydroxyproline or hydroxylysine), organic acids or amides and/or derivatives of common amino acids, such as amino acids having the C-terminal carboxylate esterified (e.g., benzyl, methyl or ethyl ester) or amidated and/or having modifications of the N-terminal amino group (e.g., acetylation or alkoxycarbonylamino), with or without any of a wide variety of side chain modifications and/or substitutions (e.g., methylation, benzylation, t-butylation, tosylation, alkoxycarbonylamino, and the like).


Residues other than common amino acids that may be present include, but are not limited to, penicillamine, tetramethylene cysteine, pentamethylene cysteine, mercaptopropionic acid, pentamethylene-mercaptopropionic acid, 2-mercaptobenzene, 2-mercaptoaniline, 2-mercaptoproline, ornithine, diaminobutyric acid, aminoadipic acid, m-aminomethylbenzoic acid, and diaminopropionic acid.


Peptides of the present embodiments can be produced and obtained by various methods known to those skilled in the art. For example, the peptide may be produced by genetic engineering, based on the nucleotide sequence coding for the peptide of the present embodiments, or chemically synthesized by means of peptide solid-phase synthesis and the like, or produced and obtained in their combination.


One skilled in the art can synthesize the custom peptide derivatives based on the present disclosure of the conserved γc-box motif and knowledge of the biochemical properties of amino acids as described in FIG. 2. Some embodiments also relate to polynucleotides comprising nucleotide sequences encoding the peptides of the present invention. “Nucleotide sequence,” “polynucleotide,” or “nucleic acid” can be used interchangeably, and are understood to mean either double-stranded DNA, a single-stranded DNA or products of transcription of the said DNAs (e.g., RNA molecules). Polynucleotides can be administered to cells or subjects and expressed by the cells or subjects, rather than administering the peptides themselves. Several embodiments also relate to genetic constructs comprising a polynucleotide sequence encoding the peptides of the present invention. Genetic constructs can also contain additional regulatory elements such as promoters and enhancers and, optionally, selectable markers.


Methods of Treating γc-Cytokine Mediated Diseases


Several embodiments relate to the use of γc-antagonist peptides in the treatment of γc-cytokine mediated diseases. Use of custom peptide derivative according to the present embodiments allows for flexibility in the design of the therapeutic agent (custom design of the peptide) and enables more comprehensive outcomes, which would not be accomplished by conventional strategies employing anti-cytokine or anti-cytokine receptor antibodies.


Described herein is a novel method of blocking the action of γc-family cytokines. Such manipulations can yield effective methods of clinical interventions in treating diseases related to the dysregulation or dysfunction of γc-cytokines. Examples of disease that may be treated by disrupting the interaction between the γc-cytokine and the γc-subunit include autoimmune diseases such as systemic lupus erythematosis, Sjögren's syndrome. Wegener's granulomatosis Celiac disease. Hashimoto's or auto-immune thyroiditis; collagen diseases including rheumatoid arthritis, inflammatory bowel disease, diabetes mellitus, autoimmune diseases of the skin such as psoriasis; degenerative neuronal diseases such as multiple sclerosis, uvietis or inflammation of the eye and sympathetic ophthalmia, graft-versus-host disease (GvHD) and myasthenia gravis.


In some embodiments, the γc-antagonist peptides described herein may be used in the treatment of 1-Human T-cell Lymphotropic type I and II (HTLV-I and HTLV-II)-associated diseases including Adult T-cell Leukemia (ATL), HTLV-associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy and myositis. In some embodiments, the γc-antagonist peptides described herein may be used in the treatment of other viral diseases such as influenza. AIDS, HBV and Herpes or parasitic diseases.


In several embodiments, the γc-antagonist peptides may be administered before, during, and or after transplantation of various organs as an immunosuppressant agent.


In some embodiments, the γc-antagonist peptides described herein may be used in the treatment of immune-mediated diseases such as asthma and other inflammatory respiratory diseases, such as, but not limited to sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, lung fibrosis. In some embodiments. γc-antagonist peptides may be administered to treat or prevent allergic reactions due to exposure to allergens, chemical agents or other common causes of acute respiratory disease. In some embodiments, γc-antagonist peptides may be administered to treat or prevent inflammatory responses caused by viruses, bacteria, chemical reagents, and biochemical reagents.


In several embodiments, the γc-antagonist peptides may be administered to treat some types of malignancies such as LGL-leukemia, Intraepithelial lymphoma and leukemia in Refractory Celiac Disease, NK leukemia/lymphoma and NK-T leukemia/lymphoma


In some embodiments, custom peptide derivatives according to the embodiments described herein can be used for cosmetic purposes, such as the treatment of acne, hair loss, sunburn and nail maintenance, included to ointment as anti-aging component because of the anti-inflammatory nature of them.


Several embodiments relate to therapeutic antagonist peptides that would inhibit the function of all or selective members of the γc-cytokines. In some embodiments, therapeutic antagonist peptides selectively inhibit individual γc-cytokine family members (custom peptides). In other embodiments, therapeutic antagonist peptides can comprehensively inhibit all γc-cytokine family members (Simul-Block). In some embodiments, therapeutic antagonist peptides selectively inhibit subsets of the γc-cytokines. Not wishing to be bound by a particular theory, the peptide antagonists can inhibit the function of all or selective members of the γc-cytokines by diminishing the binding of γc-cytokines to the γc-subunit, for example, as a competitive inhibitor.


Several members of the γc-cytokine family, IL-2, IL-7, and IL-15, but not IL-4 have been implicated as being involved in graft versus host disease (GvHD) in an experimental mouse model. (Miyagawa et al., 2008 J. Immunol. 181:1109-19.) One embodiment relates to the use of therapeutic antagonist peptides that selectively inhibit IL-2, IL-7, and IL-15 activity for the treatment of GvHD in humans, allowing survival of the grafted tissues or bone marrow cells. Other embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit a combination of IL-2 and IL-7, IL-2, and IL-15, or IL-7 and IL-15 to treat GvHD. Other embodiments relate to the use of a combination of therapeutic antagonist peptides that selectively inhibit IL-2, IL-7, or IL-15.


Some embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-2 function for the treatment of autoimmune disorders where T-regs have been implicated as playing a role. In some embodiments, peptide-mediated inhibition of T-regs can enhance the natural anti-cancer immunity in humans, providing a novel means of anti-cancer therapy.


Several embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-4 to treat asthma.


Some embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-7 either alone or in combination with therapeutic antagonist peptides that selectively inhibit the γc-cytokine family member, IL-15, as a therapeutic agent for LGL leukemia. In some embodiments therapeutic antagonist peptides that selectively inhibit both IL-7 and IL-15 activity can be used to treat LGL leukemia. Several embodiments relate to the use of BNZ-γ to treat LGL leukemia. In some embodiments, specific γc-antagonist peptides that selectively IL-15 alone or specific γc-antagonist peptides that selectively IL-15 and IL-7 are used as a therapeutic agent for CD4/CD8 T lymphocyte-associated leukemia including that caused by the HTLV-I.


Several embodiments relate to the use of γc-antagonist peptides that selectively inhibit the activity of IL-9, either alone or in combination with the other γc-cytokine family members, as a therapeutic agent for human diseases that involve the abnormal development of Th17 cells.


Several embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-15 activity as a therapeutic agent for treating CD. One recent publication suggested that IL-21, in addition to IL-15, may play a role in CD pathogenesis. (See Bodd et al., 2010, Mucosal Immunol. 3:594-601.) This suggests that optimum treatment of CD by conventional anti-cytokine or cytokine-receptor antibodies would benefit from a combination of at least two antibodies recognizing component that belong to the IL-15 and IL-21 systems. In some embodiments, custom derivative antagonist peptides that selectively inhibit both IL-15 and IL-21 activity are used as a therapeutic agent for treating CD.


In addition to having therapeutic applications, γc-antagonist peptides have applications in consumer products as well. Several embodiments relate to the use of γc-antagonist peptides in skin care products such as anti-aging, anti-inflammatory, anti-acne, and other related applications. Some embodiments relate to the use of γc-antagonist peptides in hair products as anti-hair loss ingredient to treat hair loss caused by autoimmune disorders.


Another embodiment relates to the development of chemical compounds (non-peptide, non-protein) that have a spatial structure which resembles the 19-mer amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) and can fit into the pocket of the γc-subunit to structurally hinder the access of a γc-cytokine to the γc-subunit for binding. Some embodiments relate to the use of structurally similar chemical compounds as inhibitors of γc-cytokine activity. Such molecular mimicry strategy to further refine the development of synthetic compounds resembling in structure to existing biological peptide/proteins is described in Orzaez et al., 2009 Chem. Med. Chem. 4:146-160. Another embodiment relates to administration of chemical compounds (non-peptide, non-protein) that have a resembling 3D structure as the 19-mer amino acids sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) to treat γc-cytokine-mediated diseases.


Several embodiments relates to the administration of a peptide of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) to treat γc-cytokine-mediated diseases. Another embodiment relates to the administration of derivative peptides of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S(SEQ ID NO: 1), wherein the amino acid sequence of the derivative peptide has similar physico-chemical properties as a peptide of the amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), but has distinct biological activity, to treat γc-cytokine-mediated diseases. Another embodiment relates to administration of a peptide of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) conjugated to the N- and C-termini or to the side residues of existing biological proteins/peptides into patients to treat γc-cytokine-mediated diseases.


Several embodiments relate to administration of polyclonal and monoclonal antibodies raised against a peptide comprising of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) into patients as an immunogen to treat γc-cytokine-mediated diseases. Another embodiment relates to administration of polyclonal and monoclonal antibodies that were raised against derivative peptides of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) wherein the amino acid sequence of the derivative peptide has similar physico-chemical properties as a peptide of the amino acid sequence 1-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), but has distinct biological activity, into patients as an immunogen to treat γc-cytokine-mediated diseases.


Administration of γc-Antagonist Peptides


The present embodiments also encompass the use of γc-antagonist peptides for the manufacture of a medicament for the treatment of a disease. The present embodiments also encompass a pharmaceutical composition that includes γc-antagonist peptides in combination with a pharmaceutically acceptable carrier. The pharmaceutical composition can include a pharmaceutically acceptable carrier and a non-toxic therapeutically effective amount of γc-antagonist peptides, or other compositions of the present embodiments.


The present embodiments provide methods of using pharmaceutical compositions comprising an effective amount of antagonists for γc-cytokines in a suitable diluent or carrier. A γc-antagonist of the present embodiments can be formulated according to known methods used to prepare pharmaceutically useful compositions. A γc-antagonist can be combined in admixture, either as the sole active material or with other known active materials, with pharmaceutically suitable diluents (e.g., phosphate, acetate, Tris-HCl), preservatives (e.g., thimerosal, benzyl alcohol, parabens), emulsifying compounds, solubilizers, adjuvants, and/or carriers such as bovine serum albumin.


Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 16th ed. 1980 Mack Publishing CO. Additionally, such compositions can contain a γc-antagonist complexed with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels etc., or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts, or spheroblasts. Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance or a γc-antagonist. A γc-antagonist can be conjugated to antibodies against cell-specific antigens, receptors, ligands, or coupled to ligands for tissue-specific receptors.


Methods of administrating γc-antagonists of the present embodiments may be selected as appropriate, depending on factors, such as the type of diseases, the condition of subjects, and/or the site to be targeted. The γc-antagonists can be administered topically, orally, parenterally, rectally, or by inhalation. The term “parenteral” includes subcutaneous injections, intravenous, intramuscular, intraperitoneal, intracisternal injection, or infusion techniques. These compositions will typically include an effective amount of a γc-antagonist, alone or in combination with an effective amount of any other active material.


The amount of the peptide contained in pharmaceutical compositions of the present embodiments, dosage form of the pharmaceutical compositions, frequency of administration, and the like may be selected as appropriate, depending on factors, such as the type of diseases, the condition of subjects, and/or the site to be targeted. Such dosages and desired drug concentrations contained in the compositions may vary affected by many parameters, including the intended use, patient's body weight and age, and the route of administration. Pilot studies will first be conducted using animal studies and the scaling to human administration will be performed according to art-accepted practice.


