WO2010085495A1 - Compositions and methods of treating inflammatory and autoimmune diseases - Google Patents

Compositions and methods of treating inflammatory and autoimmune diseases Download PDF

Info

Publication number
WO2010085495A1
WO2010085495A1 PCT/US2010/021519 US2010021519W WO2010085495A1 WO 2010085495 A1 WO2010085495 A1 WO 2010085495A1 US 2010021519 W US2010021519 W US 2010021519W WO 2010085495 A1 WO2010085495 A1 WO 2010085495A1
Authority
WO
WIPO (PCT)
Prior art keywords
variant
foxp3
cells
seq
stat5
Prior art date
Application number
PCT/US2010/021519
Other languages
French (fr)
Inventor
Marc A. Gavin
Li Li
Original Assignee
Amgen Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42356180&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2010085495(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US13/145,537 priority Critical patent/US20110274650A1/en
Priority to JP2011548074A priority patent/JP5766124B2/en
Priority to MX2011007647A priority patent/MX2011007647A/en
Priority to ES10733814T priority patent/ES2825173T3/en
Priority to CA2749539A priority patent/CA2749539C/en
Application filed by Amgen Inc. filed Critical Amgen Inc.
Priority to AU2010206840A priority patent/AU2010206840B2/en
Priority to EP10733814.7A priority patent/EP2382228B1/en
Publication of WO2010085495A1 publication Critical patent/WO2010085495A1/en
Priority to US13/672,417 priority patent/US9732134B2/en
Priority to US15/649,520 priority patent/US11560415B2/en
Priority to US18/068,970 priority patent/US20230139534A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • IL-2 binds three transmembrane receptor subunits: IL-2R ⁇ and IL-2R ⁇ which together activate intracellular signaling events upon IL-2 binding, and CD25 (IL-2R ⁇ ) which serves to present IL-2 to the other 2 receptor subunits.
  • the signals delivered by IL-2R ⁇ include those of the PI3-kinase, Ras-MAP-kinase, and STAT5 pathways.
  • T cells require expression of CD25 to respond to the low concentrations of IL-
  • T cells that express CD25 include both CD4 + FOXP3 + regulatory T cells (T-reg cells) — which are essential for suppressing autoimmune inflammation — and FOXP3 T cells that have been activated to express CD25.
  • FOXP3 CD25 + T effector cells (T-eff) may be either CD4 + or CD8 + cells, both of which can be pro-inflammatory and may contribute to autoimmunity, organ graft rejection or graft- versus-host disease.
  • IL-2-stimulated STAT5 signaling is crucial for normal T-reg cell growth and survival and for high FOXP3 expression.
  • IL-2R ⁇ and/or IL-2R ⁇ could be offset by an increased affinity for CD25.
  • Mutational variants of IL-2 have been generated that exhibit up to 170-fold higher affinity for CD25 (US Patent Application Publication No. 2005/0142106; Rao et al, Biochemistry 44, 10696-701 (2005)). These variants were reported to associate for several days with cell surface CD25 and to chronically promote growth of an IL-2-dependent cell line. The authors report that the mutants stimulate persistent T cell growth and, thus, may be useful in methods of viral immunotherapy and in treating cancer or other hyperproliferative disorders.
  • IL-2 Proleukin
  • US Pat. No. 6,955,807 describes IL-2 variants that are said to have reduced toxicity.
  • the patent attributes the toxicity to IL-2-induced stimulation of natural killer (NK) cells, which only express IL-2R ⁇ and IL-2R ⁇ .
  • NK natural killer
  • the IL-2 variants described therein were said to have reduced toxicity because they selectively activate CD25 + T cells over NK cells.
  • the IL-2 variants were said to be useful in therapeutic methods wherein it is beneficial to generally stimulate the immune system, e.g., the treatment of cancer or infectious diseases.
  • immunosuppressive mutational variants of IL-2 that preferentially promote the growth/survival of FOXP3 + regulatory T cells (T-reg cells) over the growth/survival of potentially proinflammatory FOXP3 CD25 + T cells.
  • T-reg cells regulatory T cells
  • FOXP3 CD25 + T cells By increasing the ratio of T-reg to other T cells and/or by increasing FOXP3 expression in T-reg without activating FOXP3 CD25 + T cells, these variants should suppress undesirable inflammation.
  • the unique properties of these IL-2 variants stem from two sets of mutations.
  • One set of mutations results in a reduced affinity for the signaling chains of the IL-2 receptor (IL-2R ⁇ /CD122 and/or IL-2R ⁇ /CD132) and/or a reduced capacity to induce a signaling event from one or both subunits of the IL-2 receptor.
  • the second set of mutations confers higher affinity for CD25 (IL-2R ⁇ ) and may include mutations described by Rao et al. (US Patent Application Publication No. 2005/0142106).
  • certain IL-2 variants induce signaling events that preferentially induce survival, proliferation, activation and/or function of T-reg cells.
  • the IL-2 variant retains the capacity to stimulate, in T-reg cells, STAT5 phosphorylation and/or phosphorylation of one or more of signaling molecules downstream of the IL-2R, e.g., p38, ERK, SYK and LCK.
  • the IL-2 variant retains the capacity to stimulate, in T-reg cells, transcription or protein expression of genes or proteins, such as FOXP3 or IL-10, that are important for T-reg cell survival, proliferation, activation and/or function.
  • the IL-2 variant exhibits a reduced capacity to stimulate endocytosis of IL-2/IL-2R complexes on the surface of CD25 + T cells.
  • the IL-2 variant demonstrates inefficient, reduced, or absence of stimulation of PI3 -kinase signaling , such as inefficient, reduced or absent phosphorylation of AKT and/or mTOR (mammalian target of rapamycin).
  • the IL-2 variant retains the ability of wt IL-2 to stimulate STAT5 phosphorylation and/or phosphorylation of one or more of signaling molecules downstream of the IL-2R in T-reg cells, yet demonstrates inefficient, reduced, or absent phosphorylation of STAT5, AKT and/or mTOR or other signaling molecules downstream of the IL-2R in FOXP3 CD4 + or CD8 + T cells or NK cells.
  • the IL-2 variant is inefficient or incapable of stimulating survival, growth, activation and/or function of FOXP3 CD4 + or CD8 + T cells or NK cells.
  • the methods comprise administering a therapeutically effective amount of one or more immunosuppressive IL-2 variants to a subject.
  • the methods comprising contacting a FOXP3- positive (FOXP3 ) T cell with an immunosuppressive IL-2 variant.
  • an immunosuppressive IL-2 variant in the preparation of a medicament for the treatment of an inflammatory or autoimmune disorder.
  • compositions of IL-2 variants conjugated to additional protein sequences or other chemical that prolong the stability and/or half- life of the therapeutic in vivo.
  • FIG. 1. Sequences of IL-2 variants. Sequences that vary from germline human IL-2 are not shaded in gray.
  • FIG. 2A-2F An example of flow cytometric data and gating strategy. Representative data is shown in FIG. IA where 9.5% of CD4 + cells are
  • CD25 + FOXP3 + , 9.9% of CD4 + cells are CD25 + FOXP3 and 6.7% of CD8 + cells are CD25 + FOXP3 .
  • FOXP3 + CD8 + T cells are typically very infrequent.
  • FIG. 2B Relative number of CD8 + CD25 + FOXP3 T cells.
  • FIG. 2C Relative number of CD4 + CD25 + FOXP3 T cells.
  • FIG. 2D Relative number of CD4 + CD25 + FOXP3 + T cells.
  • FIG. 2E Ratio of FOXP3 + /FOXP3 cells among CD25 + CD4 + T cells.
  • FIG. 2F IL-2-mediated FOXP3 upregulation in FOXP3 + CD4 + T cells.
  • FIG 3. IL-2 muteins stimulate phospho-STAT5 in T-reg but are inefficient at stimulating signals in other T cells.
  • T cells were stimulated with IL-2 as described in Example 3.
  • Phospho-STAT5 was measured by flow cytometry and phospho-AKT was measured by ELISA (MesoScale Discovery). Abbreviations are as follows: T-reg, FOXP3 + CD4 + T cells; CD4 T-eff, CD4 + CD25 + FOXP3 "effector” T cells; CD8 T-eff, CD8 + CD25 + FOXP3 "effector” T cells.
  • T-reg cells are essential for maintaining normal immune homeostasis and immune tolerance to self tissues, as well as for suppressing undesirable inflammation. T-reg cells exert their suppressor and regulatory functions through multiple mechanisms which are likely to be regulated by temporal and environmental factors.
  • Current immunosuppressive therapeutics generally target individual proinflammatory pathways and as such often exhibit partial efficacy or are applicable to specific diseases.
  • An alternative immunosuppressive modality might involve the elevation of the numbers and activation state of natural suppressor cells to better enable them to deliver appropriate suppressor molecules/activities at sites of inflammation.
  • selectively promote it is meant the therapeutic agent promotes the activity in T-reg cells but has limited or lacks the ability to promote the activity in non-regulatory T cells.
  • assays to screen for agents that selectively promote T-reg cell proliferation, survival, activation and/or function include, but are not limited to, small molecules, peptides, polypeptides, proteins including antibodies, e.g., monoclonal, humanized, human, monovalent, bivalent, and multivalent antibodies.
  • the agent is an IL-2 variant.
  • the IL-2 variant promotes these activities of T-reg cell growth/survival but have a reduced ability, as compared to wild-type IL-2, to promote non-regulatory T-cell (FOXP3 CD25 ) and NK cell proliferation, survival, activation and/or function.
  • such IL-2 variants function through a combination of elevated affinity for the IL-2R subunit IL-2R ⁇ (CD25) and a reduced affinity for the signaling subunits IL-2R ⁇ and/or IL-2R ⁇ .
  • IL-2 and variants thereof have been used in the art as immunostimulatory agents, e.g., in methods of treating cancer or infectious diseases
  • the IL-2 variants described herein are particularly useful as immunosuppressive agents, e.g., in methods of treating inflammatory disorders.
  • IL-2 variants comprise a sequence of amino acids at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93% at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to wild-type IL-2.
  • IL-2 variants further include a sequence of amino acids at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93% at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to a functional fragment of wild-type IL-2.
  • wild-type IL-2 shall mean the polypeptide having the following amino acid sequence: APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATEL KHLQCLEEELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTFMCEY ADETATIVEFLNRWITFXQSIISTLT wherein X is C, S, A or V (SEQ ID NO: 1).
  • Variants may contain one or more substitutions, deletions, or insertions within the wild-type IL-2 amino acid sequence. Residues are designated herein by the one letter amino acid code followed by the IL-2 amino acid position, e.g., K35 is the lysine residue at position 35 of SEQ ID NO: 1. Substitutions are designated herein by the one letter amino acid code followed by the IL-2 amino acid position followed by the substituting one letter amino acid code., e.g., K35A is a substitution of the lysine residue at position 35 of SEQ ID NO:1 with an alanine residue.
  • the invention provides immunosuppressive IL-2 variants that have a higher affinity for IL-2R ⁇ than wild-type IL-2.
  • IL-2 variants that have higher affinity for IL-2R ⁇ than does wild-type IL-2 and methods of making and screening for such variants.
  • Preferred IL-2 variants contain one or more mutations in positions of the IL-2 sequence that either contact IL- 2R ⁇ or alter the orientation of other positions contacting IL-2R ⁇ , resulting in higher affinity for IL-2R ⁇ .
  • the mutations may be in or near areas known to be in close proximity to IL-2R ⁇ based on published crystal structures (Xinquan Wang, Mathias Rickert, K. Christopher Garcia. Science 310:1159 2005).
  • IL-2 residues believed to contact IL-2R ⁇ include K35, R38, F42, K43, F44, Y45, E61, E62, K64, P65, E68, V69, L72, and Y107.
  • IL-2 variants having greater affinity for IL-2R ⁇ can include a change in N29, N30, Y31, K35, T37, K48, E68, V69, N71, Q74, S75, or K76.
  • Preferred variants include those having one or more of the following mutations: N29S, N30S, N30D, Y31H, Y31S, K35R, T37A, K48E, V69A, N71R, and Q74P.
  • Immunosuppressive IL-2 variants also include variants that demonstrate altered signaling through certain pathways activated by wild-type IL-2 via the IL-2R and result in preferential proliferation/survival/activation of T-reg.
  • Molecules known to be phosphorylated upon activation of the IL-2R include STAT5, p38, ERK, SYK, LCK, AKT and mTOR.
  • the immunosuppressive IL-2 variant can possess a reduced PI3K signaling ability in FOXP3 T cells, which can be measured by a reduction in the phosphorylation of AKT and/or mTOR as compared to wild-type IL-2.
  • Such variants may include mutations in positions that either contact IL-2R ⁇ or IL-2R ⁇ or alter the orientation of other positions contacting IL-2R ⁇ or IL- 2R ⁇ .
  • IL-2 residues believed to contact IL-2R ⁇ include L 12, Q 13, H 16, L 19, D20, M23, R81, D84, S87, N88, V91, 192, and E95.
  • IL-2 residues believed to contact IL- 2R ⁇ include QI l, L18, Q22, El 10, Nl 19, T123, Q126, S127, 1129, S130, and T133.
  • the IL-2 variant comprises a mutation at E 15, H 16, Q22, D84, N88, or E95. Examples of such mutations include E15Q, H16N, Q22E, D84N, N88D, and E95Q.
  • the IL-2 variant comprises a combination of mutations.
  • Examples of IL-2 variants having a combination of mutations are provided in FIG. 1 and include haWT (SEQ ID NO:2), haD, (SEQ ID NO:3), haD.1 (SEQ ID NO:4), haD.2 (SEQ ID NO:5), haD.4 (SEQ ID NO:6), haD.5 (SEQ ID NO:7), haD.6 (SEQ ID NO:8), haD.8 (SEQ ID NO:9), and haD.11 (SEQ ID NO:10)d.
  • the IL-2 variant stimulates STAT5 phosphorylation in FOXP3 -positive regulatory T cells but has reduced ability to induce STAT5 and AKT phosphorylation in FOXP3 -negative T cells as compared to wild-type IL-2.
  • Preferred variants having such properties include haD, haD.l, haD.2, haD.4, haD.5, haD.6, and haD.8.
  • the IL-2 variants may further comprise one or more mutations as compared to the wild-type IL-2 sequence that do not have an effect on the affinity for IL-2R ⁇ or IL-2R ⁇ , provided the IL-2 variant promotes the preferential proliferation, survival, activation or function of FOXP3 + T-reg over that of other T cells that do not express FOXP3.
  • mutations are conservative mutations.
  • the IL-2 variant may comprise one or more compounds to increase the serum- half-life of the IL-2 variant when administered to a patient.
  • Such half-life extending molecules include water soluble polymers (e.g., polyethylene glycol (PEG)), low- and high- density lipoproteins, antibody Fc (monomer or dimer), transthyretin (TTR), and TGF- ⁇ latency associated peptide (LAP).
  • PEG polyethylene glycol
  • TTR transthyretin
  • LAP TGF- ⁇ latency associated peptide
  • IL-2 variants comprising a combination of serum half- life extending molecules, such as PEGylated TTR (US Pat. Appl. Publ. No. 2003/0195154).
  • the immunosuppressive IL-2 variants can be produced using any suitable method known in the art, including those described in U.S. Pat. No. 6,955,807 for producing immunostimulatory IL-2 variants (incorporated herein by reference). Such methods include constructing a DNA sequence encoding the IL-2 variant and expressing those sequences in a suitably transformed host. This method will produce the recombinant variant of this invention. However, the variants may also be produced by chemical synthesis or a combination of chemical synthesis and recombinant DNA technology. Batch-wise production or perfusion production methods are known in the art. See Freshey, R. I.
  • a DNA sequence is constructed by isolating or synthesizing a DNA sequence encoding the wild type IL-2 and then changing one or more codons by site-specific mutagenesis. This technique is well known. See, e.g., Mark et. al., "Site-specific Mutagenesis Of The Human Fibroblast Interferon Gene", Proc. Natl. Acad. Sci. USA 81, pp. 5662-66 (1984); and U.S. Pat. No. 4,588,585, incorporated herein by reference.
  • Another method of constructing a DNA sequence encoding the IL-2 variant would be chemical synthesis. This for example includes direct synthesis of a peptide by chemical means of the protein sequence encoding for an IL-2 variant exhibiting the properties described herein. This method may incorporate both natural and unnatural amino acids at positions that affect the interactions of IL-2 with the IL2R ⁇ , IL-2R ⁇ , or IL-2R ⁇ . Alternatively, a gene which encodes the desired IL-2 variant may be synthesized by chemical means using an oligonucleotide synthesizer.
  • Such oligonucleotides are designed based on the amino acid sequence of the desired IL-2 variant, and preferably selecting those codons that are favored in the host cell in which the recombinant variant will be produced.
  • the genetic code is degenerate—that an amino acid may be coded for by more than one codon.
  • Phe (F) is coded for by two codons, TTC or TTT
  • Tyr (Y) is coded for by TAC or TAT
  • his (H) is coded for by CAC or CAT.
  • Trp (W) is coded for by a single codon, TGG. Accordingly, it will be appreciated that for a given DNA sequence encoding a particular IL-2 variant, there will be many DNA degenerate sequences that will code for that IL-2 variant.
  • the DNA sequence encoding the IL-2 variant may or may not also include DNA sequences that encode a signal sequence.
  • Such signal sequence should be one recognized by the cell chosen for expression of the IL-2 variant. It may be prokaryotic, eukaryotic or a combination of the two. It may also be the signal sequence of native IL-2. The inclusion of a signal sequence depends on whether it is desired to secrete the IL-2 variant from the recombinant cells in which it is made. If the chosen cells are prokaryotic, it generally is preferred that the DNA sequence not encode a signal sequence. If the chosen cells are eukaryotic, it generally is preferred that a signal sequence be encoded and most preferably that the wild-type IL-2 signal sequence be used.
  • Standard methods may be applied to synthesize a gene encoding an IL-2 variant.
  • the complete amino acid sequence may be used to construct a back-translated gene.
  • a DNA oligomer containing a nucleotide sequence coding for an IL-2 variant may be synthesized.
  • several small oligonucleotides coding for portions of the desired polypeptide may be synthesized and then ligated.
  • the individual oligonucleotides typically contain 5 ' or 3' overhangs for complementary assembly.
  • the DNA sequences encoding an IL-2 variant will be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the IL-2 variant in the desired transformed host. Proper assembly may be confirmed by nucleotide sequencing, restriction mapping, and expression of a biologically active polypeptide in a suitable host. As is well known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to transcriptional and translational expression control sequences that are functional in the chosen expression host. The choice of expression control sequence and expression vector will depend upon the choice of host. A wide variety of expression host/vector combinations may be employed.
  • Any suitable host may be used to produce the IL-2 variant, including bacteria, fungi (including yeasts), plant, insect, mammal, or other appropriate animal cells or cell lines, as well as transgenic animals or plants. More particularly, these hosts may include well known eukaryotic and prokaryotic hosts, such as strains of E.coli, Pseudomonas, Bacillus, Streptomyces, fungi, yeast, insect cells such as Spodoptera frugiperda (Sf9), animal cells such as Chinese hamster ovary (CHO) and mouse cells such as NS/O, African green monkey cells such as COS 1, COS 7, BSC 1, BSC 40, and BNT 10, and human cells, as well as plant cells in tissue culture. For animal cell expression, CHO cells and COS 7 cells in cultures and particularly the CHO cell line CHO (DHFR-) or the HKB line are preferred.
  • DHFR- CHO cell line CHO
  • vectors and expression control sequences will function equally well to express the DNA sequences described herein. Neither will all hosts function equally well with the same expression system. However, one of skill in the art may make a selection among these vectors, expression control sequences and hosts without undue experimentation. For example, in selecting a vector, the host must be considered because the vector must replicate in it. The vectors copy number, the ability to control that copy number, and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered.
  • preferred vectors for use in this invention include those that allow the DNA encoding the IL-2 variants to be amplified in copy number. Such amplifiable vectors are well known in the art.
  • IL-2 variants may be glycosylated or unglycosylated depending on the host organism used to produce the variant.
  • the IL-2 variant produced by the transformed host can be purified according to any suitable method. Various methods are known for purifying IL-2. See, e.g Current Protocols in Protein Science, VoI 2. Eds: John E. Coligan, Ben M. Dunn, Hidde L. Ploehg, David W. Speicher, Paul T. Wingf ⁇ eld, Unit 6.5 (Copyright 1997, John Wiley and Sons, Inc).
  • the biological activity of the IL-2 variants can be assayed by any suitable method known in the art. Such assays include those described in the Examples below.
  • Diseases, disorders, or conditions may be amenable to treatment with or may be prevented by administration of a T-reg-selective IL-2 variant to a subject.
  • diseases, disorders, and conditions include, but are not limited to, inflammation, autoimmune disease, paraneoplastic autroimmune diseases, cartilage inflammation, fibrotic disease and/or bone degradation, arthritis, rheumatoid arthritis, juvenile arthritis, juvenile rheumatoid arthritis, pauciarticular juvenile rheumatoid arthritis, polyarticular juvenile rheumatoid arthritis, systemic onset juvenile rheumatoid arthritis, juvenile ankylosing spondylitis, juvenile enteropathic arthritis, juvenile reactive arthritis, juvenile Reter's Syndrome, SEA Syndrome (Seronegativity, Enthesopathy, Arthropathy Syndrome), juvenile dermatomyositis, juvenile psoriatic arthritis, juvenile scleroderma, juvenile systemic lupus erythematosus, juvenile vascu
  • treatment encompasses alleviation or prevention of at least one symptom or other aspect of a disorder, or reduction of disease severity, and the like.
  • a T-reg-selective IL-2 variant need not effect a complete cure, or eradicate every symptom or manifestation of a disease, to constitute a viable therapeutic agent.
  • drugs employed as therapeutic agents may reduce the severity of a given disease state, but need not abolish every manifestation of the disease to be regarded as useful therapeutic agents.
  • a prophylactically administered treatment need not be completely effective in preventing the onset of a condition in order to constitute a viable prophylactic agent.
  • One embodiment of the invention is directed to a method comprising administering to a patient A T-reg-selective IL-2 variant in an amount and for a time sufficient to induce a sustained improvement over baseline of an indicator that reflects the severity of the particular disorder.
  • compositions comprising a therapeutically effective amount of one or a plurality of T-reg-selective IL-2 variants of the invention together with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative, and/or adjuvant.
  • the invention provides methods of treating a patient by administering such pharmaceutical composition.
  • patient includes human and animal subjects.
  • acceptable formulation materials preferably are nontoxic to recipients at the dosages and concentrations employed.
  • the pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolality, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta- cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides; and other carbohydrates (such as glucose, sucrose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emul
  • T-reg-selective IL-2 variant- containing pharmaceutical composition to be employed will depend, for example, upon the therapeutic context and objectives.
  • dosage levels for treatment will vary depending, in part, upon the molecule delivered, the indication for which the T-reg-selective IL-2 variant is being used, the route of administration, and the size (body weight, body surface or organ size) and/or condition (the age and general health) of the patient.
  • the clinician may titer the dosage and modify the route of administration to obtain the optimal therapeutic effect.
  • a typical dosage may range from about 0.1 ⁇ g/kg to up to about 30 mg/kg or more, depending on the factors mentioned above. In specific embodiments, the dosage may range from 0.1 ⁇ g/kg up to about 30 mg/kg, optionally from 1 ⁇ g/kg up to about 30 mg/kg or from 10 ⁇ g/kg up to about 5 mg/kg.
  • Dosing frequency will depend upon the pharmacokinetic parameters of the particular T-reg-selective IL-2 variant in the formulation used.
  • a clinician administers the composition until a dosage is reached that achieves the desired effect.
  • the composition may therefore be administered as a single dose, or as two or more doses (which may or may not contain the same amount of the desired molecule) over time, or as a continuous infusion via an implantation device or catheter. Further refinement of the appropriate dosage is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them.
  • the route of administration of the pharmaceutical composition is in accord with known methods, e.g., orally, through injection by intravenous, intraperitoneal, intracerebral (intra- parenchymal), intracerebroventricular, intramuscular, intra-ocular, intraarterial, intraportal, or intralesional routes; by sustained release systems or by implantation devices.
  • the compositions may be administered by bolus injection or continuously by infusion, or by implantation device.
  • T-reg-selective IL-2 variant is administered in combination with other agents useful for treating the condition with which the patient is afflicted.
  • agents include both proteinaceous and non- proteinaceous drugs.
  • dosages may be adjusted accordingly, as is recognized in the pertinent art.
  • “Co-administration” and combination therapy are not limited to simultaneous administration, but also include treatment regimens in which a T-reg-selective IL-2 variant is administered at least once during a course of treatment that involves administering at least one other therapeutic agent to the patient.
  • a T-reg-selective IL-2 variant is administered in combination with an inhibitor of the PI3-K/AKT/mTOR pathway, e.g., rapamycin (rapamune, sirolimus). Inhibitors of this pathway in combination with IL-2 favor T- reg enrichment.
  • an inhibitor of the PI3-K/AKT/mTOR pathway e.g., rapamycin (rapamune, sirolimus).
  • Inhibitors of this pathway in combination with IL-2 favor T- reg enrichment.
  • Example 1 Panel of IL-2 mutants.
  • variant haD, haD.l, haD.2, etc, also contained mutations predicted to alter interactions with IL-2R ⁇ and/or IL-2R ⁇ .
  • variant haD.11 was not capable of inducing any signal or altering any cellular phenotype and, as such, served as a control for CD25 binding without IL-2R signaling.
  • All the IL-2 variants contained the C 125 S mutation for improved manufacturability and terminated with FLAG and HIS-tag sequences (DYKDDDDKG S SHHHHHH) (SEQ ID NO:11)
  • Cell signaling e.g. detection of phosphorylated STAT5 and AKT using flow cytometric and ELISA-based methods.
  • Example 2 Enrichment of FOXP3 + cells and retention of FOXP3 upregulation during long term T cell culture.
  • PBMC peripheral blood mononuclear cells
  • CD8 + CD25 + T cells were especially responsive to WT IL-2 and variant ha WT, however, all variants that contained mutated IL-2R ⁇ and/or ⁇ contact residues were very inefficient at promoting accumulation of activated CD8 CD25 + T cells (FIG. 2B).
  • a similar trend was observed for CD4 + CD25 + FOXP3 T cells (FIG. 2C).
  • the growth/survival of FOXP3 + CD4 + T cells was stimulated by several IL-2 variants to a degree similar to that of WT IL-2 (FIG. 2D).
  • the ratio of FOXP3 + to FOXP3 T cells among CD4 + CD25 + T cells was increased by several IL-2 variants with IL-2R ⁇ -contact residue mutations (FIG. 2E). Furthermore, the mutations did not impair IL-2-stimulated FOXP3 upregulation in T-reg (FIG. 2F).
  • Example 3 Mutations that reduce signaling in FOXP3 T cells but stimulate STAT5 signaling in T-reg.
  • the IL-2 variants were screened for their ability to stimulate AKT and STAT5 phosphorylation in T cell subsets.
  • Several IL-2 variants were as potent, or nearly as potent, as wt IL-2 at stimulating STAT5 in FOXP3 + T cells 10 min after stimulation.
  • Three hours after washing IL-2 from the media some IL-2 variants (haD, haD.l, haD.2, haD.4, haD.6, and haD.8) continued to stimulate sustained STAT5 signaling at levels higher than that seen with wt IL-2.
  • STAT5 and AKT responses to the haD variants after 10 min stimulation were not nearly as high as those stimulated by wt IL-2 or ha WT.
  • T cells Previously activated (with anti-CD3 for 2-3 days) and rested (in fresh culture medium for 2-5 days) T cells were exposed to 1 nM wt or mutant IL-2 for 10 min at 37°C. Cells were then stained (10 min timepoint) or washed and cultured for an additional 3 hrs (3 hr timepoint).
  • a 50 ⁇ l culture was stopped by adding an equal volume of 2x lysis buffer and lysates were measured for phospho-AKT with multiplex ELISA plates according to the manufacturer's protocols (MesoScale Discovery, Gaithersburg, Maryland).
  • a 50 ⁇ l culture was stopped by adding 1 ml of FOXP3 Fix/Perm Buffer (BioLegend, San Diego, CA), incubation at 25°C for 20 min, and staining for cell surface markers, FOXP3 and phospho-STAT5 according the BioLegend FOXP3 staining protocol.
  • FOXP3 Fix/Perm Buffer BioLegend, San Diego, CA