In one embodiment, host cells that have been genetically modified with a polynucleotide encoding at least one γc-antagonist peptide are administered to a subject to treat a proliferation disorder and/or to reduce the growth of malignant cells. The polynucleotide is expressed by the host cells, thereby producing the peptides within the subject. Preferably, the host cells are allogeneic or autogenic to the subject.


In a further aspect, γc-antagonist peptides can be used in combination with other therapies, for example, therapies inhibiting cancer cell proliferation and growth. The phrase “combination therapy” embraces the administration of γc-antagonist peptides and an additional therapeutic agent as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of these therapeutic agents. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually minutes, hours, days or weeks depending upon the combination selected).


A combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by an appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. There therapeutic agents can be administered by the same route or by different routes. The sequence in which the therapeutic agents are administered is not narrowly critical.


Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients (such as, but not limited to, a second and different therapeutic agent) and non-drug therapies (such as, but not limited to, surgery or radiation treatment). Where the combination therapy further comprises radiation treatment, the radiation treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and radiation treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the radiation treatment is temporarily removed from the administration of the therapeutic agents, perhaps by days or even weeks.


In certain embodiments, γc-antagonist peptides can be administered in combination with at least one anti-proliferative agent selected from the group consisting of chemotherapeutic agent, an antimetabolite, and antitumorgenic agent, and antimitotic agent, and antiviral agent, and antineoplastic agent, an immunotherapeutic agent, and a radiotherapeutic agent.


In certain embodiments, γc-antagonist peptides can be administered in combination with at least one anti-inflammatory agent selected from the group consisting of steroids, corticosteroids, and nonsteroidal anti-inflammatory drugs.


Also provided are kits for performing any of the methods provided herein. In some embodiments, kits may include one or more γc-antagonist according to any of the embodiments provided herein. In some embodiments, the kit may include instructions. Instructions may be in written or pictograph form, or may be on recorded media including audio tape, audio CD, video tape, DVD, CD-ROM, or the like. The kits may comprise packaging.


Definitions


As used herein, the term “patient” refers to the recipient of a therapeutic treatment and includes all organisms within the kingdom animalia. In preferred embodiments, the animal is within the family of mammals, such as humans, bovine, ovine, porcine, feline, buffalo, canine, goat, equine, donkey, deer, and primates. The most preferred animal is human.


As used herein, the term “treat” or any variation thereof (e.g., treatment, treating, etc.), refers to any treatment of a patient diagnosed with a biological condition, such as CD4-, CD8-, and LGL-leukemia, an autoimmune disease, systemic lupus erythematosis, Sjögren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, a collagen disease, rheumatoid arthritis, inflammatory bowel disease, diabetes mellitus, psoriasis, a degenerative neuronal disease, multiple sclerosis, uvietis, inflammation of the eye, graft-versus-host disease (GvHD), myasthenia gravis, 1-Human T-cell Lymphotropic type I and II (HTLV-I and HTLV-II)-associated diseases, Adult T-cell Leukemia (ATL), HTLV-associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, myositis, influenza, AIDS, HBV, Herpes, asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, lung fibrosis, NK leukemia/lymphoma and NK-T leukemia/lymphoma.


The term treat, as used herein, includes: (i) preventing or delaying the presentation of symptoms associated with the biological condition of interest in an at-risk patient who has yet to display symptoms associated with the biological condition; (ii) ameliorating the symptoms associated with the biological condition of interest in a patient diagnosed with the biological condition; (iii) preventing, delaying, or ameliorating the presentation of symptoms associated with complications, conditions, or diseases associated with the biological condition of interest in either an at-risk patient or a patient diagnosed with the biological condition: (iv) slowing, delaying or halting the progression of the biological condition; and/or (v) preventing, delaying, slowing, halting or ameliorating the cellular events of inflammation.


The term “symptom(s)” as used herein, refers to common signs or indications that a patient is suffering from a specific condition or disease.


The term “effective amount,” as used herein, refers to the amount necessary to elicit the desired biological response. In accordance with the present embodiments, an effective amount of a γc-antagonist is the amount necessary to provide an observable effect in at least one biological factor for use in treating a biological condition.


“Recombinant DNA technology” or “recombinant” refers to the use of techniques and processes for producing specific polypeptides from microbial (e.g., bacterial, yeast), invertebrate (insect), mammalian cells or organisms (e.g., transgenic animals or plants) that have been transformed or transfected with cloned or synthetic DNA sequences to enable biosynthesis of heterologous peptides. Native glycosylation pattern will only be achieved with mammalian cell expression system. Prokaryotic expression systems lack the ability to add glycosylation to the synthesized proteins. Yeast and insect cells provide a unique glycosylation pattern that may be different from the native pattern.


A “Nucleotide sequence” refers to a polynucleotide in the form of a separate fragment or as a component of a larger DNA construct that has been derived from DNA or RNA isolated at least once in substantially pure form, free of contaminating endogenous materials and in a quantity or concentration enabling identification, manipulation, and recovery of its component nucleotide sequences by standard molecular biology methods (as outlined in Current Protocols in Molecular Biology).


“Recombinant expression vector” refers to a plasmid comprising a transcriptional unit containing an assembly of (1) a genetic element or elements that have a regulatory role in gene expression including promoters and enhances, (2) a structure or coding sequence that encodes the polypeptide according to the present embodiments, and (3) appropriate transcription and translation initiation sequence and, if desired, termination sequences. Structural elements intended for use in yeast and mammalian system preferably include a signal sequence enabling extracellular secretion of translated polypeptides by yeast or mammalian host cells.


“Recombinant microbial expression system” refers to a substantially homogenous monoculture of suitable hot microorganisms, for example, bacteria such as E. coli, or yeast such as S. cerevisiae, that have stably integrated a recombinant transcriptional unit into chromosomal DNA or carry the recombinant transcriptional unit as a component of a residual plasmid. Generally, host cells constituting a recombinant microbial expression system are the progeny of a single ancestral transformed cell. Recombinant microbial expression systems will express heterologous polypeptides upon induction of the regulatory elements linked to a structural nucleotide sequence to be expressed.


As used herein, the section headings are for organizational purposes only and are not to be construed as limiting the described subject matter in any way. All literature and similar materials cited in this application, including but not limited to, patents, patent applications, articles, books, treatises, and internet web pages are expressly incorporated by reference in their entirety for any purpose. When definitions of terms in incorporated references appear to differ from the definitions provided in the present teachings, the definition provided in the present teachings shall control. It will be appreciated that there is an implied “about” prior to the temperatures, concentrations, times, etc discussed in the present teachings, such that slight and insubstantial deviations are within the scope of the present teachings herein.


Although this invention has been disclosed in the context of certain embodiments and examples, those skilled in the art will understand that the present invention extends beyond the specifically disclosed embodiments to other alternative embodiments and/or uses of the invention and obvious modifications and equivalents thereof. In addition, while several variations of the invention have been shown and described in detail, other modifications, which are within the scope of this invention, will be readily apparent to those of skill in the art based upon this disclosure.


It is also contemplated that various combinations or sub-combinations of the specific features and aspects of the embodiments may be made and still fall within the scope of the invention. It should be understood that various features and aspects of the disclosed embodiments can be combined with, or substituted for, one another in order to form varying modes or embodiments of the disclosed invention. Thus, it is intended that the scope of the present invention herein disclosed should not be limited by the particular disclosed embodiments described above.


It should be understood, however, that this detailed description, while indicating preferred embodiments of the invention, is given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art.


EXAMPLES

The following Examples are presented for the purposes of illustration and should not be construed as limitations.


Example 1—Method for Assessing the Inhibitory Activity of γc-Antagonist Peptide

The capacity of any custom derivative peptide prepared according to the present embodiments for inhibiting the action of one γc-cytokine family member is determined using mammalian cellular assays to measure their proliferative response to the γc-cytokine family member.


For each of the six γc-cytokines, indicator cell lines: CTLL-2, a murine CD8 T cells line available from American Type Culture Collection, and PT-18, a murine mast cell line and its subclone PT-18β, is transfected with human IL-2Rβ gene to make the cells responsive to IL-2 and IL-15 (Tagaya et al., 1996, EMBO J. 15:4928-39), and is used to quantitatively determine the γc-cytokine's growth-promoting activity (See Current protocols in Immunology from Wiley and Sons for a methodological reference). The indicator cells demonstrate semi-linear dose-dependent response when measured by a colorimetric WST-1 assay over a range of concentrations (See Clontech PT3946-1 and associated user manual, incorporated herein by reference, for a detailed description of the reagents and methods).


Once the appropriate doses of the cytokine that yield the 50% and 95% maximum response from the indicator cell line is determined, various concentrations (ranging from 1 pM to 10 μM) of the purified or synthesized custom derivative peptide is added to each well containing the cytokine and indicator cells. The reduction in light absorbance at 450 nm is used as an indicator of inhibition of cytokine-stimulated cellular proliferation. Typically, the cells are stimulated by the cytokines such that the absorbance of the well containing indicator cell line and the cytokine is between 2.0 and 3.0, which is reduced to a range of 0.1 to 0.5 by the addition of inhibitory peptides.


Example 2—BNZ-γ Peptide Specifically Inhibits the Growth-Promoting Activities of IL-9 and IL-15

Using PT-183 cells as described above, the ability of the BNZ-γ peptide to specifically inhibit the growth-promoting activity of select γc-cytokines was determined (FIG. 3A). IL-3, a non-γc-cytokine that supports the growth of PT-183 cells, was used as a negative control. Briefly, PT-185 cells were incubated either with two different dilutions of BNZ-γ peptide produced by HEK293T cells (1:20 or 1:50 dilution of the original supernatant of HEK293T cells transfected with a BNZ-γ expression construct) or without BNZ-γ peptide in the presence of IL-3, IL-9, IL-15, or IL-4 (1 nM of each cytokine in the culture).


The growth-responses of the cells were determined 2 days after the introduction of BNZ-γ peptide and the cytokine using the WST-1 assay. The growth-promoting activity of IL-3 (a non γc-cytokine) was not inhibited by BNZ-γ. In contrast, the activity of IL-15 and IL-9 were significantly (p<0.01 Student's T test) reduced by the BNZ-γ peptide. Cellular proliferation stimulated by IL-4, another γc-cytokine, was not affected by the by the addition of BNZ-γ peptide. Results for IL-3, IL-9, IL-15, and IL-4 are shown at FIG. 3A.


In a similar assay, the murine cell line CTTL2 was used. In this assay the cells were cultured with 0.5 nM of recombinant IL-2 in RPMI 10% fetal Calf Serum. To set up the proliferation assay, cells were washed from the cytokines 3 times. Cells were seeded at 1×10(5) cells per well of a 96-well plate with final concentration of 50 pM of IL-2 or IL-15. Various concentration of BNZ-γ peptide (0.1, 1, and 10 ug/ml) was added to each well. Cells were cultured for 20 hours and in the last 4 hours, 3H-thymidine was added to the plates. Cells were harvested using a plate reader. The data is shown in FIG. 3B.


Example 3—Method for Measuring Inhibition γc-Cytokine Activity by Assaying 3H-Thymidine Incorporation of as a Marker of Cellular Proliferation

Inhibition of γc-cytokine-induced proliferation of an indicator cell population by antagonist custom derivative peptides is measured by the 3H-thymidine incorporation assay. Briefly, radiolabeled thymidine (1 microCi) is given to 20-50.000 cells undergoing proliferation in the presence of cytokines. The cell-incorporated radioactivity is measured by trapping cell-bound radioactivity to a glass-fiber filter using a conventional harvester machines (for example, Filtermate Universal Harvester from Perkin-Elmer), after which the radioactivity is measured using a b-counter (for example, 1450 Trilux microplate scintillation counter).


Example 4—Method for Measuring Inhibition γc-Cytokine Activity by Assaying Incorporation of a Cell-Tracker Dye as a Marker of Cellular Proliferation

Indicator cells are incubated in the presence of a selected γc-cytokine or in the presence of a selected γc-cytokine and a selected custom derivative peptide. The cell population is then labeled in vitro using a cell-tracker dye, for example, CMFDA, C2925 from Invitrogen, and the decay of cellular green fluorescence at each cellular division is monitored using a flow-cytometer (for example, Beckton-Dickinson FACScalibur). Typically, in response to γc-cytokine stimulation 7˜10 different peaks corresponding to the number of divisions that the cells have undergone will appear on the green fluorescence channel. Incubation of the cells with the selected γc-cytokine and antagonist custom derivative peptide reduces the number of peaks to only 1 to 3, depending on the degree of the inhibition.