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Pulmonology (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Described herein are immunosuppressive molecules including immunosuppressive variants of IL-2, and use of such molecules to treat inflammatory and autoimmune disorders.

Description

COMPOSITIONS AND METHODS OF TREATING INFLAMMATORY AND AUTOIMMUNE DISEASES
This application claims priority to U.S. Provisional Patent Application No. 61/146,111, filed January 21, 2009, which is incorporated herein by reference in its entirety.
Background
IL-2 binds three transmembrane receptor subunits: IL-2Rβ and IL-2Rγ which together activate intracellular signaling events upon IL-2 binding, and CD25 (IL-2Rα) which serves to present IL-2 to the other 2 receptor subunits. The signals delivered by IL-2Rβγ include those of the PI3-kinase, Ras-MAP-kinase, and STAT5 pathways. T cells require expression of CD25 to respond to the low concentrations of IL-
2 that typically exist in tissues. T cells that express CD25 include both CD4+ FOXP3+ regulatory T cells (T-reg cells) — which are essential for suppressing autoimmune inflammation — and FOXP3 T cells that have been activated to express CD25. FOXP3 CD25+ T effector cells (T-eff) may be either CD4+ or CD8+ cells, both of which can be pro-inflammatory and may contribute to autoimmunity, organ graft rejection or graft- versus-host disease. IL-2-stimulated STAT5 signaling is crucial for normal T-reg cell growth and survival and for high FOXP3 expression.
Because of the low affinity IL-2 possesses for each of the three IL-2R chains, a further reduction in affinity for IL-2Rβ and/or IL-2Rγ could be offset by an increased affinity for CD25. Mutational variants of IL-2 have been generated that exhibit up to 170-fold higher affinity for CD25 (US Patent Application Publication No. 2005/0142106; Rao et al, Biochemistry 44, 10696-701 (2005)). These variants were reported to associate for several days with cell surface CD25 and to chronically promote growth of an IL-2-dependent cell line. The authors report that the mutants stimulate persistent T cell growth and, thus, may be useful in methods of viral immunotherapy and in treating cancer or other hyperproliferative disorders. High doses of IL-2 (Proleukin) are administered to cancer patients to induce anti-tumor immunity, a treatment that is often associated with undesirable toxicity. US Pat. No. 6,955,807 describes IL-2 variants that are said to have reduced toxicity. The patent attributes the toxicity to IL-2-induced stimulation of natural killer (NK) cells, which only express IL-2Rβ and IL-2Rγ. The IL-2 variants described therein were said to have reduced toxicity because they selectively activate CD25+ T cells over NK cells. Again the IL-2 variants were said to be useful in therapeutic methods wherein it is beneficial to generally stimulate the immune system, e.g., the treatment of cancer or infectious diseases.
Summary Provided herein are immunosuppressive mutational variants of IL-2 that preferentially promote the growth/survival of FOXP3+ regulatory T cells (T-reg cells) over the growth/survival of potentially proinflammatory FOXP3 CD25+ T cells. By increasing the ratio of T-reg to other T cells and/or by increasing FOXP3 expression in T-reg without activating FOXP3 CD25+ T cells, these variants should suppress undesirable inflammation.
The unique properties of these IL-2 variants stem from two sets of mutations. One set of mutations results in a reduced affinity for the signaling chains of the IL-2 receptor (IL-2Rβ/CD122 and/or IL-2Rγ/CD132) and/or a reduced capacity to induce a signaling event from one or both subunits of the IL-2 receptor. The second set of mutations confers higher affinity for CD25 (IL-2Rα) and may include mutations described by Rao et al. (US Patent Application Publication No. 2005/0142106).
As described herein, certain IL-2 variants induce signaling events that preferentially induce survival, proliferation, activation and/or function of T-reg cells. In certain embodiments, the IL-2 variant retains the capacity to stimulate, in T-reg cells, STAT5 phosphorylation and/or phosphorylation of one or more of signaling molecules downstream of the IL-2R, e.g., p38, ERK, SYK and LCK. In other embodiments, the IL-2 variant retains the capacity to stimulate, in T-reg cells, transcription or protein expression of genes or proteins, such as FOXP3 or IL-10, that are important for T-reg cell survival, proliferation, activation and/or function. In other embodiments, the IL-2 variant exhibits a reduced capacity to stimulate endocytosis of IL-2/IL-2R complexes on the surface of CD25+ T cells. In other embodiments, the IL-2 variant demonstrates inefficient, reduced, or absence of stimulation of PI3 -kinase signaling , such as inefficient, reduced or absent phosphorylation of AKT and/or mTOR (mammalian target of rapamycin). In yet other embodiments, the IL-2 variant retains the ability of wt IL-2 to stimulate STAT5 phosphorylation and/or phosphorylation of one or more of signaling molecules downstream of the IL-2R in T-reg cells, yet demonstrates inefficient, reduced, or absent phosphorylation of STAT5, AKT and/or mTOR or other signaling molecules downstream of the IL-2R in FOXP3 CD4+ or CD8+ T cells or NK cells. In other embodiments, the IL-2 variant is inefficient or incapable of stimulating survival, growth, activation and/or function of FOXP3 CD4+ or CD8+ T cells or NK cells.
Provided are methods of treating an inflammatory or autoimmune disorder. The methods comprise administering a therapeutically effective amount of one or more immunosuppressive IL-2 variants to a subject.
Further provided are methods of promoting proliferation, survival, growth, or activation of regulatory T cells. The methods comprising contacting a FOXP3- positive (FOXP3 ) T cell with an immunosuppressive IL-2 variant.
Also provided is the use of an immunosuppressive IL-2 variant in the preparation of a medicament for the treatment of an inflammatory or autoimmune disorder.
Also provided are compositions of IL-2 variants conjugated to additional protein sequences or other chemical that prolong the stability and/or half- life of the therapeutic in vivo.
Brief Explanation of the Drawings
FIG. 1. . Sequences of IL-2 variants. Sequences that vary from germline human IL-2 are not shaded in gray.
FIG. 2A-2F. FIG. 2A An example of flow cytometric data and gating strategy. Representative data is shown in FIG. IA where 9.5% of CD4+ cells are
CD25+FOXP3+, 9.9% of CD4+ cells are CD25+FOXP3 and 6.7% of CD8+ cells are CD25+FOXP3 . FOXP3+CD8+ T cells are typically very infrequent. FIG. 2B. Relative number of CD8+CD25+FOXP3 T cells. FIG. 2C. Relative number of CD4+CD25+FOXP3 T cells. FIG. 2D. Relative number of CD4+CD25+FOXP3+ T cells. FIG. 2E. Ratio of FOXP3+/FOXP3 cells among CD25+CD4+ T cells. FIG. 2F. IL-2-mediated FOXP3 upregulation in FOXP3+CD4+ T cells.
FIG 3. IL-2 muteins stimulate phospho-STAT5 in T-reg but are inefficient at stimulating signals in other T cells. T cells were stimulated with IL-2 as described in Example 3. Phospho-STAT5 was measured by flow cytometry and phospho-AKT was measured by ELISA (MesoScale Discovery). Abbreviations are as follows: T-reg, FOXP3+CD4+ T cells; CD4 T-eff, CD4+CD25+FOXP3 "effector" T cells; CD8 T-eff, CD8+CD25+FOXP3 "effector" T cells.
Detailed Description of Preferred Embodiments
FOXP3+ regulatory T cells (T-reg cells) are essential for maintaining normal immune homeostasis and immune tolerance to self tissues, as well as for suppressing undesirable inflammation. T-reg cells exert their suppressor and regulatory functions through multiple mechanisms which are likely to be regulated by temporal and environmental factors. Current immunosuppressive therapeutics generally target individual proinflammatory pathways and as such often exhibit partial efficacy or are applicable to specific diseases. An alternative immunosuppressive modality might involve the elevation of the numbers and activation state of natural suppressor cells to better enable them to deliver appropriate suppressor molecules/activities at sites of inflammation.
Described herein are therapeutic agents that selectively promote T-reg cell proliferation, survival, activation and/or function. By "selectively promote," it is meant the therapeutic agent promotes the activity in T-reg cells but has limited or lacks the ability to promote the activity in non-regulatory T cells. Further described herein are assays to screen for agents that selectively promote T-reg cell proliferation, survival, activation and/or function. Agents that may be screened include, but are not limited to, small molecules, peptides, polypeptides, proteins including antibodies, e.g., monoclonal, humanized, human, monovalent, bivalent, and multivalent antibodies. In certain embodiments, the agent is an IL-2 variant. In particular, the IL-2 variant promotes these activities of T-reg cell growth/survival but have a reduced ability, as compared to wild-type IL-2, to promote non-regulatory T-cell (FOXP3 CD25 ) and NK cell proliferation, survival, activation and/or function. In certain embodiments, such IL-2 variants function through a combination of elevated affinity for the IL-2R subunit IL-2Rα (CD25) and a reduced affinity for the signaling subunits IL-2Rβ and/or IL-2Rγ. Whereas IL-2 and variants thereof have been used in the art as immunostimulatory agents, e.g., in methods of treating cancer or infectious diseases, the IL-2 variants described herein are particularly useful as immunosuppressive agents, e.g., in methods of treating inflammatory disorders. IL-2 variants comprise a sequence of amino acids at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93% at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to wild-type IL-2. IL-2 variants further include a sequence of amino acids at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93% at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to a functional fragment of wild-type IL-2. As used herein, "wild-type IL-2" shall mean the polypeptide having the following amino acid sequence: APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATEL KHLQCLEEELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTFMCEY ADETATIVEFLNRWITFXQSIISTLT wherein X is C, S, A or V (SEQ ID NO: 1).
Variants may contain one or more substitutions, deletions, or insertions within the wild-type IL-2 amino acid sequence. Residues are designated herein by the one letter amino acid code followed by the IL-2 amino acid position, e.g., K35 is the lysine residue at position 35 of SEQ ID NO: 1. Substitutions are designated herein by the one letter amino acid code followed by the IL-2 amino acid position followed by the substituting one letter amino acid code., e.g., K35A is a substitution of the lysine residue at position 35 of SEQ ID NO:1 with an alanine residue. In one aspect, the invention provides immunosuppressive IL-2 variants that have a higher affinity for IL-2Rα than wild-type IL-2. U.S. Published Patent Application No. 2005/0142106 (incorporated herein by reference in its entirety) describes IL-2 variants that have higher affinity for IL-2Rα than does wild-type IL-2 and methods of making and screening for such variants. Preferred IL-2 variants contain one or more mutations in positions of the IL-2 sequence that either contact IL- 2Rα or alter the orientation of other positions contacting IL-2Rα, resulting in higher affinity for IL-2Rα. The mutations may be in or near areas known to be in close proximity to IL-2Rα based on published crystal structures (Xinquan Wang, Mathias Rickert, K. Christopher Garcia. Science 310:1159 2005). IL-2 residues believed to contact IL-2Rα include K35, R38, F42, K43, F44, Y45, E61, E62, K64, P65, E68, V69, L72, and Y107.
IL-2 variants having greater affinity for IL-2Rα can include a change in N29, N30, Y31, K35, T37, K48, E68, V69, N71, Q74, S75, or K76. Preferred variants include those having one or more of the following mutations: N29S, N30S, N30D, Y31H, Y31S, K35R, T37A, K48E, V69A, N71R, and Q74P.
Immunosuppressive IL-2 variants also include variants that demonstrate altered signaling through certain pathways activated by wild-type IL-2 via the IL-2R and result in preferential proliferation/survival/activation of T-reg. Molecules known to be phosphorylated upon activation of the IL-2R include STAT5, p38, ERK, SYK, LCK, AKT and mTOR. Compared to wild-type IL-2, the immunosuppressive IL-2 variant can possess a reduced PI3K signaling ability in FOXP3 T cells, which can be measured by a reduction in the phosphorylation of AKT and/or mTOR as compared to wild-type IL-2. Such variants may include mutations in positions that either contact IL-2Rβ or IL-2Rγ or alter the orientation of other positions contacting IL-2Rβ or IL- 2Rγ. IL-2 residues believed to contact IL-2Rβ include L 12, Q 13, H 16, L 19, D20, M23, R81, D84, S87, N88, V91, 192, and E95. IL-2 residues believed to contact IL- 2Rγ include QI l, L18, Q22, El 10, Nl 19, T123, Q126, S127, 1129, S130, and T133. In certain embodiments, the IL-2 variant comprises a mutation at E 15, H 16, Q22, D84, N88, or E95. Examples of such mutations include E15Q, H16N, Q22E, D84N, N88D, and E95Q.
In certain embodiments, the IL-2 variant comprises a combination of mutations. Examples of IL-2 variants having a combination of mutations are provided in FIG. 1 and include haWT (SEQ ID NO:2), haD, (SEQ ID NO:3), haD.1 (SEQ ID NO:4), haD.2 (SEQ ID NO:5), haD.4 (SEQ ID NO:6), haD.5 (SEQ ID NO:7), haD.6 (SEQ ID NO:8), haD.8 (SEQ ID NO:9), and haD.11 (SEQ ID NO:10)d. In preferred embodiments, the IL-2 variant stimulates STAT5 phosphorylation in FOXP3 -positive regulatory T cells but has reduced ability to induce STAT5 and AKT phosphorylation in FOXP3 -negative T cells as compared to wild-type IL-2. Preferred variants having such properties include haD, haD.l, haD.2, haD.4, haD.5, haD.6, and haD.8.
The IL-2 variants may further comprise one or more mutations as compared to the wild-type IL-2 sequence that do not have an effect on the affinity for IL-2Rβ or IL-2Rγ, provided the IL-2 variant promotes the preferential proliferation, survival, activation or function of FOXP3+ T-reg over that of other T cells that do not express FOXP3. In preferred embodiments, such mutations are conservative mutations. The IL-2 variant may comprise one or more compounds to increase the serum- half-life of the IL-2 variant when administered to a patient. Such half-life extending molecules include water soluble polymers (e.g., polyethylene glycol (PEG)), low- and high- density lipoproteins, antibody Fc (monomer or dimer), transthyretin (TTR), and TGF-β latency associated peptide (LAP). Also contemplated are IL-2 variants comprising a combination of serum half- life extending molecules, such as PEGylated TTR (US Pat. Appl. Publ. No. 2003/0195154).
Methods of Making an Immunosuppressive IL-2 variant The immunosuppressive IL-2 variants can be produced using any suitable method known in the art, including those described in U.S. Pat. No. 6,955,807 for producing immunostimulatory IL-2 variants (incorporated herein by reference). Such methods include constructing a DNA sequence encoding the IL-2 variant and expressing those sequences in a suitably transformed host. This method will produce the recombinant variant of this invention. However, the variants may also be produced by chemical synthesis or a combination of chemical synthesis and recombinant DNA technology. Batch-wise production or perfusion production methods are known in the art. See Freshey, R. I. (ed), "Animal Cell Culture: A Practical Approach," 2nd ed., 1992, IRL Press. Oxford, England; Mather, J. P. "Laboratory Scaleup of Cell Cultures (0.5-50 liters)," Methods Cell Biolog 57: 219- 527 (1998); Hu, W. S., and Aunins, J. G., "Large-scale Mammalian Cell Culture," Curr Opin Biotechnol 8: 148-153 (1997); Konstantinov, K. B., Tsai, Y., Moles, D., Matanguihan, R., "Control of long-term perfusion Chinese hamster ovary cell culture by glucose auxostat.," Biotechnol Prog 12:100-109 (1996). In one embodiment of a recombinant method for producing a variant, a DNA sequence is constructed by isolating or synthesizing a DNA sequence encoding the wild type IL-2 and then changing one or more codons by site-specific mutagenesis. This technique is well known. See, e.g., Mark et. al., "Site-specific Mutagenesis Of The Human Fibroblast Interferon Gene", Proc. Natl. Acad. Sci. USA 81, pp. 5662-66 (1984); and U.S. Pat. No. 4,588,585, incorporated herein by reference.
Another method of constructing a DNA sequence encoding the IL-2 variant would be chemical synthesis. This for example includes direct synthesis of a peptide by chemical means of the protein sequence encoding for an IL-2 variant exhibiting the properties described herein. This method may incorporate both natural and unnatural amino acids at positions that affect the interactions of IL-2 with the IL2Rα, IL-2Rβ, or IL-2Rγ. Alternatively, a gene which encodes the desired IL-2 variant may be synthesized by chemical means using an oligonucleotide synthesizer. Such oligonucleotides are designed based on the amino acid sequence of the desired IL-2 variant, and preferably selecting those codons that are favored in the host cell in which the recombinant variant will be produced. In this regard, it is well recognized that the genetic code is degenerate—that an amino acid may be coded for by more than one codon. For example, Phe (F) is coded for by two codons, TTC or TTT, Tyr (Y) is coded for by TAC or TAT and his (H) is coded for by CAC or CAT. Trp (W) is coded for by a single codon, TGG. Accordingly, it will be appreciated that for a given DNA sequence encoding a particular IL-2 variant, there will be many DNA degenerate sequences that will code for that IL-2 variant.
The DNA sequence encoding the IL-2 variant, whether prepared by site directed mutagenesis, chemical synthesis or other methods, may or may not also include DNA sequences that encode a signal sequence. Such signal sequence, if present, should be one recognized by the cell chosen for expression of the IL-2 variant. It may be prokaryotic, eukaryotic or a combination of the two. It may also be the signal sequence of native IL-2. The inclusion of a signal sequence depends on whether it is desired to secrete the IL-2 variant from the recombinant cells in which it is made. If the chosen cells are prokaryotic, it generally is preferred that the DNA sequence not encode a signal sequence. If the chosen cells are eukaryotic, it generally is preferred that a signal sequence be encoded and most preferably that the wild-type IL-2 signal sequence be used.
Standard methods may be applied to synthesize a gene encoding an IL-2 variant. For example, the complete amino acid sequence may be used to construct a back-translated gene. A DNA oligomer containing a nucleotide sequence coding for an IL-2 variant may be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide may be synthesized and then ligated. The individual oligonucleotides typically contain 5 ' or 3' overhangs for complementary assembly.
Once assembled (by synthesis, site-directed mutagenesis or another method), the DNA sequences encoding an IL-2 variant will be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the IL-2 variant in the desired transformed host. Proper assembly may be confirmed by nucleotide sequencing, restriction mapping, and expression of a biologically active polypeptide in a suitable host. As is well known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to transcriptional and translational expression control sequences that are functional in the chosen expression host. The choice of expression control sequence and expression vector will depend upon the choice of host. A wide variety of expression host/vector combinations may be employed.
Any suitable host may be used to produce the IL-2 variant, including bacteria, fungi (including yeasts), plant, insect, mammal, or other appropriate animal cells or cell lines, as well as transgenic animals or plants. More particularly, these hosts may include well known eukaryotic and prokaryotic hosts, such as strains of E.coli, Pseudomonas, Bacillus, Streptomyces, fungi, yeast, insect cells such as Spodoptera frugiperda (Sf9), animal cells such as Chinese hamster ovary (CHO) and mouse cells such as NS/O, African green monkey cells such as COS 1, COS 7, BSC 1, BSC 40, and BNT 10, and human cells, as well as plant cells in tissue culture. For animal cell expression, CHO cells and COS 7 cells in cultures and particularly the CHO cell line CHO (DHFR-) or the HKB line are preferred.
It should of course be understood that not all vectors and expression control sequences will function equally well to express the DNA sequences described herein. Neither will all hosts function equally well with the same expression system. However, one of skill in the art may make a selection among these vectors, expression control sequences and hosts without undue experimentation. For example, in selecting a vector, the host must be considered because the vector must replicate in it. The vectors copy number, the ability to control that copy number, and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered. For example, preferred vectors for use in this invention include those that allow the DNA encoding the IL-2 variants to be amplified in copy number. Such amplifiable vectors are well known in the art. They include, for example, vectors able to be amplified by DHFR amplification (see, e.g., Kaufman, U.S. Pat. No. 4,470,461, Kaufman and Sharp, "Construction OfA Modular Dihydrafolate Reductase cDNA Gene: Analysis Of Signals Utilized For Efficient Expression", MoI. Cell. Biol., 2, pp. 1304-19 (1982)) or glutamine synthetase ("GS") amplification (see, e.g., U.S. Pat. No. 5,122,464 and European published application 338,841). The IL-2 variants may be glycosylated or unglycosylated depending on the host organism used to produce the variant. If bacteria are chosen as the host, then the IL-2 variant produced will be unglycosylated. Eukaryotic cells, on the other hand, will glycosylate the IL-2 variant, although perhaps not in the same way as native IL-2 is glycosylated. The IL-2 variant produced by the transformed host can be purified according to any suitable method. Various methods are known for purifying IL-2. See, e.g Current Protocols in Protein Science, VoI 2. Eds: John E. Coligan, Ben M. Dunn, Hidde L. Ploehg, David W. Speicher, Paul T. Wingfϊeld, Unit 6.5 (Copyright 1997, John Wiley and Sons, Inc). The biological activity of the IL-2 variants can be assayed by any suitable method known in the art. Such assays include those described in the Examples below.
Indications