Example 5—Inhibition of Intracellular Signaling by BNZ-γ and its Derivative Antagonists

In addition to stimulating cellular proliferation, binding of the γc-cytokines to their receptors causes a diverse array of intracellular events. (Rochman et al. 2009 Nat. Rev. Immunol. 9:480-90, Pesu et al. 2005 Immunol. Rev. 203:127-142.) Immediately after the cytokine binds to its receptor, a tyrosine kinase called Jak3 (Janus-kinase 3) is recruited to the receptor at the plasma membrane. This kinase phosphorylates the tyrosine residues of multiple proteins including the γc-subunit, STAT5 (Signal Transducer and Activator of Transcription 5) and subunits of the PI3 (Phosphatidylinositol 3) kinase. Among these, the phosphorylation of STAT5 has been implicated in many studies as being linked to the proliferation of cells initiated by the γc-cytokine. (Reviewed in Hennighausen and Robinson, 2008 Genes Dev. 22:711-21.) In accordance with these published data, whether or not the BNZ-γ peptide inhibits the tyrosine phosphorylation of STAT5 molecule in PT-18β cells stimulated by IL-15 was examined (results shown in FIG. 3C).


PT-18β cells were stimulated by IL-15 in the presence or absence of BNZ-γ peptide. Cytoplasmic proteins were extracted from the cells according to a conventional method as described in Tagaya et al. 1996 EMBO J. 15:4928-39. The extracted cytoplasmic proteins were resolved using a standard SDS-PAGE (Sodium Dodecyl-Sulfate PolyAcrylamide Gel Electrophoresis) and the phorphorylation status was confirmed by an anti-phospho-STAT5 antibody (Cell Signaling Technology, Catalog #9354, Danvers Mass.) using immunoblotting (see FIG. 3C, top panel). To confirm that each lane represented a similar total protein load, the membrane was then stripped, and re-probed with an anti-STAT5 antibody (Cell Signaling Technology, Catalog #9358) (see FIG. 3C, bottom panel).


These results demonstrated that tyrosine phosphorylation of STAT5, a marker of signal transduction, was induced by IL-15 in PT-18β cells, and tyrosine phosphorylation of STAT5 was markedly reduced by the BNZ-γ peptide.


Example 6—Rational Design for BNZ-γ Derivative Antagonistic Peptides

Derivative peptides are prepared based from the core sequence D/E-F-L-E/Q/N-S/R-X-I/K-X-L/I-X-Q (SEQ ID NO: 2) (where X denotes any amino acid) by substituting the defined amino acids of the core sequence with amino acids having identical physico-chemical properties as designated in FIG. 2.


Alternatively, custom peptides or their derivative peptides can be prepared based on the sequence alignment of the D-helix regions of different γc-cytokine family members. For example, as shown in FIG. 5, one or more sequences conserved in γc-cytokine family (SEQ ID NO: 4-SEQ ID NO: 9) members can be combined to form a peptide such as SEQ ID NO: 3.


Example 7—Method of Identifying the Inhibitory Specificity of Antagonistic Custom Derivative Peptides

The γc-cytokine inhibitory specificity of antagonistic custom derivative peptides is determined by assaying the ability of a custom derivative peptide to inhibit the proliferative response of a cytokine-responsive cell line to each of the 6 γc-cytokines. For example, a mouse cell line, CTLL-2, is used to determine if a candidate peptide inhibits the function of IL-2 and IL-15. PT-18(β) cells are used to determine if a candidate peptide inhibits the function of IL-4 and IL-9. PT-18 (7α) cells are used to determine if a candidate peptide inhibits the function of IL-7, and PT-18(21α) cells are used to determine if a candidate peptide inhibits the function of IL-21. PT-18(β) denotes a subclone of PT-18 cells that exogenously express human IL-2Rβ by gene transfection (See Tagaya et al. 1996), PT-18(7α) denotes a subclone that expresses human IL-7Rα by gene transfection and PT-18(21Rα) cells express human IL-21Rα.


Another alternative is to use other cell lines that respond to an array of cytokines. An example of this cell line in a human NK cell line NK92 that is commercially available by ATCC (catalog #CRL-2407). This cell line is an IL-2 dependent cell line that responds to other cytokines including IL-9, IL-7, IL-15, IL-12, IL-18, IL-21 (Gong et al. 1994 Leukemia 8: 652-658, Kingemann et al., 1996, Biol Blood Marrow Transplant 2:68; 75, Hodge D L et al., 2002 J. Immunol. 168:9090-8)


Example 8—Preparation of γc-Antagonist Peptides

Custom derivative γc-antagonist peptides are synthesized chemically by manual and automated processes.


Manual Synthesis:


Classical liquid-phase synthesis is employed, which involves coupling the carboxyl group or C-terminus of one amino acid to the amino group or N-terminus of another. Alternatively, solid-phase peptide synthesis (SPPS) is utilized.


Automated Synthesis:


Many commercial companies provide automated peptide synthesis for a cost. These companies use various commercial peptide synthesizers, including synthesizers provided by Applied Biosystems (ABI). Custom derivative γc-antagonist peptides are synthesized by automated peptide synthesizers.


Example 9—Biological Production of Custom Derivative γc-Antagonist Peptides Using Recombinant Technology

A custom derivative γc-antagonist peptides is synthesized biologically as a pro-peptide that consists of an appropriate tagging peptide, a signal peptide, or a peptide derived from a known human protein that enhances or stabilizes the structure of the BNZ-γ peptide or a peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof, and improves their biological activities. If desired, an appropriate enzyme-cleavage sequence proceeding to the N-terminus of the peptide shall be designed to remove the tag or any part of the peptide from the final protein.


A nucleotide sequence encoding the custom derivative peptide with a stop codon at the 3′ end is inserted into a commercial vector with a tag portion derived from thioredoxin of E. coli and a special peptide sequence that is recognized and digested by an appropriate proteolytic enzyme (for example, enterokinase) intervening between the tag portion and the nucleotide sequence encoding the custom derivative peptide and stop codon. One example of a suitable vector is the pThioHis plasmid available from Invitrogen, Calif. Other expression vectors may be used.


Example 10—Conjugation of Custom Peptides and Derivative to Carrier Proteins for Immunization Purposes and Generation of Antibody Against the Custom Peptides

BNZ-γ and other custom derivative peptides, such as a peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof are used to immunize animals to obtain polyclonal and monoclonal antibodies. Peptides are conjugated to the N- or the C-terminus of appropriate carrier proteins (for example, bovine serum albumin, Keyhold Limpet Hemocyanin (KLH), etc.) by conventional methods using Glutaraldehyde or m-Maleimidobenzoyl-N-Hydroxysuccinimide Ester. The conjugated peptides in conjunction with an appropriate adjuvant are then used to immunize animals such as rabbits, rodents, or donkeys. The resultant antibodies are examined for specificity using conventional methods. If the resultant antibodies react with the immunogenic peptide, they are then tested for the ability to inhibit individual γc-cytokine activity according to the cellular proliferation assays described in Examples 1-3. Due to the composite nature of the derivative peptides it is possible to generate a single antibody that recognizes two different cytokines simultaneously, because of the composite nature of these peptides.


Example 11—Method for Large Scale Production of Custom Derivative γc-Antagonist Peptides

Recombinant proteins are produced in large scale by the use of cell-free system as described elsewhere. (See Takai et al., 2010 Curr. Pharm. Biotechnol. 11(3):272-8.) Briefly, cDNAs encoding the γc-antagonist peptide and a tag are subcloned into an appropriate vector (See Takai et al., 2010 Curr. Pharm. Biotechnol. 11(3):272-8), which is subjected to in vitro transcription, followed immediately by an in vitro translation to produce the tagged peptide. The pro-polypeptide is then purified using an immobilized antibody recognizing the tagged epitope, treated by the proteolytic enzyme and the eluate (which mostly contains the custom derivative peptide of interest) is tested for purity using conventional 18% Tricine-SDS-PAGE (Invitrogen) and conventional comassie staining. Should the desired purity of the peptide not be met (>98%), the mixture is subjected to conventional HPLC (high-performance liquid chromatography) for further purification.


Example 12—Use of Custom Derivative γc-Antagonist Peptides to Block Cytokine Function in HAM/TSP

HTLV-1-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) is a chronic progressive myelopathy seen in some people infected with Human T-Lymphotropic Virus Type I (HTLV-I). Infiltration of lymphocytes in the spinal cord is associated with the immune response to HTLV-I and results in the release of certain cytokines. Some of these cytokines may also damage nerves.


Patients with HAM/TSP show an elevated state of the immune system that is similar to that observed in autoimmune diseases (Oh et al. 2008 Neurol Clin. 26:781-785). This elevated state is demonstrated by the ability of HAM/TSP patient's T-cells to undergo spontaneous proliferation in an ex vivo culture for about a week in the absence of exogenously added cytokines. The spontaneous proliferation of T-cells in HAM/TSP patients is attributed, at least partly, to autocrine/paracrine loops of IL-2, IL-9, and IL-15. It has been shown that adding blocking antibody against the IL-2 or IL-15 receptors can inhibit spontaneous T-cell proliferation in a HAM/TSP ex vivo culture system.


These observations, along with other data derived from ex vivo studies, have provided the rationale for taking two monoclonal antibodies (an anti-IL-2 receptor alpha or anti-Tac and an anti-IL-15 receptor beta chain) into the clinic for treatment of HAM/TSP (Azimi et al. 2001 Proc. Natl. Acad. Sci. 98:14559-64, Azimi et al., 1999 J. Immunol 163:4064-72).


Anti-cytokine receptor antagonists according to the embodiments described herein, would not only be valuable as a therapeutic immuno-modulatory agent for treatment of HAM/TSP, but modulation of immune response in HAM/TSP by anti-cytokine receptor antagonists according to the present embodiments acts proof-of-concept for the use of the anti-cytokine receptor antagonists according to the present embodiments in the treatment of other auto-immune diseases.


To demonstrate the efficacy of custom derivative γc-antagonist peptides according to the embodiments described herein, we tested the ability of BNZ-γ peptide to block immune response to HTLV-1 in a spontaneous T-cell proliferation assay using a HAM/TSP ex vivo culture system. Proliferation assays were performed on HAM/TSP patient blood samples with and without the addition of BNZ-γ. These assays evaluated the ability of BNZ-γ to block the function of cytokines, such as IL-2 and IL-15, present in the ex vivo HAM/TSP patient blood culture and prevent spontaneous T-cell proliferation in these samples.


In an ex vivo spontaneous T-cell proliferation assay, PBMC from HAM/TSP patient was cultured at 1×10(6) cells per well of a 96 well plate in RPMI-10% FCS. Increasing concentrations of BNZ-γ peptide were added to each well. As a control, an irrelevant peptide was used in similar fashion. The cells were incubated in a 37° C. CO2 incubator for 3, 4, and 6 days. The amount of 1 uCi of 3H-thymidine was added to the cells. After an additional 6 hour incubation, cells were harvested their proliferation rate was measured. The data for a representative HAM/TSP patient is shown in FIG. 4A-FIG. 4D. As indicated in FIG. 4A-FIG. 4D, BNZ-γ peptide inhibits the spontaneous proliferation of T-cells in HAM/TSP culture at a concentration of about 1 ug/ml.


Other immunological markers were additionally measured in this assay. The percentage of the viral specific CD8 cells was measured during the ex vivo culture using viral protein tetramers. The population of CD4+CD25+ cells, a marker of T-cell activation, as well as Ki67 staining, a marker of T-cell proliferation, was monitored in a flow cytometry assay.


Other forms of the conjugated BNZ-γ peptide derivative or a custom peptide comprising the sequence of SEQ ID NO: 3, and a derivative thereof can be used in a similar future assay. They include albumin, BSA, PEG that can be conjugated to the peptide after chemical synthesis. Other biological forms of custom peptides such as the BNZ-γ peptide conjugate or a custom peptide comprising the sequence of SEQ ID NO: 3, and a derivative thereof may include regions of known protein entities (including but not limited to Fc region of human IgG) that are fused to the custom peptides.