Diseases, disorders, or conditions may be amenable to treatment with or may be prevented by administration of a T-reg-selective IL-2 variant to a subject. Such diseases, disorders, and conditions include, but are not limited to, inflammation, autoimmune disease, paraneoplastic autroimmune diseases, cartilage inflammation, fibrotic disease and/or bone degradation, arthritis, rheumatoid arthritis, juvenile arthritis, juvenile rheumatoid arthritis, pauciarticular juvenile rheumatoid arthritis, polyarticular juvenile rheumatoid arthritis, systemic onset juvenile rheumatoid arthritis, juvenile ankylosing spondylitis, juvenile enteropathic arthritis, juvenile reactive arthritis, juvenile Reter's Syndrome, SEA Syndrome (Seronegativity, Enthesopathy, Arthropathy Syndrome), juvenile dermatomyositis, juvenile psoriatic arthritis, juvenile scleroderma, juvenile systemic lupus erythematosus, juvenile vasculitis, pauciarticular rheumatoid arthritis, polyarticular rheumatoid arthritis, systemic onset rheumatoid arthritis, ankylosing spondylitis, enteropathic arthritis, reactive arthritis, Reter's Syndrome, SEA Syndrome (Seronegativity, Enthesopathy, Arthropathy Syndrome), dermatomyositis, psoriatic arthritis, scleroderma, systemic lupus erythematosus, vasculitis, myolitis, polymyolitis, dermatomyolitis, osteoarthritis, polyarteritis nodossa, Wegener's granulomatosis, arteritis, ploymyalgia rheumatica, sarcoidosis, scleroderma, sclerosis, primary biliary sclerosis, sclerosing cholangitis, Sjogren's syndrome, psoriasis, plaque psoriasis, guttate psoriasis, inverse psoriasis, pustular psoriasis, erythrodermic psoriasis, dermatitis, atopic dermatitis, atherosclerosis, lupus, Still's disease, Systemic Lupus Erythematosus (SLE), myasthenia gravis, inflammatory bowel disease (IBD), Crohn's disease, ulcerative colitis, celiac disease, multiple schlerosis (MS), asthma, COPD, Guillain-Barre disease, Type I diabetes mellitus, thyroiditis(e.g., Graves' disease), Addison's disease, Raynaud's phenomenon, autoimmune hepatitis, GVHD, transplantation rejection, and the like. In specific embodiments, pharmaceutical compositions comprising a therapeutically effective amount of a T-reg-selective IL-2 variant are provided.
The term "treatment" encompasses alleviation or prevention of at least one symptom or other aspect of a disorder, or reduction of disease severity, and the like. A T-reg-selective IL-2 variant need not effect a complete cure, or eradicate every symptom or manifestation of a disease, to constitute a viable therapeutic agent. As is recognized in the pertinent field, drugs employed as therapeutic agents may reduce the severity of a given disease state, but need not abolish every manifestation of the disease to be regarded as useful therapeutic agents. Similarly, a prophylactically administered treatment need not be completely effective in preventing the onset of a condition in order to constitute a viable prophylactic agent. Simply reducing the impact of a disease (for example, by reducing the number or severity of its symptoms, or by increasing the effectiveness of another treatment, or by producing another beneficial effect), or reducing the likelihood that the disease will occur or worsen in a subject, is sufficient. One embodiment of the invention is directed to a method comprising administering to a patient A T-reg-selective IL-2 variant in an amount and for a time sufficient to induce a sustained improvement over baseline of an indicator that reflects the severity of the particular disorder.
Pharmaceutical Compositions In some embodiments, the invention provides pharmaceutical compositions comprising a therapeutically effective amount of one or a plurality of T-reg-selective IL-2 variants of the invention together with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative, and/or adjuvant. In addition, the invention provides methods of treating a patient by administering such pharmaceutical composition. The term "patient" includes human and animal subjects.
In certain embodiments, acceptable formulation materials preferably are nontoxic to recipients at the dosages and concentrations employed. In certain embodiments, the pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolality, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition. In such embodiments, suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta- cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides; and other carbohydrates (such as glucose, sucrose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emulsifying agents; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; salt-forming counterions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (such as sucrose or sorbitol); tonicity enhancing agents (such as alkali metal halides, preferably sodium or potassium chloride, mannitol sorbitol); delivery vehicles; diluents; excipients and/or pharmaceutical adjuvants. See, REMINGTON'S PHARMACEUTICAL SCIENCES, 18" Edition, (A.R. Genrmo, ed.), 1990, Mack Publishing Company. The therapeutically effective amount of T-reg-selective IL-2 variant- containing pharmaceutical composition to be employed will depend, for example, upon the therapeutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment will vary depending, in part, upon the molecule delivered, the indication for which the T-reg-selective IL-2 variant is being used, the route of administration, and the size (body weight, body surface or organ size) and/or condition (the age and general health) of the patient.
In certain embodiments, the clinician may titer the dosage and modify the route of administration to obtain the optimal therapeutic effect. A typical dosage may range from about 0.1 μg/kg to up to about 30 mg/kg or more, depending on the factors mentioned above. In specific embodiments, the dosage may range from 0.1 μg/kg up to about 30 mg/kg, optionally from 1 μg/kg up to about 30 mg/kg or from 10 μg/kg up to about 5 mg/kg.
Dosing frequency will depend upon the pharmacokinetic parameters of the particular T-reg-selective IL-2 variant in the formulation used. Typically, a clinician administers the composition until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose, or as two or more doses (which may or may not contain the same amount of the desired molecule) over time, or as a continuous infusion via an implantation device or catheter. Further refinement of the appropriate dosage is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them.
The route of administration of the pharmaceutical composition is in accord with known methods, e.g., orally, through injection by intravenous, intraperitoneal, intracerebral (intra- parenchymal), intracerebroventricular, intramuscular, intra-ocular, intraarterial, intraportal, or intralesional routes; by sustained release systems or by implantation devices. In certain embodiments, the compositions may be administered by bolus injection or continuously by infusion, or by implantation device.
Combination therapies In further embodiments, T-reg-selective IL-2 variant is administered in combination with other agents useful for treating the condition with which the patient is afflicted. Examples of such agents include both proteinaceous and non- proteinaceous drugs. When multiple therapeutics are co-administered, dosages may be adjusted accordingly, as is recognized in the pertinent art. "Co-administration" and combination therapy are not limited to simultaneous administration, but also include treatment regimens in which a T-reg-selective IL-2 variant is administered at least once during a course of treatment that involves administering at least one other therapeutic agent to the patient.
In certain embodiments, a T-reg-selective IL-2 variant is administered in combination with an inhibitor of the PI3-K/AKT/mTOR pathway, e.g., rapamycin (rapamune, sirolimus). Inhibitors of this pathway in combination with IL-2 favor T- reg enrichment.
The invention having been described, the following examples are offered by way of illustration, and not limitation. Examples
Example 1 : Panel of IL-2 mutants.
To examine the potential for generating IL-2 variants with reduced capacity to stimulate FOXP3 CD25+ "effector" T cells (T-eff) but not T-reg, a series of IL-2 mutants was generated in which amino acids predicted to interact with the IL-2Rβ and/or IL-2Rγ chain were altered. These variants also contained a set of previously described mutations that conferred high affinity for CD25 (variant "2-4" in Rao et al., Biochemistry 44, 10696-701 (2005)). This series of variants is shown in Figure 1. Variant ha WT contained only the mutations that contributed to the high affinity for CD25. Variants haD, haD.l, haD.2, etc, also contained mutations predicted to alter interactions with IL-2Rβ and/or IL-2Rγ. In all assays, variant haD.11 was not capable of inducing any signal or altering any cellular phenotype and, as such, served as a control for CD25 binding without IL-2R signaling. All the IL-2 variants contained the C 125 S mutation for improved manufacturability and terminated with FLAG and HIS-tag sequences (DYKDDDDKG S SHHHHHH) (SEQ ID NO:11)
Several assays were used to assess the ability of the IL-2 variants to induce signaling events and T cell growth. These included assays to detect:
1. Growth and survival of T cell subsets and measurement of FOXP3 expression.
2. Cell signaling (e.g. detection of phosphorylated STAT5 and AKT using flow cytometric and ELISA-based methods).
Example 2: Enrichment of FOXP3+ cells and retention of FOXP3 upregulation during long term T cell culture.
Total PBMC were activated in 24-well plates at 4x106 cells per well with 100 ng/ml anti-CD3 (OKT3). On day 3 of culture, cells were washed 3 times and rested in fresh media for 3 days. Cells were then washed and seeded in 96 well flat-bottom plates with IL-2 variants at either 10 nM or 100 pM. Three days later cells were counted and analyzed by flow cytometry. (FIG. 2A)
As expected, CD8+CD25+ T cells were especially responsive to WT IL-2 and variant ha WT, however, all variants that contained mutated IL-2Rβ and/or γ contact residues were very inefficient at promoting accumulation of activated CD8 CD25+ T cells (FIG. 2B). A similar trend was observed for CD4+CD25+FOXP3 T cells (FIG. 2C). In contrast, the growth/survival of FOXP3+ CD4+ T cells was stimulated by several IL-2 variants to a degree similar to that of WT IL-2 (FIG. 2D). As a result, the ratio of FOXP3+ to FOXP3 T cells among CD4+ CD25+ T cells was increased by several IL-2 variants with IL-2Rβγ-contact residue mutations (FIG. 2E). Furthermore, the mutations did not impair IL-2-stimulated FOXP3 upregulation in T-reg (FIG. 2F).
Example 3 : Mutations that reduce signaling in FOXP3 T cells but stimulate STAT5 signaling in T-reg.
The IL-2 variants were screened for their ability to stimulate AKT and STAT5 phosphorylation in T cell subsets. Several IL-2 variants were as potent, or nearly as potent, as wt IL-2 at stimulating STAT5 in FOXP3+ T cells 10 min after stimulation. Three hours after washing IL-2 from the media, some IL-2 variants (haD, haD.l, haD.2, haD.4, haD.6, and haD.8) continued to stimulate sustained STAT5 signaling at levels higher than that seen with wt IL-2. In contrast, for FOXP3 T cells, STAT5 and AKT responses to the haD variants after 10 min stimulation were not nearly as high as those stimulated by wt IL-2 or ha WT. After 3 hrs, weak STAT5 and AKT signals similar to those seen with wt IL-2 were observed in T-eff, however, at this late timepoint wt IL-2 signaling had diminished greatly. In FOXP3+ T cells, AKT signaling is not normally stimulated by IL-2 (Zeiser R, et al, 2008 Blood 111 :453) thus the phospho-AKT signal observed in total T cell lysates can be attributed to T-eff.
Methods: Previously activated (with anti-CD3 for 2-3 days) and rested (in fresh culture medium for 2-5 days) T cells were exposed to 1 nM wt or mutant IL-2 for 10 min at 37°C. Cells were then stained (10 min timepoint) or washed and cultured for an additional 3 hrs (3 hr timepoint). To measure phospho-AKT by ELISA, a 50 μl culture was stopped by adding an equal volume of 2x lysis buffer and lysates were measured for phospho-AKT with multiplex ELISA plates according to the manufacturer's protocols (MesoScale Discovery, Gaithersburg, Maryland). To measure phospho-STAT5 by flow cytometry, a 50 μl culture was stopped by adding 1 ml of FOXP3 Fix/Perm Buffer (BioLegend, San Diego, CA), incubation at 25°C for 20 min, and staining for cell surface markers, FOXP3 and phospho-STAT5 according the BioLegend FOXP3 staining protocol.