Example 13—Method of Treating Adult T-Cell Leukemia (ATL) in a Human Patient by Administration of Custom Derivative γc-Antagonist Peptide

A human patient suffering from Adult T-cell Leukemia is identified. An effective dose, as determined by the physician, of custom derivative γc-antagonist peptide, for example, BNZ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient enters remission.


Example 14—Method of Treating HAM/TSP in a Human Patient by Administration of Custom Derivative γc-Antagonist Peptide

A human patient suffering from HAM/TSP is identified. An effective dose, as determined by the physician, of custom derivative γc-antagonist peptide, for example, BNZ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient's symptoms improve or if the progression of the disease has been stopped or slowed down.


Example 15—Use of Custom Derivative γc-Antagonist Peptides to Block Cytokine Function

A human patient suffering who is in need of reducing the function of at least IL-15 and IL-21 is identified. An effective dose, as determined by the physician, of custom derivative γc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient's symptoms improve or if the progression of the disease has been stopped or slowed down.


Example 16—Method of Treating Celiac Disease in a Human Patient by Administration of Custom Derivative γc-Antagonist Peptide

A human patient suffering from Celiac disease is identified. An effective dose, as determined by the physician, of custom derivative γc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient's symptoms improve or if the progression of the disease has been stopped or slowed down.


REFERENCES



  • Antony, P. A., Paulos, C. M., Ahmadzadeh, M., Akpinarli, A., Palmer, D. C., Sato, N., Kaiser A., Heinrichs, C. S., Klebanoff, C. A., Tagaya, Y., and Restifo, N P., Interleukin-2-dependent mechanisms of tolerance and immunity in vivo. 2006 J. Immunol. 176:5255-66.

  • Azimi, N., Nagai, M., Jacobson, S., Waldmann, T. A., IL-15 plays a major role in the persistence of Tax-specific CD8 cells in HAM/TSP patients. 2001 Proc. Natl. Acad. Sci. 98:14559-64.

  • Azimi, N., Mariner J., Jacobson S., Waldmann T. A., How does interleukin 15 contribute to the pathogenesis of HTLV type-1 associated myelopathy/tropical spastic paraparesis? 2000 AIDS Res. Hum. Retroviruses 16:1717-22.

  • Azimi, N., Jacobson, S., Leist, T., Waldmann, T. A., Involvement of IL-15 in the pathogenesis of human T lymphotropic virus type-I-associated myelopathy/tropical spastic paraparesis: implications for therapy with a monoclonal antibody directed to the IL-2/15R beta receptor. 1999 J. Immunol. 163:4064-72.

  • Azimi, N., Brown, K., Bamford, R. N., Tagaya, Y., Siebenlist, U., Waldmann, T. A., Human T cell lymphotropic virus type I Tax protein trans-activates interleukin 15 gene transcription through an NF-kappaB site. 1998 Proc. Natl. Acad. Sci. USA 95:2452-7.

  • Bazan, J. F., Hematopoietic receptors and helical cytokines. 1990 Immunol. Today 11:350-354.

  • Bettini, M., and Vignali, D. A., Regulatory T cells and inhibitory cytokines in autoimmunity. 2009 Curr. Opin. Immunol. 21:612-8.

  • Bodd, M., Raki, M., Tollefsen, S., Fallang, L. E., Bergseng, E., Lundin, K. E., Sollid, L. M., HLA-DQ2-restricted gluten-reactive T cells produce IL-21 but not IL-17 or IL-22. 2010 Mucosal Immunol. 3:594-601.

  • De Rezende, L. C., Silva I. V., Rangel, L. B., Guimaraes, M. C., Regulatory T cells as a target for cancer therapy. 2010 Arch. Immunol. Ther. Exp. 58:179-90.

  • Dubois, S., Mariner, J., Waldmann, T. A., Tagaya, Y., IL-15Ralpha recycles and presents IL-15 In trans to neighboring cells. 2002 Immunity 17:537-47.

  • Dodge D L. Et al., IL-2 and IL-12 alter NK cell responsiveness to IFN-gamma-inducible protein 10 by down-regulating CXCR3 expression. J. Immun. 168:6090-8.

  • Fehniger, T. A., Suzuki, K., Ponnappan, A., VanDeusen, J. B., Cooper, M. A., Florea, S. M., Freud, A. G., Robinson, M. L., Durbin, J., Caligiuri, M. A., Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells. 2001 J. Exp. Med. 193:219-31.

  • Fisher, A. G., Burdet, C., LeMeur, M., Haasner, D., Gerber, P., Cerediq, R., Lymphoproliferative disorders in an IL-7 transgenic mouse line. 1993 Leukemia 2:S66-68.

  • Gong J H, et al. Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells Leukemia 8: 652-658, 1994.

  • Hennighausen, L., Robinson, G. W., Interpretation of cytokine signaling through the transcription factors STAT5A and STAT5B. 2008 Genes Dev. 22:711-21.

  • Klingemann H G. et al. A cytotoxic NK-cell line (NK-92) for ex vivo purging of leukemia from blood. Biol. Blood Marrow Transplant. 2: 68-75, 1996.

  • Krause, C. D, and Pestka, S., Evolution of the Class 2 cytokines and receptors, and discovery of new friends and relatives. 2005 Pharmacol, and Therapeutics 106:299-346.

  • Kundig, T. M., Schorle, H., Bachmann, M. F., Hengartener, H., Zinkernagel, R. M., Horak, I., Immune Responses of the interleukin-2-deficient mice. 1993 Science 262:1059-61.

  • Le Buanec, H., Paturance, S., Couillin, I., Schnyder-Candrian, S., Larcier, P., Ryffel, B., Bizzini, B., Bensussan, A., Burny, A., Gallo, R., Zagury, D., Peltre, G., Control of allergic reactions in mice by an active anti-murine IL-4 immunization. 2007 Vaccine 25:7206-16.

  • Littman, D. R., Rudensky, A Y., Th17 and regulatory T cells in mediating and restraining inflammation. 2010 Cell 140(6):845-58.

  • Miyagawa, F., Tagaya, Y., Kim, B. S., Patel, H. J., Ishida, K., Ohteki, T., Waldmann, T. A., Katz, S. I., IL-15 serves as a costimulator in determining the activity of autoreactive CD8 T cells in an experimental mouse model of graft-versus-host-like disease. 2008 J. Immunol. 181:1109-19.

  • Noguchi, M., Yi, H., Rosenblatt, H. M., Filipovich, A. H., Adelstein, S., Modi, W. S., McBride, O. W., Leonard, W. J., Interleukin 2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans. 1993 Cell 73:147-157.

  • OH, U., Jacobson S., Treatment of HTLV-I-Associated Myelopathy/Tropical Spastic Paraparesis: Towards Rational Targeted Therapy 2008 Neurol Clin. 2008 26: 781-785.

  • Orzaez, M., Gortat, A., Mondragon, L., Perez-Paya, E., Peptides and Peptide Mimics as Modulators of Apototic Pathways. 2009 Chem. Med. Chem. 4:146-160.

  • O'Shea, J. J., Targeting the Jak/STAT pathway for immunosuppression. 2004 Ann. Rheum. Dis. 63:(suppl II): ii67-71.

  • Paul, W. E., Pleiotropy and redundancy: T cell-derived lymphokines in the immune response. 1989 Cell 57:521-4.

  • Pesu M, Candotti F, Husa M, Hofmann S R, Notarangelo L D, and O'Shea J J. Jak3, severe combined immunodeficiency, and a new class of immunosuppressive drugs. 2005 Immunol. Rev. 203:127-142.

  • Pesu, M., Laurence, A., Kishore, N., Zwillich, S., Chan, G., O'Shea, J. J., Therapeutic targeting of Janus kinases. Immunol. 2008 Rev. 223:132-142.

  • Rochman, Y., Spolski, R., Leonard, W. J., New Insights into the regulation of T cells by gamma c family cytokines. 2009 Nat. Rev. Immunol. 9:480-90.

  • Sakaguchi, S., Yamaguchi, T., Nomura, T., One, M., Regulatory T cells and immune tolerance. 2008 Cell 133: 775-87.

  • Sato, N., Sabzevari, H., Fu, S., Ju, W., Bamford, R. N., Waldmann, T. A., and Tagaya, Y., Development of an IL-15-Autocrine CD8 T-cell Leukemia in IL-15 Transgenic mice requires the cis-expression of IL-15R alpha. Blood 2011 in press.

  • Sugamura, K., Asao, H., Kondo, M., Tanaka, N., Ishii, N., Nakamura, M., Takeshita, T., The common gamma-chain for multiple cytokine receptors. 1995 Adv. Immunol. 59: 225-277.

  • Sugamura, K., Asao, H., Kondo, M., Tanaka, N., Ishii, N., Ohbo, K., Nakamura, M., Takeshita. T., The interleukin-2 receptor gamma chain: its role in the multiple cytokine receptor complexes and T cell development in XSCID. 1996 Annu. Rev. Immunol. 14:179-205.

  • Tagaya, Y., Burton, J. D., Miyamoto, Y., Waldmann, T A., Identification of a novel receptor/signal transduction pathway for IL-15/T in mast cells. 1996 EMBO J. 15:4928-39.

  • Tagaya, Y., Memory CD8 T cells now join “Club 21”. 2010 J. Leuk. Biol. 87:13-15.

  • Takai, K., Sawasaki, T., and Endo, Y. The Wheat-Germ Cell-Free Expression System, 2010 Curr. Pharm. Biotechnol. 11:272-8.

  • Tanaka, T., et al., A novel monoclonal antibody against murine IL-2 receptor beta-chain. Characterization of receptor expression in normal lymphoid cells and EL-4 cells. 1991 J. Immunol. 147:2222-28.

  • Takeshita, T., Asao, H., Ohtani, K., Ishii, N., Kumaki, S., Tanaka, N., Manukata, H., Nakamura, M., Sugamura, K., Cloning of the Gamma chain of the Human IL2 receptor. 1992 Science 257:379-382.

  • Waldmann, T. A., Anti-Tac (daclizumab, Zenapax) in the treatment of leukemia, autoimmune diseases, and in the prevention of allograft rejection: a 25-year personal odyssey. 2007 J. Clin. Immunol. 27: 1-18.


Claims
  • 1. A nucleotide sequence encoding a composite peptide that comprises amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises an amino acid sequence having at least 70% sequence identity to the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), wherein the composite peptide inhibits the activity of IL-15 and IL-21.
  • 2. The nucleotide sequence of claim 1, wherein the nucleotide sequence is selected from the group consisting of dsDNA, ssDNA, dsRNA, and ssRNA.
  • 3. The nucleotide sequence of claim 1, wherein the composite peptide consists of the amino acid sequence of SEQ ID NO: 3.
  • 4. A pharmaceutical composition comprising: a nucleotide sequence encoding a peptide conjugate, or a derivative thereof; anda pharmaceutically acceptable carrier, diluent, excipient or combination thereof;wherein the peptide conjugate or the derivative thereof modulates the activity of IL-15 and IL-21;wherein the peptide conjugate or the derivative thereof comprises an amino acid sequence having at least 70% sequence identity to P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).
  • 5. The pharmaceutical composition of claim 4, wherein the nucleotide sequence further comprises a sequence encoding a signal peptide.
  • 6. The pharmaceutical composition of claim 4, wherein the nucleotide sequence is selected from the group consisting of dsDNA, ssDNA, dsRNA, and ssRNA.
  • 7. The pharmaceutical composition of claim 4, wherein the peptide conjugate consists of the amino acid sequence of SEQ ID NO: 3.
  • 8. A method of treating a γc-cytokine-mediated disease, the method comprising administering the pharmaceutical composition of claim 4 to a subject in need of treatment, wherein the γc-cytokine-mediated disease is selected from the group consisting of myasthenia gravis, inflammatory bowel disease, CD4-leukemia, CD8-leukemia, LGL-leukemia, systemic lupus erythematosus, Sjögren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, and graft-versus-host disease (GvHD).
  • 9. A method of treating uveitis, arthropathy, dermatitis, exocrinopathy, and myositis, the method comprising administering the pharmaceutical composition of claim 4 to a subject in need of treatment.
  • 10. A method of treating an inflammatory respiratory disease, the method comprising administering the pharmaceutical composition of claim 4 to a subject in need of treatment, wherein the inflammatory respiratory disease is selected from the group consisting of sinusitis, bronchitis, chronic obstructive pulmonary disease (COPD), and lung fibrosis.
  • 11. A method of treating a cosmetic condition, the method comprising administering the pharmaceutical composition of claim 4 to a subject in need of treatment, wherein the cosmetic condition is selected from the group consisting of acne, hair loss, and sunburn.
  • 12. A genetic construct comprising: a nucleotide sequence encoding a composite peptide; andat least one regulatory element, wherein the at least one regulatory element is configured to regulate expression of the composite peptide,wherein the composite peptide, when expressed, can modulate the activity of IL-15 and IL-21;wherein the composite peptide comprises an amino acid sequence having at least 70% sequence identity to P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).
  • 13. The genetic construct of claim 12, wherein the genetic construct is a DNA.
  • 14. The genetic construct of claim 12, wherein the genetic construct is an RNA.
  • 15. The genetic construct of claim 12, wherein the composite peptide consists of the amino acid sequence of SEQ ID NO: 3.
  • 16. The composite peptide encoded by the nucleotide sequence of claim 1.
  • 17. The composite peptide of claim 16, wherein the composite peptide is conjugated to one or more additional moieties at the N-terminus, C-terminus, or a side residue of the composite peptide.
  • 18. The composite peptide of claim 17, wherein the one or more additional moieties is selected from the group consisting of albumin, Keyhold Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell-specific antigen, a receptor, a ligand, a metal ion, and Poly Ethylene Glycol (PEG).
  • 19. The composite peptide of claim 17, wherein the one or more additional moieties comprises Poly Ethylene Glycol (PEG).
  • 20. A method for nail maintenance, the method comprising administering the pharmaceutical composition of claim 4 to a subject in need of nail maintenance.
PRIORITY AND CROSS REFERENCE TO RELATED APPLICATIONS