Claims

Claims
1. A method of treating an inflammatory disorder in a subject, said method comprising administering to a subject in need thereof a therapeutically effective amount of an IL-2 variant, wherein said IL-2 variant
(a) comprises a sequence of amino acids at least 80% identical to SEQ ID NO:1;
(b) stimulates STAT5 phosphorylation in FOXP3 -positive regulatory T cells; and
(c) has a reduced ability compared to the polypeptide set forth as SEQ ID NO:1 to induce phosphorylation of STAT5 in FOXP3 -negative T cells.
2. The method of claim 1, wherein the inflammatory disorder is selected from the group consisting of asthma, diabetes, arthritis, allergy, organ graft rejection and graft- versus-host disease.
3. The method of claim 1, wherein said IL-2 variant comprises a sequence of amino acids at least 90% identical to SEQ ID NO: 1.
4. The method of claim 1, wherein said IL-2 variant comprises a sequence of amino acids at least 95% identical to SEQ ID NO: 1.
5. The method of claim 1 , wherein the IL-2 variant has a higher affinity for IL- 2Rα than does the polypeptide set forth as SEQ ID NO:1;
6. The method of claim 1 , wherein the IL-2 variant promotes FOXP3 -positive regulatory T cell growth or survival in vitro.
7. The method of claim 1 , wherein the IL-2 variant comprises a mutation in the polypeptide sequence set forth in SEQ ID NO:1 at a position selected from the group consisting of amino acid 30, amino acid 31, amino acid 35, amino acid 69, and amino acid 74.
8. The method of claim 7, wherein the mutation at position 30 is N30S.
9. The method of claim 7, wherein the mutation at position 31 is Y3 IH.
10. The method of claim 7, wherein the mutation at position 35 is K35R.
11. The method of claim 7, wherein the mutation at position 69 is V69A.
12. The method of claim 7, wherein the mutation at position 74 is Q74P.
13. The method of claim 1 , wherein the IL-2 variant induces STAT5 phosphorylation in ex vivo FOXP3-positive T cells comprising a functional IL- 2 receptor complex but has a reduced ability to induce phosphorylation of STAT5.
14. The method of claim 13, wherein the IL-2 variant comprises a mutation in the polypeptide sequence set forth in SEQ ID NO:1 at position 88.
15. The method of claim 14, wherein the mutation at position 88 is N88D.
16. The method of claim 1, wherein the IL-2 variant is conjugated to a chemical or polypeptide that extends the serum half- life of said IL-2 variant in vivo.
17. A method of promoting FOXP3-postive regulatory T cell growth or survival, said method comprising contacting a FOXP3 -positive regulatory T cell with an IL-2 variant, wherein said IL-2 variant
(a) comprises a sequence of amino acids at least 80% identical to SEQ ID NO:1;
(b) stimulates STAT5 phosphorylation in said FOXP3 -positive regulatory T cells; and
(c) has a reduced ability compared to the polypeptide set forth as SEQ ID NO:1 to induce phosphorylationof STAT5 in FOXP3 -negative T cells.
18. The method of claim 16, wherein the FOXP3 -positive regulatory T cell is contacted in vitro.
19. The method of claim 18, wherein the IL-2 variant is conjugated to a chemical or polypeptide that extends the serum half-life of said IL-2 variant in vivo.
20. Use of an IL-2 variant in the preparation of a medicament for the treatment of an inflammatory disease, wherein said IL-2 variant
(a) comprises a sequence of amino acids at least 80% identical to SEQ ID NO:1;
(b) stimulates STAT5 phosphorylation in FOXP3 -positive regulatory T cells; and
(c) has a reduced ability compared to the polypeptide set forth as SEQ ID NO:1 to induce phosphorylation of STAT5 in FOXP3 -negative T cells.
21. The method of claim 20, wherein the IL-2 variant is conjugated to a chemical or polypeptide that extends the serum half-life of said IL-2 variant in vivo.
PCT/US2010/021519 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases WO2010085495A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
EP10733814.7A EP2382228B1 (en) 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases
JP2011548074A JP5766124B2 (en) 2009-01-21 2010-01-20 Compositions and methods for the treatment of inflammatory and autoimmune diseases
MX2011007647A MX2011007647A (en) 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases.
ES10733814T ES2825173T3 (en) 2009-01-21 2010-01-20 Compositions and methods of treatment of inflammatory and autoimmune diseases
CA2749539A CA2749539C (en) 2009-01-21 2010-01-20 Compositions and methods comprising interleukin-2 mutants for treating inflammatory and autoimmune diseases
US13/145,537 US20110274650A1 (en) 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases
AU2010206840A AU2010206840B2 (en) 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases
US13/672,417 US9732134B2 (en) 2009-01-21 2012-11-08 Method of treating graft versus host disease using IL-2 muteins
US15/649,520 US11560415B2 (en) 2009-01-21 2017-07-13 Method of promoting regulatory T-cell proliferation
US18/068,970 US20230139534A1 (en) 2009-01-21 2022-12-20 Methods of treating graft versus host disease using il-2 muteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US14611109P 2009-01-21 2009-01-21
US61/146,111 2009-01-21

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/145,537 A-371-Of-International US20110274650A1 (en) 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases
US13/672,417 Continuation US9732134B2 (en) 2009-01-21 2012-11-08 Method of treating graft versus host disease using IL-2 muteins

Publications (1)

Publication Number Publication Date
WO2010085495A1 true WO2010085495A1 (en) 2010-07-29

Family

ID=42356180

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/021519 WO2010085495A1 (en) 2009-01-21 2010-01-20 Compositions and methods of treating inflammatory and autoimmune diseases

Country Status (8)

Country Link
US (4) US20110274650A1 (en)
EP (1) EP2382228B1 (en)
JP (6) JP5766124B2 (en)
AU (1) AU2010206840B2 (en)
CA (1) CA2749539C (en)
ES (1) ES2825173T3 (en)
MX (1) MX2011007647A (en)
WO (1) WO2010085495A1 (en)

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2014500868A (en) * 2010-11-12 2014-01-16 セントロ ド インムノロジア モレキュラー IL-2 derived polypeptides with agonist activity for the treatment of cancer and chronic infections
WO2014153063A1 (en) 2013-03-14 2014-09-25 Amgen Inc. AGLYCOSYLATED Fc-CONTAINING POLYPEPTIDES
WO2015118016A1 (en) * 2014-02-06 2015-08-13 F. Hoffmann-La Roche Ag Interleukin-2 fusion proteins and uses thereof
US9266938B2 (en) 2011-02-10 2016-02-23 Roche Glycart Ag Mutant interleukin-2 polypeptides
WO2016164937A2 (en) 2015-04-10 2016-10-13 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US9669071B2 (en) 2011-03-11 2017-06-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for stimulating T lymphocytes with IL-2
US9732134B2 (en) 2009-01-21 2017-08-15 Amgen Inc. Method of treating graft versus host disease using IL-2 muteins
US10035836B1 (en) 2014-08-11 2018-07-31 Delinia, Inc. Modified IL-2 variants that selectively activate regulatory T cells
EP2683395B1 (en) 2011-03-11 2018-08-01 Assistance Publique - Hôpitaux de Paris Use of low dose il-2 for treating type 1 diabetes
US10202464B2 (en) 2011-04-29 2019-02-12 Roche Glycart Ag Immunoconjugates
US10294287B2 (en) 2016-01-20 2019-05-21 Delinia, Inc. Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
WO2019131964A1 (en) 2017-12-27 2019-07-04 協和発酵キリン株式会社 Il-2 variant
WO2019158764A1 (en) 2018-02-16 2019-08-22 Iltoo Pharma Use of interleukin 2 for treating sjögren's syndrome
WO2020007937A1 (en) 2018-07-03 2020-01-09 Iltoo Pharma Use of interleukin-2 for treating systemic sclerosis
US10562949B2 (en) 2012-08-10 2020-02-18 Roche Glycart Ag Interleukin-2 fusion proteins and uses thereof
WO2020035482A1 (en) 2018-08-13 2020-02-20 Iltoo Pharma Combination of interleukin-2 with an interleukin 1 inhibitor, conjugates and therapeutic uses thereof
US10610571B2 (en) 2017-08-03 2020-04-07 Synthorx, Inc. Cytokine conjugates for the treatment of proliferative and infectious diseases
US10654905B2 (en) 2014-04-24 2020-05-19 The Board Of Trustees Of The Leland Stanford Junior University Method of treating graft versus host disease with an interleukin-2 mutein
WO2020201095A1 (en) 2019-03-29 2020-10-08 Institut Curie Interleukin-2 variants with modified biological activity
US10851144B2 (en) 2015-04-10 2020-12-01 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
CN112279906A (en) * 2020-10-30 2021-01-29 浙江新码生物医药有限公司 Human interleukin 2-polyethylene glycol conjugate, preparation method and application thereof
WO2021030602A1 (en) 2019-08-13 2021-02-18 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US10946068B2 (en) 2017-12-06 2021-03-16 Pandion Operations, Inc. IL-2 muteins and uses thereof
US10961310B2 (en) 2017-03-15 2021-03-30 Pandion Operations, Inc. Targeted immunotolerance
WO2021116444A1 (en) 2019-12-12 2021-06-17 Iltoo Pharma Interleukin 2 chimeric constructs
WO2021127262A1 (en) 2019-12-17 2021-06-24 Amgen Inc. Dual interleukin-2 /tnf receptor agonist for use in therapy
US11077195B2 (en) 2019-02-06 2021-08-03 Synthorx, Inc. IL-2 conjugates and methods of use thereof
US11077172B2 (en) 2016-11-08 2021-08-03 Delinia, Inc. IL-2 variants for the treatment of psoriasis
AU2019264645B2 (en) * 2015-04-10 2021-08-12 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US11091526B2 (en) 2017-12-06 2021-08-17 Pandion Operations, Inc. IL-2 muteins and uses thereof
US20220002672A1 (en) * 2011-12-16 2022-01-06 The Usa, As Represented By The Secretary, Department Of Health And Human Services. Methods of producing t cell populations enriched for stable regulatory t-cells
US11248046B2 (en) 2019-02-15 2022-02-15 Integral Molecular, Inc. Claudin 6 antibodies and uses thereof
US11254736B2 (en) 2019-02-15 2022-02-22 Integral Molecular, Inc. Antibodies comprising a common light chain and uses thereof
WO2022013696A3 (en) * 2020-07-14 2022-02-24 Pfizer Inc. Recombinant vaccinia virus
WO2022043493A1 (en) * 2020-08-28 2022-03-03 Ascendis Pharma Oncology Division A/S Glycosylated il-2 proteins and uses thereof
CN114133456A (en) * 2014-07-21 2022-03-04 德里尼亚公司 Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
WO2022120224A1 (en) * 2020-12-04 2022-06-09 Visterra, Inc. Methods of using interleukin-2 agents
US11466068B2 (en) 2017-05-24 2022-10-11 Pandion Operations, Inc. Targeted immunotolerance
US11479595B2 (en) 2017-03-15 2022-10-25 Cue Biopharma, Inc. Methods for modulating an immune response
US11505588B2 (en) 2016-12-22 2022-11-22 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11505591B2 (en) 2016-05-18 2022-11-22 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11542312B2 (en) 2017-06-19 2023-01-03 Medicenna Therapeutics, Inc. IL-2 superagonists in combination with anti-PD-1 antibodies
WO2023057588A1 (en) 2021-10-06 2023-04-13 Iltoo Pharma Interleukin 2 chimeric constructs with targeting specificy to inflamed tissues
US11680090B2 (en) 2013-09-24 2023-06-20 Medicenna Therapeutics, Inc. Interleukin-2 fusion proteins and uses thereof
US11692020B2 (en) 2019-11-20 2023-07-04 Anwita Biosciences, Inc. Cytokine fusion proteins, and their pharmaceutical compositions and therapeutic applications
US11702461B2 (en) 2018-01-09 2023-07-18 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides comprising reduced-affinity immunomodulatory polypeptides
US11725034B2 (en) 2019-12-20 2023-08-15 Regeneron Pharmaceuticals, Inc. IL2 agonists and methods of use thereof
US11739146B2 (en) 2019-05-20 2023-08-29 Pandion Operations, Inc. MAdCAM targeted immunotolerance
US11827684B2 (en) 2020-04-22 2023-11-28 Merck Sharp & Dohme Llc Human interleukin-2 conjugates biased for the interleukin-2 receptor beta GAMMAc dimer and conjugated to a nonpeptidic, water-soluble polymer
US11834689B2 (en) 2017-07-11 2023-12-05 The Scripps Research Institute Incorporation of unnatural nucleotides and methods thereof
US11851471B2 (en) 2017-01-09 2023-12-26 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11879001B2 (en) 2020-06-03 2024-01-23 Ascendis Pharma Oncology Division A/S Conjugate comprising an IL-2 moiety
US11878062B2 (en) 2020-05-12 2024-01-23 Cue Biopharma, Inc. Multimeric T-cell modulatory polypeptides and methods of use thereof
US11897930B2 (en) 2020-04-28 2024-02-13 Anwita Biosciences, Inc. Interleukin-2 polypeptides and fusion proteins thereof, and their pharmaceutical compositions and therapeutic applications
WO2024056154A1 (en) 2022-09-12 2024-03-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Interleukin-2 for use in treating autism spectrum disorder
US11981717B2 (en) 2017-12-19 2024-05-14 Xencor, Inc. Engineered IL-2 Fc fusion proteins
US11981715B2 (en) 2020-02-21 2024-05-14 Pandion Operations, Inc. Tissue targeted immunotolerance with a CD39 effector
US12006347B2 (en) 2010-12-22 2024-06-11 The Board Of Trustees Of The Leland Stanford Junior University Superagonists and antagonists of interleukin-2
US12024559B2 (en) 2020-10-23 2024-07-02 Asher Biotherapeutics, Inc. Fusions with CD8 antigen binding molecules for modulating immune cell function
US12029782B2 (en) 2020-09-09 2024-07-09 Cue Biopharma, Inc. MHC class II T-cell modulatory multimeric polypeptides for treating type 1 diabetes mellitus (T1D) and methods of use thereof
WO2024151515A2 (en) 2023-01-09 2024-07-18 Odyssey Therapeutics, Inc. Anti-tnfr2 antigen-binding proteins and uses thereof
US12049502B2 (en) 2023-11-29 2024-07-30 Integral Molecular, Inc. Antibodies directed to claudin 6, including bispecific formats thereof

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200064083A (en) * 2017-09-07 2020-06-05 큐 바이오파마, 인크. Antigen-presenting polypeptides and methods of use thereof
WO2019173832A2 (en) 2018-03-09 2019-09-12 AskGene Pharma, Inc. Novel cytokine prodrugs
AU2019244091B2 (en) 2018-03-28 2023-12-07 Bristol-Myers Squibb Company Interleukin-2/Interleukin-2 receptor alpha fusion proteins and methods of use
EP3875475A4 (en) 2018-12-21 2022-12-07 Jiangsu Hengrui Pharmaceuticals Co., Ltd. Human interleukin-2 variant or derivative thereof
AU2020291012A1 (en) 2019-06-12 2021-12-16 AskGene Pharma, Inc. Novel IL-15 prodrugs and methods of use thereof
US11633488B2 (en) 2020-01-10 2023-04-25 Bright Peak Therapeutics Ag Modified IL-2 polypeptides and uses thereof
JP7303391B2 (en) 2020-01-14 2023-07-04 シンセカイン インコーポレイテッド Biased IL2 muteins, methods, and compositions
JP2023548311A (en) 2020-10-29 2023-11-16 ブリストル-マイヤーズ スクイブ カンパニー Fusion proteins for the treatment of diseases
US20230398183A1 (en) 2020-11-13 2023-12-14 Jiangsu Hengrui Pharmaceuticals Co., Ltd. Pharmaceutical composition comprising human interleukin 2 variant or derivative thereof and use thereof
JP2024526297A (en) 2021-07-09 2024-07-17 ブライト ピーク セラピューティクス エージー Antibody conjugates and their production
WO2023281483A1 (en) 2021-07-09 2023-01-12 Bright Peak Therapeutics Ag Modified tnf-antibodies and uses thereof
WO2023005680A1 (en) 2021-07-30 2023-02-02 西安龙腾景云生物科技有限公司 Human interleukin-2 variant and use thereof
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4470461A (en) 1982-09-30 1984-09-11 Phillips Petroleum Company Organic nitro compounds as cosurfactants in enhanced oil recovery processes
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
EP0338841A1 (en) 1988-04-18 1989-10-25 Celltech Limited Recombinant DNA methods, vectors and host cells
US5122464A (en) 1986-01-23 1992-06-16 Celltech Limited, A British Company Method for dominant selection in eucaryotic cells
US20030195154A1 (en) 2002-04-04 2003-10-16 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
US20050142106A1 (en) 2003-07-18 2005-06-30 Wittrup K. D. Mutant interleukin-2 (IL-2) polypeptides
US6955807B1 (en) 1998-05-15 2005-10-18 Bayer Pharmaceuticals Corporation IL-2 selective agonists and antagonists