This application is the U.S. National Stage Application under 35 U.S.C. § 371 of International Application No. PCT/US2016/055845, filed Oct. 6, 2016, designating the U.S. and published in English as WO 2017/062685 A1 on Apr. 13, 2017, which claims the benefit of U.S. Provisional Application No. 62/239,761, filed Oct. 9, 2015, which are hereby incorporated by reference in its entireties.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2016/055845 10/6/2016 WO 00
Publishing Document Publishing Date Country Kind
WO2017/062685 4/13/2017 WO A
US Referenced Citations (96)
Number Name Date Kind
4518584 Mark et al. May 1985 A
5700913 Taniguchi et al. Dec 1997 A
5795966 Grabstein et al. Aug 1998 A
6013480 Grabstein et al. Jan 2000 A
6028186 Tasset et al. Feb 2000 A
6127387 Huang et al. Oct 2000 A
6168783 Grabstein et al. Jan 2001 B1
6261559 Levitt et al. Jul 2001 B1
6307024 Novak et al. Oct 2001 B1
6323027 Burkly et al. Nov 2001 B1
6686178 Novak et al. Feb 2004 B2
6770745 Burkly et al. Aug 2004 B2
6793919 Mohler Sep 2004 B2
6797263 Strom et al. Sep 2004 B2
6811780 Furfine et al. Nov 2004 B2
6838433 Serlupi-Crescenzi Jan 2005 B2
6955807 Shanafelt et al. Nov 2005 B1
7105653 Shanafelt et al. Sep 2006 B2
7148333 Cox, III Dec 2006 B2
7192578 Levitt et al. Mar 2007 B2
7235240 Grabstein et al. Jun 2007 B2
7314623 Grusby et al. Jan 2008 B2
7347995 Strom et al. Mar 2008 B2
7423123 Boisvert et al. Sep 2008 B2
7473765 Novak et al. Jan 2009 B2
7632814 Hazlehurst et al. Dec 2009 B2
7645449 Stassi et al. Jan 2010 B2
7700088 Levitt et al. Apr 2010 B2
7731946 Grusby et al. Jun 2010 B2
7785580 Pan et al. Aug 2010 B2
7786072 Verdine et al. Aug 2010 B2
7910123 McKay Mar 2011 B2
7959908 Nelson et al. Jun 2011 B2
8110180 Novak et al. Feb 2012 B2
8211420 Bondensgaard Jul 2012 B2
8455449 Tagaya Jun 2013 B2
8512946 Mirkin Aug 2013 B2
9133243 Tagaya Sep 2015 B2
9133244 Tagaya Sep 2015 B2
9675672 Tagaya Jun 2017 B2
9951105 Tagaya Apr 2018 B2
9959384 Azimi et al. May 2018 B2
10030058 Azimi Jul 2018 B2
10030059 Azimi Jul 2018 B2
10227382 Tagaya Mar 2019 B2
10808009 Tagya et al. Oct 2020 B2
10854312 Azimi et al. Dec 2020 B2
20020114781 Strom et al. Aug 2002 A1
20030049798 Carter et al. Mar 2003 A1
20030108549 Carter et al. Jun 2003 A1
20040009150 Nelson et al. Jan 2004 A1
20040126900 Barry Jul 2004 A1
20040136954 Grusby et al. Jul 2004 A1
20050124044 Cunningham et al. Jun 2005 A1
20060034892 Ueno Feb 2006 A1
20060039902 Young et al. Feb 2006 A1
20060236411 Dreher et al. Oct 2006 A1
20060263857 Lefrancois et al. Nov 2006 A1
20070048266 Nelson Mar 2007 A1
20070122413 Sivakumar et al. May 2007 A1
20080038275 Martin Feb 2008 A1
20080108552 Hazlehurst et al. May 2008 A1
20080166338 Leonard Jul 2008 A1
20090136511 Santos Savio et al. May 2009 A1
20090148403 Bosivert et al. Jun 2009 A1
20090253864 Peschke et al. Oct 2009 A1
20090258357 Ruebn Oct 2009 A1
20100099742 Stassi Apr 2010 A1
20100135958 Hwu Jun 2010 A1
20100196309 Bondensgaard et al. Aug 2010 A1
20100266531 Hsieh Oct 2010 A1
20110081327 Nicolette Apr 2011 A1
20110091515 Zilberman Apr 2011 A1
20110142833 Young Jun 2011 A1
20110245090 Abbas Oct 2011 A1
20110311475 Borte Dec 2011 A1
20120329728 Tagaya Dec 2012 A1
20130052127 Sasaki Feb 2013 A1
20130095102 Levin Apr 2013 A1
20130217858 Bioniz Aug 2013 A1
20160000877 Bioniz Jan 2016 A1
20160306918 Azimi Oct 2016 A1
20170051015 Bioniz Feb 2017 A1
20170101452 Bioniz Apr 2017 A1
20170204153 Tagaya Jul 2017 A1
20170240607 Azimi Aug 2017 A1
20180125941 Greve May 2018 A1
20180237475 Tagaya Aug 2018 A1
20180258174 Mortier et al. Sep 2018 A1
20180349550 Azimi et al. Dec 2018 A1
20190070263 Azimi Mar 2019 A1
20190194255 Tagaya Jun 2019 A1
20200347128 Tagaya et al. Nov 2020 A1
20200399316 Tagaya et al. Dec 2020 A1
20210082537 Azimi et al. Mar 2021 A1
20210324029 Doerr et al. Oct 2021 A1
Foreign Referenced Citations (41)
Number Date Country
1478098 Feb 2004 CN
1703423 Nov 2005 CN
103501805 Jan 2014 CN
1525213 Apr 2005 EP
2665486 Nov 2013 EP
2004-525076 Aug 2004 JP
2005-508179 Mar 2005 JP
WO 8702990 May 1987 WO
WO 200072864 Dec 2000 WO
WO 2003040313 May 2003 WO
WO 03087320 Oct 2003 WO
WO 2003103589 Dec 2003 WO
WO 2004084835 Oct 2004 WO
WO 2005014642 Feb 2005 WO
WO 2005030196 Apr 2005 WO
WO 2005067956 Jul 2005 WO
WO 2005105830 Nov 2005 WO
WO 2005112983 Dec 2005 WO
WO 2006105538 May 2006 WO
WO 2006111524 Oct 2006 WO
WO 2006113331 Oct 2006 WO
WO 2008049920 Feb 2008 WO
WO 2009100035 Aug 2009 WO
WO 2009108341 Sep 2009 WO
WO 2009132821 Nov 2009 WO
WO 2010011313 Jan 2010 WO
WO 2010039533 Apr 2010 WO
WO 2010054667 May 2010 WO
WO 2010076339 Jul 2010 WO
WO 2010103038 Sep 2010 WO
WO 2010133828 Nov 2010 WO
WO 2011070214 Jun 2011 WO
WO 2011133948 Oct 2011 WO
WO 2012006585 Jan 2012 WO
WO 2012012531 Jan 2012 WO
WO 2012099886 Jul 2012 WO
WO 2012175222 Dec 2012 WO
WO 2015089217 Jun 2015 WO
WO 2015089217 Jun 2015 WO
WO 2017062685 Apr 2017 WO
WO 2018187499 Oct 2018 WO
Non-Patent Literature Citations (286)
Entry
Seffernick et al., J. Bacteriol., 2001, 183: 2405-2410.
Witkowski et al., Biochemistry, 1999, 38: 11643-11650.
Nozuma et al., Journal of NeuroVirology, 2020, 26: 652-663.
Nata et al., JBC, 2015, 290: 22338-22351.
Boraschi et al., Encyclopedia of Endocrine Diseases, 2004.
Rajaei et al., Med. Microbiol. Immunol., 2017, 206: 195-201.
Enose-Akahata et al., Annals of Clinical and Translational Neurology, 2019, 6: 1383-1394.
Venkayeshaiah et al., J. Allergy Clin. Immunol., 2017, 141:906-917.
Wu et al., Biochem. Biophys. Res. Commun., 2018, 501: 92-99.
Aoi et al., J. Allergy Clin. Immunol., 2006, 117: 1359-1366.
Hiromura et al., J. Immunol., 2007, 179: 7157-7165.
Cagdas et al., J. Clin. Immunol., 2021, 41: 1272-1290.
Pepper et al., Nat. Immunol., 2010, 11:83-89.
Crane et al., Aging Cell, 2015, 14: 625-634.
Huang et al., FASEB J., 2008, 22: 2142-2150.
Mayo Clinic, 2021.
Antony, et al., “lnterleukin-2-Dependent Mechanisms of Tolerance and Immunity In Vivo,” J. Immunol. 176: 5255-5266, 2006.
Azimi, N., “Human T Cell Lymphotropic Virus Type I Tax Protein Trans-Activates Interleukin 15 Gene Transcription Through an NF-kappaB Site,” Proc. Natl. Acad. Sci. USA 95:2452-2457, 1998.
Azimi, N., “Involvement of IL-15 In The Pathogenesis of Human T Lymphotropic Virus Type-I-Associated Myelopathy/Tropical Spastic Paraparesis: Implications for Therapy with a Monoclonal Antibody Directed to the IL-2/15Rbeta Receptor,” J. Immunol. 163:4064-4072, 1999.
Azimi, N., et al., “How Does Interleukin 15 Contribute to the Pathogenesis of HTLV Type-1 Associated Myelopathy/Tropical Spastic Paraparesis?” AIDS Res. Hum. Retroviruses 16:1717-1722, 2000.
Azimi, N., et al., “IL-15 Plays a Major Role in the Persistence of Tax-specific CD8 Cells in HAM/TSP patients,” Proc. Natl. Acad. Sci. 98:14559-14564, 2001.
Bazan, J.F., “Hematopoietic Receptors and Helical cytokines,” Immunol. Today 11:350-354, 1990.
Bernard et al., dentification of an Interleukin-15α Receptor-binding Site on Human Interleukin-15*, The Journal of Biological Chemistry (2004)279:24313-24322.
Bettini, M., and D.A. Vignali, “Regulatory T Cells and Inhibitory Cytokines in Autoimmunity,” Curr. Opin. Immunol. 21:612-618, 2009.
Bodd, M., et al., “HLA-DQ2-Restricted Gluten-Reactive T cells Produce IL-21 but not IL-17 or IL-22,” Mucosal Immunol. 3:594-601, 2010.
Bonsch, et al. Species-specific Agonist/Antagonist Activities of Human Interleukin-4 Variants Suggest Distinct Ligand Binding Properties of Human and Murine Common Receptor γChain*, The Journal of Biological Chemistry (1995) 270:8452-8457.
De Rezende, L.C., et al., “Regulatory T Cells as a Target for Cancer Therapy,” Arch. Immunol. Ther. Exp. 58:179-190, 2010.
Decision of Rexamination in Chinese Application 201280010348.8.
Definition of composite from www.merriam-sebster.com/dictionary/composite, pp. 1-5. Accessed Feb. 17, 2015.
Definition of derivative from http://cancerweb.ncl.ac.uk/omd/about.html, pp. 1-5. Accessed Jul. 7, 2005.
Dubois, S., et al., “IL-15R alpha Recycles and Presents IL-15 In Trans to Neighboring Cells,” Immunity 17:537-547, 2002.
Examination Report dated Nov. 14, 2018 in Australian Patent Application No. 2016334085.
Extended EP Search report dated May 22, 2014 for PCT/US2012/021566.
Fehniger, T.A., “Fatal Leukemia in Interleukin 15 Transgenic Mice Follows Early Expansions in Natural Killer and Memory Phenotype CD8+ T Cells,” J. Exp. Med. 193:219-231, 2001.
Final Office Action dated Feb. 24, 2015 for U.S. Appl. No. 13/868,725.
Final Office Action dated Nov. 29, 2016 for JP Patent Application 2013-550541.
Final Office Action dated Dec. 13, 2017for U.S. Appl. No. 15/585,666.
Fisher, A.G. et al., “Lymphoproliferative disorders in an IL-7 transgenic mouse line,” Leukemia 2, 1993, pp. 66-68.
Gong, J.H. et al., Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells. Leukemia 8, 1994, pp. 652-658.
Hennighausen, L., and G.W. Robinson, “Interpretation of Cytokine Signaling Through the Transcription Factors STAT5A and STAT5B,” Genes Dev. 22:711-721, 2008.
Hines, L et at. Interleukin 15, partial [synthetic construct]. NCBI PDS Accession No. AAX36174, interleukin 15, partial [synthetic construct]. Submitted Jan. 5, 2005; downloaded from the internet <https://www.ncbi.nlm.nih.gov/protein/60811495/> on Dec. 14, 2016, p. 1.
Hodge, D.L., et al., IL-2 and IL-12 Alter NK Cell Responsiveness to IFN-Gamma-Inducible Protein 10 by Down-Regulating CXCR3 Expression, J. Immun. 168:6090-6098, 2002.
International Preliminary Report on Patentability dated Jul. 23, 2013 for PCT/US2012/021566.
International Preliminary Report on Patentability dated Jun. 14, 2016 for PCT/US2012/062870.
International Preliminary Report on Patentability dated Apr. 10, 2018 for PCT/US2016/055845.
International Search Report and Written Opinion dated Jan. 17, 2017 for PCT/US2016/055845.
International Search Report and Written Opinion of the International Searching Authority in International Application No. PCT/US2012/021566 dated May 10, 2012.
International Search Report dated Jun. 26, 2015 for PCT/US14/69597.
International Search Report and Written Opinion dated Aug. 23, 2018 for PCT/US2018/026125.
Klingemann, H.G. et al., “A cytotoxic NK-cell line (NK-92) for ex vivo purging of leukemia from blood,” Blood Marrow Transplant 2, 1996, pp. 68-75.
Krause, C.D. and S. Pestka, “Evolution of the Class 2 Cytokines and Receptors, and Discovery of New Friends and Relatives,” Pharmacol, and Therapeutics 106:299-346, 2005.
Kundig, T.M. et al. “Immune Responses of the interleukin-2-deficient mice,” Science 262, 1993, pp. 1059-1061.
Le Buanec, H., et al., “Control of Allergic Reactions in Mice by an Active Anti-Murine IL-4 Immunization,” Vaccine 25:7206-7216, 2007.
Littman, D.R., and A.Y. Rudensky, “Th17 and Regulatory T Cells in Mediating and Restraining Inflammation,” Cell 140(6):845-858, 2010.
Miyagawa, F., et al., “IL-15 Serves as a Costimulator in Determining the Activity of Autoreactive CD8 T Cells in an Experimental Mouse Model of Graft-Versus-Host-Like Disease,” J. Immunol. 181:1109-1119, 2008.
NCBI Accession No. ABF82250, Accessed Aug. 11, 2014.
NCBI Accession No. BAA96385, Accessed Aug. 11, 2014.
NCBI Accession No. NP_999580, Accessed Aug. 11, 2014.
NCBI Accession No. ACT78884, Accessed Aug. 11, 2014.
NCBI Accession No. NP_999288, Accessed Aug. 11, 2014.
Noguchi, M., et al., “Interleukin 2 Receptor Gamma Chain Mutation Results in X-linked Severe Combined Immunodeficiency in Humans,” Cell 73:147-157, 1993.
Notice of Acceptance dated Oct. 19, 2016 for AU Application 2012207456.
Notice of Allowance dated Feb. 19, 2013 for U.S. Appl. No. 13/589,017.
Notice of Allowance dated Feb. 6, 2017 for U.S. Appl. No. 14/852,240.
Notice of Allowance dated May 11, 2015 for U.S. Appl. No. 13/868,725.
Notice of Allowance dated May 4, 2015 for U.S. Appl. No. 13/980,305.
Notice of Allowance dated Dec. 15, 2017 in U.S. Appl. No. 15/179,900.
Notice of Allowance dated Dec. 19, 2017 for U.S. Appl. No. 15/103,804.
Notification of Allowance dated May 31, 2018 for CN Patent Application 201280010348.8.
Notice of Allowance dated Mar. 26, 2018 in U.S. Appl. No. 15/287,517.
Notice of Allowance dated Mar. 26, 2018 in U.S. Appl. No. 15/585,666.
Notice of Allowance dated Oct. 22, 2018 for U.S. Appl. No. 15/474,312.
Notification of Re-examination dated Mar. 24, 2017 for CN Patent Application 201280010348.8.
O'Shea, J.J., “Targeting the Jak/STAT Pathway for Immunosuppression,” Ann. Rheum. Dis. 63:(Suppl. II):ii67-71, 2004.
Office Action dated Aug. 18, 2014 for U.S. Appl. No. 13/868,725.
Office Action dated Aug. 3, 2017for U.S. Appl. No. 15/103,804.
Office Action dated Aug. 8, 2017for U.S. Appl. No. 15/585,666.
Office Action dated Jan. 26, 2016 for JP Patent Application 2013-550541.
Office Action dated Jul. 2, 2014 for corresponding CN Application 201280010348.8.
Office Action dated Jul. 24, 2017 in U.S. Appl. No. 15/179,900.
Office Action dated Jul. 4, 2016 for Chinese Patent Application No. 201280010348.8.
Office Action dated Nov. 12, 2014 for U.S. Appl. No. 13/980,305.
Office Action dated Oct. 26, 2016 in EP Patent Application No. 12736203.6.
Office Action dated Oct. 18, 2014 for U.S. Appl. No. 13/868,725.
Office Action dated Oct. 22, 2015 for AU Application 2012207456.
Office Action dated Apr. 28, 2015 for Chinese Patent Application No. 201280010348.8.
Office Action dated Oct. 4, 2017 in European Patent Application No. 12736203.6.
Office Action dated Oct. 23, 2017 in U.S. Appl. No. 15/287,517.
Office Action dated Nov. 22, 2017 in Canadian Application No. 2,824,51.
Office Action dated Feb. 9, 2018 in Australian Application No. 2017200489.
Office Action dated Feb. 20, 2018 for JP Patent Application 2013-550541.
Office Action dated May 8, 2018 in JP Patent Application No. 2017-90501.
Office Action dated Jun. 22, 2018 for U.S. Appl. No. 15/474,312.
Office Action dated Nov. 23, 2018 in Canadian Application No. 2,824,51.
Office Action dated Nov. 26, 2018 in European Patent Application No. 12736203.6.
Oh, U., and S. Jacobson, “Treatment of HTLV-I-Associated Myelopathy/Tropical Spastic Paraparesis: Towards Rational Targeted Therapy,” Neurol. Clin. 26:781-785, 2008.
Orzaez, M., et al., “Peptides and Peptide Mimics as Modulators of Apoptotic Pathways,” Chem. Med. Chem. 4:146-160, 2009.
Paul, W.E., “Pleiotropy and Redundancy: T Cell-Derived Lymphokines in the Immune Response,” Cell 57:521-524, 1989.
Pesu, M., “Jak3, Severe Combined Immunodeficiency, and a New Class of Immunosuppressive Drugs,” Immunol. Rev. 203:127-142, 2005.
Pesu, M., Laurence, et al., “Therapeutic Targeting of Janus Kinases,” Immunol. Rev. 223:132-142, 2008.
Restriction Requirement dated Feb. 28, 2017 for U.S. Appl. No. 15/179,900.
Restriction Requirement dated Jul. 25, 2017 in U.S. Appl. No. 15/287,517.
Restriction Requirement dated Jun. 24, 2014 for U.S. Appl. No. 13/980,305.
Restriction Requirement dated Mar. 24, 2017 for U.S. Appl. No. 15/103,804.
Restriction Requirement dated May 9, 2014 for U.S. Appl. No. 13/868,725.
Restriction Requirement dated Jan. 26, 2018 for U.S. Appl. No. 15/474,312.
Rochman, Y., et al., “New Insights into the Regulation of T Cells by Gamma C Family Cytokines,” Nat. Rev. Immunol. 9:480-490, 2009.
Sakaguchi, S., et al., “Regulatory T Cells and Immune Tolerance,” Cell 133:775-787, 2008.
Sato, N., et al., “Development of an IL-15-Autocrine CD8 T-cell Leukemia in IL-15 Transgenic mice requires the cis-expression of IL-15R apha,” Blood 2011.
Sugamura, K., et al., “The Common Gamma-Chain for Multiple Cytokine Receptors,” Adv. Immunol. 59:225-277, 1995.
Sugamura, K., et al., “The lnterleukin-2 Receptor Gamma Chain: Its Role in the Multiple Cytokine Receptor Complexes and T Cell Development in XSCID,” Annu. Rev. Immunol. 14:179-205, 1996.
Supplementary European Search Report dated May 22, 2014 for European Application No. 12784848.9.
Tagaya, Y., “Memory CD8 T Cells Now Join Club 21,” J. Leuk. Biol. 87:13-15, 2010.
Tagaya, Y., et al., “Identification of a Novel Receptor/Signal Transduction Pathway for IL-15/T in Mast Cells,” EMBO J. 15:4928-4939, 1996.
Takai, K. et al., The Wheat-Germ Cell-Free Expression System, Curr. Pharm. Biotechnol. 11, 2010, pp. 272-278.
Takeshita, T., et al., “Cloning of the Gamma Chain of the Human IL-2 Receptor,” Science 257:379-382, 1992.
Tanaka, T., et al., “A Novel Monoclonal Antibody Against Murine IL-2 Receptor Beta-Chain. Characterization of Receptor Expression in Normal Lymphoid Cells and EL-4 Cells,” J. Immunol. 147:2222-2228, 1991.
Waldmann, T.A., Anti-Tac (daclizumab, Zenapax) in the Treatment of Leukemia, Autoimmune Diseases, and in the Prevention of Allograft Rejection: A 25-Year Personal Odyssey, J. Clin. Immunol. 27:1-18, 2007.
Water is naturally occurring from www.biology-online.org/dictionary/Water, pp. 1-3, Accesssed Apr. 24, 2014.
Extended European Search Report dated Mar. 28, 2019 in European Patent Application No. 16854367.6.
Office Action dated Apr. 23, 2019 in JP Patent Application No. 2017-90501.
Office Action dated Jun. 4, 2019 in JP Patent Application No. 2018-517887.
Office Action dated Aug. 16, 2019 for U.S. Appl. No. 15/957,806.
Office Action dated Sep. 9, 2019 for U.S. Appl. No. 15/964,717.
Yampolsky, L. et al., The Exchangeability of Amino Acids in Proteins, Genetics, 170, pp. 1459-1472, (2005).
Notice of Allowance dated Jul. 5, 2019 for EP Patent Application No. 12736203.6.
Office Action dated Sep. 25, 2019 for KR Patent Application No. 10-2018-7013183.
Office Action dated Oct. 28, 2019 for AU Patent Application No. 2016334085.
Olosz, F. et al. Structural Baisis for Binding Multiple Ligands by the Common Cytokine Receptor [gamma]-chain, Journal of Biological Chemistry, vol. 277, No. 14, pp. 12047-12052, (2002).
Office Action dated Jan. 31, 2019 in Canadian Patent Application No. 3,000,207.
Restriction Requirement dated Mar. 22, 2019 for U.S. Appl. No. 15/957,806.
Restriction Requirement dated Apr. 8, 2019 for U.S. Appl. No. 15/964,717.
Abadie et al., “IL-15: A central regulator of celiac disease immunopathology”. Immunol Rev. (2014) 260(1): 221-234.
Aringer et al., “Serum interleukin-15 is elevated in systemic lupus erythematosus”, Rheumatology (2001) 40(8):876-881.
Awwad et al., “Overview of Antibody Drug Delivery”. Pharmaceutics (2018) 10(3): 83.
Baranda et al., “IL-15 and IL-15R in leucocytes from patients with systemic lupus erythematosus.” Rheumatology (2005) 44(12): 1507-1513.
Ben Ahmed et al., “IL-15 renders conventional lymphocytes resistant to suppressive functions of regulatory T cells through activation of the phosphatidylinositol 3-kinase pathway”. J Immunol. (2009) 182(11):6763-6770.
Benahmed et al., “Inhibition of TGF-beta signaling by IL-15: a new role for IL-15 in the loss of immune homeostasis in celiac disease”. Gastroenter. (2007) 132(3):994-1008.
Blaser et al., “Donor-derived IL-15 is critical for acute allogeneic graft-versus-host disease”. Blood (2005) 105(2): 894-901.
Bla{hacek over (z)}ek et al., “The production and application of single-chain antibody fragments”. Folia Microbiol. (2003) 48(5): 687-698.
Bobbala et al., “Interleukin-15 plays an essential role in the pathogenesis of autoimmune diabetes in the NOD mouse”, Diabetologia (2012) 55: 3010-3020.
Borrego et al., “Recognition of human histocompatibility leukocyte antigen (HLA)-E complexed with HLA class I signal sequence-derived peptides by CD94/NKG2 confers protection from natural killer cell-mediated lysis”. J Exp Med. (1998) 187(5): 813-818.
Broux et al., “IL-15 amplifies the pathogenic properties of CD4+CD28− T cells in multiple sclerosis”. J Immunol. (2015) 194(5): 2099-2109.
Bubier et al., “A critical role for IL-21 receptor signaling in the pathogenesis of systemic lupus erythematosus in BXSB-Yaa mice.” Proc Natl Acad Sci U S A (2009) 106(5):1518-1523.
Bucher et al., “IL-21 blockade reduces graft-versus-host disease mortality by supporting inducible T regulatory cell generation”. 2009 114:5375-84.
Cantoni et al., “The activating form of CD94 receptor complex: CD94 covalently associates with the Kp39 protein that represents the product of the NKG2-C gene”. Eur J Immunol. (1998) 28: 327-338.
Caruso et al., “Involvement of interleukin-21 in the epidermal hyperplasia of psoriasis”. Nature Med. (2009) 15(( ): 1013-1015.
Caruso et al., “Pathogenic role of interleukin-21 in psoriasis.” Cell Cycle (2009) 8: 3629-3630.
Chen et al., “Insulin-dependent diabetes induced by pancreatic betacell expression of IL-15 and IL-15R alpha”, Proc Natl Acad Sci U S A (2013) 110:13534-13539.
Chen et al., “Induction of autoimmune diabetes in non-obese diabetic mice requires interleukin-21-dependent activation of autoreactive CD8+ T cells”. Clin Exp Immunol. (2013) 173(2): 184-194.
De Nitto et al., “Involvement of interleukin-15 and interleukin-21, two gamma-chain-related cytokines, in deiac disease”, World J Gastroenter. (2009) 15:4609-4614.
De Nitto et al. “Interleukin-21 triggers effector cell responses in the gut.” World J Gastroenterol. (2010) 16(29):3638-3641.
DePaolo et al., “Co-adjuvant effects of retinoic acid and IL-15 induce inflammatory immunity to dietary antigens”. Nature (2011) 471 (7337): 220-224.
Di Sabatino A. et al., “Epithelium derived interleukin 15 regulates intraepithelial lymphocyte Th1 cytokine production, cytotoxicity, and survival in coeliac disease”, Gut (2006) 55(4):469-477.
Fang et al., “Prophylactic effects of interleukin-2 receptor antagonists against graft-versus-host disease following unrelated donor peripheral blood stem cell transplantation”. Biol Blood Marrow Transplant. (2012) 18(5): 754-762.
Ferreira et al., “IL-21 production by CD4+ effector T cells and frequency of circulating follicular helper T cells are increased in type 1 diabetes patients”, Diabetologia (2015) 58: 781-790.
Frenzel et al., “Designing Human Antibodies by Phage Display”. Transfus Med Hemother. (2017) 44(5): 312-318.
Garrity et al., “The activating NKG2D receptor assembles in the membrane with two signaling dimers into a hexameric structure”. Proc Natl Acad Sci. (2005) 102(21): 7641-7646.
Ghalamfarsa et al., “IL-21 and IL-21 receptor in theimmunopathogenesis of multiple sclerosiS”, J Immunotoxicol. (2016) 13(3):274-285.
Gilhar et al., “Alopecia areata animal models illuminate autoimmune pathogenesis and novel immunotherapeutic strategies”. Autoimmun Rev. (2016) 15(7): 726-735.
Gonzalez-Alvaro et al., “Increased serum levels of interleukin-15 in rheumatoid arthritis with long-term disease.” Clin Exp Rheumatol. (2003) 21(5):639-642.
Groh et al., “Stimulation of T cell autoreactivity by anomalous expression of NKG2D and its MIC ligands in rheumatoid arthritis”. Proc Natl Acad Sci USA. (2003) 100(16):9452-9457.
Guo-Qiang et al., “Guided selection methods through chain shuffling”. Methods Mol Biol. (2009) 562(10): 133-142.
Habib T. et al., “IL-21: a novel IL-2-family lymphokine that modulates B,T, and natural killer cell responses”, J Allergy Clin Immunol. (2003) 112(6):1033-1045.
Hammers et al., “Antibody Phage Display: Technique and Applications”. J Invest Dermatol. (2014) 134(2): e17; 13 pages.
Harada et al., “Production of interleukin-7 and interleukin-15 by fibroblast-like synoviocytes from patients with rheumatoid arthritis.” Arthritis Rheum. (1999) 42(7):1508-1516.
Hessian P.A. et al., “Cytokine profile of the rheumatoid nodule suggests that it is a Th1 granuloma.” Arthritis Rheum. (2003) 48(2):334-338.
Hippen et al., “Blocking IL-21 signaling ameliorates xenogeneic GVHD induced by human lymphocytes”. Blood (2012) 119(2): 619-628.
Hodge et al., “IL-2 and IL-12 alter NK cell responsiveness to IFN-gamma-inducible protein 10 by down-regulating CXCR3 expression”. J Immunol. (2002) 168(12): 6090-6098.
Hong et al., Regulatory and pro-inflammatory phenotypes of myelin basic protein-autoreactive T cells in multiple sclerosis. Int Immunol. (2009) 21(12): 1329-1340.
Hüe et al., “A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease.” Immunity (2004) 21(3): 367-377.
Jabri B. et al., “Selective expansion of intraepithelial lymphocytes expressing the HLA-E-specific natural killer receptor CD94 in celiac disease”, Gastroenter. (2000) 118:867-879.
Jagielska et al., “Follow-up study of the first genome-wide association scan in alopecia areata: IL13 and KIAA0350 as susceptibility loci supported with genome-wide significance.” J Invest Dermatol. (2012) 132:2192-2197.
Jespers et al., “Guiding the selection of human antibodies from phage display repertoires to a single epitope of an antigen”. Biotechnology (1994) 12: 899-903.
Kang et al., Rational Design of Interleukin-21 Antagonist through Selective Elimination of the C Binding Epitope, J Biol Chem. (2010) 285(16): 12223-12231.
Kivisäkk et al., “IL-15 mRNA expression is up-regulated in blood and cerebrospinal fluid mononuclear cells in multiple sclerosis (MS)”, Clin Exp Immunol. (1998) 111(1):193-197.
Kluczyk et al., “The “two-headed” peptide inhibitors of interleukin-1 action,” Peptides, (200), 21: 1411-1420.
Köhler et al., “Continuous cultures of fused cells secreting antibody of predefined specificity”. Nature (1975) 256(5517): 495-497.
Kooy-Winkelaar et al., “CD4 T-cell cytokines synergize to induce proliferation of malignant and nonmalignant innate intraepithelial lymphocytes”, Proc Natl Acad Sci U S A (2017)114(6):E980-989.
Liu et al., “IL-15 is highly expressed in inflammatory bowel disease and regulates local T cell-dependent cytokine production.” J Immunol. (2000) 164(7):3608-3615.
Lonberg et al., “Human antibodies from transgenic mice”. Int Rev Immunol. (1995) 13: 65-93.
Maiuri L. et al., “Interleukin 15 Mediates Epithelial Changes in Celiac Desease”, Gastroenter. (2000) 119:996-1006.
McInnes et al., “Interleukin-15 mediates T cell-dependent regulation of tumor necrosis factor-α production in rheumatoid arthritis.” Nature Med. (1997) 3(2): 189-195.
Mention J-J. et al., “Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease”, Gastroenter. (2003) 125(3):730-745.
Mingari et al., “HLA class l-specific inhibitory receptors in human T lymphocytes: interleukin 15-induced expression of CD94/NK62A in superantigen- or alloantigen-activated CD8+ T cells”. Proc Natl Acad Sci. (1998) 95(3): 1172-1177.
Monteleone et al., “Interleukin-21 enhances T-helper cell type I signaling and interferon-γ production in Crohn's disease”, Gastroenterology (2005) 128(3): 687-694.
Monteleone et al., “Characterization of IL-17A—Producing cells in celiac disease mucosa”, J Immunol. (2010) 184: 2211-2218.
Nakou et al., “Interleukin-21 is increased in active systemic lupus erythematosus patients and contributes to the generation of plasma B cells.” Clin Exp Rheumatol. (2013) 31(2):172-179.
Nohra et al., RGMA and IL21R show association with experimental inflammation and multiple sclerosis. Genes & Immunity, (2010) 11(4): 279-293.
Nowak E.C. et al., “IL-9 as a mediator of Th17-driven inflammatory disease”, J Exp Med. (2009) 206:1653-1660.
Oppenheimer-Marks et al., “Interleukin 15 is produced by endothelial cells and increases the transendothelial migration of T cells In vitro and in the SCID mouse-human rheumatoid arthritis model In vivo.” J Clin Invest (1998) 101(6):1261-1272.
Pashenkov et al., “Levels of interleukin-15-expressing blood mononuclear cells are elevated in multiple sclerosis”, Scand J Immunol. (1999) 50(3):302-308.
Petukhova et al., “Genome-wide association study in alopecia areata implicates both innate and adaptive immunity.” Nature (2010) 466(7302):113-117.
Price-Troska et al., Inhibiting IL-2 Signaling And The Regulatory T-Cell Pathway Using Computationally Designed Peptides, Invest New Drugs, (2018) 37(1): 9-16.
Recher et al., “IL-21 is the primary common γ chain-binding cytokine required for human B-cell differentiation in vivo”. Blood (2011) 118(26): 6824-6835.
Richmond et al., “Antibody blockade of IL-15 signaling has the potential to durably reverse vitiligo”. Sci Transl Med. (2018) 10(450): 1-20.
Riechmann et al., “Reshaping human antibodies for therapy”. Nature (1988) 332: 323-327.
Roguska et al., “Humanization of murine monoclonal antibodies through variable domain resurfacing”. Proc Natl Acad Sci. (1994) 91(3): 969-973.
Rückert R. et al., “Interleukin-15 stimulates macrophages to activate CD4+ T cells: a role in the pathogenesis of rheumatoid arthritis?”, Immunology (2009) 126(1):63-73.
Saha et al.,“Prediction of continuous B-cell epitopes in an antigen using recurrent neural network”. Proteins (2006) 65: 40-48.
Saikali et al., “Contribution of astrocyte-derived IL-15 to CD8 T cell effector functions in multiple sclerosis”, J Immunol. (2010) 185(10):5693-5703.
Sarra M. et al., “IL-15 positively regulates IL-21 production in celiac disease mucosa”, Mucosal Immunol. (2013) 6(2):244-255.
Sawalha et al., “Genetic association of interleukin-21 polymorphisms with systemic lupus erythematosus”, Ann Rheum Dis. (2008) 67(4):458-461.
Schneider et al., “B cell-derived IL-15 enhances CD8 T cell cytotoxicity and is increased in multiple sclerosis patients”, J Immunol. (2011) 187(8):4119-4128.
Schumacher et al., “Severe combined immunodeficiencies of the common g-chain/JAK3 signaling pathway.” Isr. Med. Assoc. J. (2002) 4: 131-135.
Shultz et al., “Humanized mice for immune system investigation: progress, promise and challenges”. Nat Rev Immunol. (2012) 12(11): 786-798.
Studnicka et al., “Human-engineered monoclonal antibodies retain full specific binding activity by preserving non-CDR complementarity-modulating residues”. P rotein Eng (1994) 7:805-814.
Suarez-Farinas et al., “Alopecia areata profiling shows TH1, TH2, and IL-23 cytokine activation without parallel TH17/TH22 skewing.” J Allergy Clin Immunol. (2015) 136(5): 1277-1287.
Tang et al., “Cytosolic PLA2 is required for CTL-mediated immunopathology of celiac disease via NKG2D and IL-15.” J Exp Med. (2009) 206(3): 707-719.
Terrier et al., “Interleukin 21 Correlates with T Cell and B Cell Subset Alterations in Systemic Lupus Erythematosus”, J Rheumatol. (2012) 39(9):1819-1828.
Tomimatsu et al., “Antigen-specific in vitro immunization: a source for human monoclonal antibodies”. Methods Mol Biol. (2014) 1060 (Chapter 15): 297-307.
Tzartos et al., “IL-21 and IL-21 Receptor Expression in Lymphocytes and Neurons in Multiple Sclerosis Brain”, Am J Pathol. (2011) 178(2):794-802.
Vaknin-Dembinsky et al., “Membrane bound IL-15 is increased on CD14 monocytes in early stages of MS”, J Neuroimmunol. (2008) 195(1-2):135-139.
Van Heel et al., “A genome-wide association study for celiac disease identifies risk variants in the region harboring IL2 and IL21.” Nat Genet. (2007) 39(7): 827-829.
Vaughan et al., “Human Antibodies with Sub-nanomolar Affinities Isolated from a Large Non-immunized Phage Display Library”. Nature (1996) 14(3): 309-314.
Villadsen et al., “Resolution of psoriasis upon blockade of IL-15 biological activity in a xenograft mouse model”. J Clin Invest. (2003) 112: 1571-1580.
Waldmann, T.A., “The biology of IL-15: implications for cancer therapy and the treatment of autoimmune disorders.” J Investig Dermatol Symp Proc. (2013) 16(1):S28-S30.
Walensky L.D. et al., “Hydrocarbon-Stapled Peptides: Principles, Practice, and Progress”, Miniperspective; J Med Chem. (Aug. 2014) 57(15):6275-6288.
Williams et al., “Humanising Antibodies by CDR Grafting.”, in: Antibody Engineering, eds R. Kontermann and S. Dubel (Springer—Berlin, Heidelberg); (2010) Chapter 21: 319-339.
Xing et al., “Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition.” Nat Med. (2014) 9:1043-1049.
Xing et al., “Interleukin-21 induces migration and invasion of fibroblast-like synoviocytes from patients with rheumatoid arthritis”, Clin Exp Immunol. (May 2016) 184(2):147-158.
Xing et al., “Interleukin-21 Induces Proliferation and Proinflammatory Cytokine Profile of Fibroblast-like Synoviocytes of Patients with Rheumatoid Arthritis”, Scand J Immunol. (Jan. 2016) 83(1):64-71.
Yao et al., “SVMTriP: A Method to Predict Antigenic Epitopes Using Support Vector Machine to Integrate Tri-Peptide Similarity and Propensity”. PLoS One (2012) 7(9): e45152.
Zanzi et al., “IL-15 interferes with suppressive activity of intestinal regulatory T cells expanded in Celiac disease.” Am J Gastroenter (2011) 106(7): 1308-1317.
Zeng et al., “Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function.” J Exp Med. (2005) 201(1): 139-148.
Notice of Acceptance dated Jan. 14, 2019 in Australian Application No. 2017200489.
Office Action dated Jun. 6, 2020 in Australian Application No. 2019202527.
Office Action regarding National Genetic Patrimony/Traditional Knowledge, dated Feb. 11, 2020, in Brazilian Application No. 1120130180463.
Office Action dated Dec. 19, 2019 for Canadian Patent Application No. 2824515.
Office Action dated Oct. 27, 2015 for Chinese Patent Application No. 201280010348.8.
Extended European Search Report dated Feb. 10, 2020 in European Application No. 19206731.2.
Office Action dated Jun. 23, 2020 in Japenese Application No. 2017-090501.
Office Action dated Sep. 29, 2020 in Japenese Application No. 2019-152602.
Office Action dated May 27, 2020 in Australian Application No. 2020201174.
Office Action dated Feb. 4, 2020 in In Canadian Application No. 3000207.
Office Action dated Mar. 27, 2020 for European Application No. 16854367.6.
Office Action dated May 19, 2020 in JP Patent Application No. 2018-517887.
Office Action dated Nov. 11, 2020 in JP Patent Application No. 2018-517887.
Extended European Search Report dated Oct. 15, 2020 for Application No. 18780544.5.
International Search Report and Written Opinion dated Aug. 14, 2020 for PCT/US2020/030772.
Restriction Requirement dated Apr. 25, 2016 for U.S. Appl. No. 14/852,240.
Office Action dated Sep. 6, 2016 for U.S. Appl. No. 14/852,240.
Notice of Allowance dated Feb. 26, 2020 in U.S. Appl. No. 15/957,806.
Notice of Allowance dated Jun. 5, 2020 in U.S. Appl. No. 15/957,806.
Office Action dated Feb. 26, 2020 for U.S. Appl. No. 15/964,717.
Notice of Allowance dated Jul. 29, 2020 for U.S. Appl. No. 15/964,717.
Asano et al., “Molecular scanning of interleukin-21 gene and genetic susceptibility to type 1 diabetes.” Hum Immunol. (2007) 68(5):384-391.
Atwa et al., “T-helper 17 cytokines (interleukins 17, 21,22, and 6, and tumor necrosis factor-α) in patients with alopecia areata: association with clinical type and severity”. Int J Dermatol. (2016) 55(6):666-672.
Botti et al., “Psoriasis, from pathogenesis to therapeutic strategies: IL-21 as a novel potential thereapeutic target”. Curr Pharm Biotechnol. (2012) 13(10): 1861-1867.
Brumbaugh et al., “Clonotypic differences in signaling from CD94 (kp43) on NK cells lead to divergent cellular responses”. J Immunol. (1996) 157(7): 2804-2812.
Chik et al., “Elevated serum interleukin-15 level in acute graft-versus-host disease after hematopoietic cell transplantation.” J Pediatr Hematol Oncol. (2003) 25(12): 960-964.
Cox et al., “Immunoassay methods”, in Assay Guidance Manual [Internet], S. Markossian, G.S. Sittampalam, N.P. Coussens, H. Nelson, et al. [Eds.] (Bethesda, MD: Eli Lilly & Company and the National Center for Advancing Translational Sciences); 2004 Edition; (TOC only).
D'Auria et al., “Increased serum interleukin-15 levels in bullous skin diseases: correlation with disease intensity”. Arch Dermatol Res. (1999) 291: 354-356.
Fina et al., “Interleukin 21 contributes to the mucosal T helper cell type 1 resonse in coeliac disease”. Gut (2008) 57(7):887-892.
Fuentes-Duculan et al., “Biomarkers of alopecia areata disease activity and response to corticosteroid treatment”. Exp Dermatol. (2016) 4:282-286.
Grando et al., “Mediators of inflammation in blister fluids from patients with pemphigus vulgaris and bullous pemphigoid”. 1989 Arch Dermatol. (1989) 125:925-930.
He et al., “Elevated serum levels of interleukin 21 are associated with disease severity in patients with psoriasis.” Br J Dermatol. (2012) 167: 191-193.
Kuczynski et al., “IL-15 is elevated in serum patients with type 1 diabetes mellitus.” Diabetes Res Clin Pract. (2005) 69(3):231-236.
Laffleur et al., “Production of human or humanized antibodies in mice”. In Antibody Methods and Protocols by G. Proetzel et al.,[Ed.]. (2012) 901:149-159 (TOC only).
Lazetic et al., “Human natural killer cell receptors involved in MHC class I recognition are disulfide-linked heterodimers of CD94 and NKG2 subunits”. J Immunol. (1996) 157(11): 4741-4745.
Meresse et al., “The cytokine interleukin 21: a new player in coeliac disease?.” Gut (2008) 57(7): 879-881.
Padlan E.A., “A possible procedure for reducing the immunogenicity of antibody variable domains while preserving their ligand-binding properties”. Mol Immunol. (1991) 28(4-5): 489-498.
Schaller et al., “Interleukin-2 receptor expression and interleukin-2 production in bullous pemphigoid”. Arch Dermatol Res. (1990) 282(4): 223-226.
Sonntag et al., “Chronic graft-versus-host-disease in CD34(+)-humanized NSG mice is associated with human susceptibility HLA haplotypes for autoimmune diseases”. J Autoimmun. (2015) 62: 55-66.
Sushama et al., “Cytokine profile (IL-2, IL-6, IL-17, IL-22, and TNF-alpha) in vitiligo—New insight into pathogenesis of disease”. J Cosmet Dermatol. (2019) 18(1): 337-341.
Vainer et al., “Colonic expression and synthesis of interleukin 13 and interleukin 15 in inflammatory bowel disease”, Cytokine (2000) 12(10):1531-1536.
Yano et al., “Interleukin 15 induces the signals of epidermal proliferation through ERK and PI 3-kinase in a human epidermal keratinocyte cell line, HaCaT.” Biochem Biophys Res Comm. (2003) 301(4): 841-847.
Office Action dated Feb. 26, 2021 for Chinese Patent Application No. 201810863447.X.
Office Action dated Mar. 18, 2021 in Australian Application No. 2020201174.
Notice of Allowance dated Mar. 23, 2021 in Japanese Patent Application No. 2018-517887.
Notice of Acceptance dated Jun. 11, 2021 for Australian Patent Application No. 2019202527.
Office Action dated Aug. 17, 2021 in Japanese Application No. 2019-152602.
Office Action dated May 12, 2021 in Australian Patent Application No. 2018250210.
Office Action dated Jul. 22, 2021 in U.S. Appl. No. 16/294,733.
Ciszewski et al., Identification of a γc receptor antagonist that prevents reprogramming of human tissue-resident cytotoxic T cells by IL15 and IL21. Gastroenterol. (Feb. 1, 2020) 158(3): 625-637.
Office Action dated Jan. 29, 2021 in Canadian Application No. 2824515.
Office Action dated Sep. 24, 2021 in Chinese Application No. 201810863447.X.
Office Action dated Feb. 19, 2021 in European Application No. 19206731.2.
Office Action dated Feb. 23, 2021 in In Canadian Application No. 3000207.
Office Action dated Dec. 24, 2020 in Chinese Application No. 201680058779.X.
Office Action dated Sep. 17, 2021 in Chinese Application No. 201680058779.X.
Examination Report dated Dec. 9, 2020 in India Application No. 201817014941.
Office Action dated Feb. 8, 2022 in Japanese Patent Application No. 2019-554995.
International Search Report and Written Opinion dated Dec. 21, 2021 for PCT/US2021/038512.
Office Action dated Dec. 24, 2021 for U.S. Appl. No. 17/012,724.
Office Action dated Dec. 29, 2021 in U.S. Appl. No. 16/294,733.
Related Publications (1)
Number Date Country
20190070263 A1 Mar 2019 US
Provisional Applications (1)
Number Date Country
62239761 Oct 2015 US