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4518584A (en) 1983-04-15 1985-05-21 Cetus Corporation Human recombinant interleukin-2 muteins
EP0262802A3 (en) 1986-09-01 1990-01-31 Takeda Chemical Industries, Ltd. Medicament for the treatment or improvement of rheumatoid arthritis or systemic lupus erythematosus
US5466447A (en) 1988-06-29 1995-11-14 Amgen Inc. Method for treating psoriasis
AU627477B2 (en) 1988-07-05 1992-08-27 Amgen, Inc. Interleukin ii analogs
CA2062582C (en) 1991-03-27 1996-03-26 Tse-Wen Chang Methods and substances for recruiting therapeutic agents to solid tissues
US5229109A (en) 1992-04-14 1993-07-20 Board Of Regents, The University Of Texas System Low toxicity interleukin-2 analogues for use in immunotherapy
DZ2788A1 (en) 1998-05-15 2003-12-01 Bayer Ag Selective IL-2 agonists and antagonists.
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6348192B1 (en) 1999-05-11 2002-02-19 Bayer Corporation Interleukin-2 mutein expressed from mammalian cells
WO2001058957A2 (en) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Enhancing the circulating half-life of antibody-based fusion proteins
WO2003015697A2 (en) 2001-08-13 2003-02-27 University Of Southern California Interleukin-2 mutants with reduced toxicity
HU229098B1 (en) 2001-12-04 2013-07-29 Merck Patent Gmbh Immunocytokines with modulated selectivity
AR043504A1 (en) 2003-03-17 2005-08-03 Novartis Ag PHARMACEUTICAL COMPOSITIONS THAT INCLUDE RAPAMYCIN FOR THE TREATMENT OF INFLAMMATORY DISEASES
US20060234205A1 (en) 2004-03-05 2006-10-19 Chiron Corporation In vitro test system for predicting patient tolerability of therapeutic agents
AU2006232374B2 (en) * 2005-04-01 2011-08-25 University Of Southern California Preventing rejection of transplanted tissue using regulatory T cells
EP1987065A4 (en) 2006-02-16 2010-01-20 Nascent Biolog Inc Methods for improving immune function and methods for prevention or treatment of disease in a mammalian subject
EP1988921A4 (en) 2006-02-28 2010-02-10 Elan Pharm Inc Methods of treating inflammatory and autoimmune diseases with natalizumab
EP2038417A2 (en) 2006-07-06 2009-03-25 Merck Patent GmbH Compositions and methods for enhancing the efficacy of il-2 mediated immune responses
CN101479570A (en) 2006-07-06 2009-07-08 三菱电机株式会社 Navigation system
WO2008112325A2 (en) 2007-03-15 2008-09-18 Biogen Idec Ma Inc. Treatment of autoimmune disorders
GB0623539D0 (en) 2006-11-24 2007-01-03 Avidex Ltd Polypeptides
RU2009135824A (en) 2007-02-27 2011-04-10 Дженентек, Инк. (Us) ANTIBODIES ANTAGONISTS AGAINST OX40 AND THEIR APPLICATION IN TREATMENT OF INFLAMMATORY AND AUTOIMMUNE DISEASES
US8906356B2 (en) 2007-11-05 2014-12-09 Massachusetts Institute Of Technology Mutant interleukin-2 (IL-2) polypeptides
JP6157046B2 (en) 2008-01-07 2017-07-05 アムジェン インコーポレイテッド Method for generating antibody Fc heterodimer molecules using electrostatic steering effect
DE102008023820A1 (en) 2008-05-08 2009-11-12 Aicuris Gmbh & Co. Kg An agent for the treatment and / or prophylaxis of an autoimmune disease and for the production of regulatory T cells
WO2010017220A1 (en) 2008-08-04 2010-02-11 The Trustees Of The University Of Pennsylvania Methods for expanding regulatory t-cells
US20110200534A1 (en) 2008-08-21 2011-08-18 Industry-Academic Cooperation Foundation, Yonsei U T1-T2 Dual Modal MRI Contrast Agents
ES2825173T3 (en) 2009-01-21 2021-05-14 Amgen Inc Compositions and methods of treatment of inflammatory and autoimmune diseases
AU2012228418B2 (en) 2011-03-11 2016-09-29 Assistance Publique - Hopitaux De Paris Use of low dose IL-2 for treating autoimmune - related or inflammatory disorders
CA2830254C (en) 2011-03-16 2019-09-10 Amgen Inc. Fc variants

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4470461A (en) 1982-09-30 1984-09-11 Phillips Petroleum Company Organic nitro compounds as cosurfactants in enhanced oil recovery processes
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US5122464A (en) 1986-01-23 1992-06-16 Celltech Limited, A British Company Method for dominant selection in eucaryotic cells
EP0338841A1 (en) 1988-04-18 1989-10-25 Celltech Limited Recombinant DNA methods, vectors and host cells
US6955807B1 (en) 1998-05-15 2005-10-18 Bayer Pharmaceuticals Corporation IL-2 selective agonists and antagonists
US20030195154A1 (en) 2002-04-04 2003-10-16 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
US20050142106A1 (en) 2003-07-18 2005-06-30 Wittrup K. D. Mutant interleukin-2 (IL-2) polypeptides

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Animal Cell Culture: A Practical Approach", 1992, IRL PRESS
"Current Protocols in Protein Science", vol. 2, 1997, JOHN WILEY AND SONS, INC, article "Unit 6.5"
A.R. GENARO: "REMINGTON'S PHARMACEUTICAL SCIENCES", 1990, MACK PUBLISHING COMPANY
FUJII ET AL.: "Activation of Stat5 by interleukin 2 requires a carboxyl-terminal region of the interleukin 2 receptor beta chain but is not essential for the proliferative signal transmission.", PNAS, vol. 92, 1995, pages 5482 - 5486 *
HU, W. S.; AUNINS, J. G.: "Large-scale Mammalian Cell Culture", CURR OPIN BIOTECHNOL, vol. 8, 1997, pages 148 - 153
KAUFMAN; SHARP: "Construction Of A Modular Dihydrafolate Reductase cDNA Gene: Analysis Of Signals Utilized For Efficient Expression", MOL. CELL. BIOL., vol. 2, 1982, pages 1304 - 1319, XP001007141
KONSTANTINOV, K. B. ET AL: "Control of long-term perfusion Chinese hamster ovary cell culture by glucose auxostat.", BIOTECHNOL PROG, vol. 12, 1996, pages 100 - 109, XP002285656, DOI: doi:10.1021/bp950044p
MARK: "Site-specific Mutagenesis Of The Human Fibroblast Interferon Gene", PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 5662 - 5666, XP002554304, DOI: doi:10.1073/pnas.81.18.5662
MATHER, J. P.: "Laboratory Scaleup of Cell Cultures (0.5-50 liters)", METHODS CELL BIOLOG, vol. 57, no. 57, February 1998 (1998-02-01), pages 219 - 227, XP008093720, DOI: doi:10.1016/S0091-679X(08)61581-2
NAGASE ET AL.: "Despite increased CD4+Foxp3+ cells within the infection site, BALB/c IL-4 receptor-deficient mice reveal CD4+ Foxp3-negative T cells as a source of IL-10 in Leishmania major susceptibility.", JOURNAL OF IMMUNOLOGY, vol. 179, 2007, pages 2435 - 2444 *
RAO ET AL., BIOCHEMISTRY, vol. 44, 2005, pages 10696 - 10701
XINQUAN WANG; MATHIAS RICKERT; K. CHRISTOPHER GARCIA, SCIENCE, vol. 310, 2005, pages 1159

Cited By (133)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11560415B2 (en) 2009-01-21 2023-01-24 Amgen Inc. Method of promoting regulatory T-cell proliferation
US9732134B2 (en) 2009-01-21 2017-08-15 Amgen Inc. Method of treating graft versus host disease using IL-2 muteins
EP2382228B1 (en) 2009-01-21 2020-08-26 Amgen Inc. Compositions and methods of treating inflammatory and autoimmune diseases
JP2014500868A (en) * 2010-11-12 2014-01-16 セントロ ド インムノロジア モレキュラー IL-2 derived polypeptides with agonist activity for the treatment of cancer and chronic infections
US12006347B2 (en) 2010-12-22 2024-06-11 The Board Of Trustees Of The Leland Stanford Junior University Superagonists and antagonists of interleukin-2
TWI577801B (en) * 2011-02-10 2017-04-11 羅齊克雷雅公司 Mutant interleukin-2 polypeptides
US11111312B2 (en) 2011-02-10 2021-09-07 Roche Glycart Ag Mutant interleukin-2 polypeptides
US10323098B2 (en) 2011-02-10 2019-06-18 Roche Glycart Ag Mutant interleukin-2 polypeptides
US9266938B2 (en) 2011-02-10 2016-02-23 Roche Glycart Ag Mutant interleukin-2 polypeptides
TWI666027B (en) * 2011-02-10 2019-07-21 羅齊克雷雅公司 Mutant interleukin-2 polypeptides
US10184009B2 (en) 2011-02-10 2019-01-22 Roche Glycart Ag Mutant interleukin-2 polypeptides
EP3443979B1 (en) 2011-03-11 2020-05-27 Assistance Publique - Hôpitaux De Paris Il-2 dosage regimen for treating systemic lupus erythematosus
US10765723B2 (en) 2011-03-11 2020-09-08 INSERM (Institut National de la Santé et de la Recherche Médicale Use of low dose IL-2 for treating autoimmune—related or inflammatory disorders
JP2021006591A (en) * 2011-03-11 2021-01-21 アシスタンス ピュブリク−オピトー ドゥ パリAssistance Publique − Hopitaux De Paris Use of low dose il-2 for treating autoimmune-related or inflammatory disorders
US9669071B2 (en) 2011-03-11 2017-06-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for stimulating T lymphocytes with IL-2
US11559566B2 (en) 2011-03-11 2023-01-24 Inserm (Institut National De La Sante Et De La Recherche Medicale) Use of low dose IL-2 for treating autoimmune-related or inflammatory disorders
US10293028B2 (en) 2011-03-11 2019-05-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of low dose IL-2 for treating autoimmune related or inflammatory disorders
EP2683395B1 (en) 2011-03-11 2018-08-01 Assistance Publique - Hôpitaux de Paris Use of low dose il-2 for treating type 1 diabetes
US11130822B2 (en) 2011-04-29 2021-09-28 Roche Glycart Ag Immunoconjugates
US10316104B2 (en) 2011-04-29 2019-06-11 Roche Glycart Ag Immunoconjugates
US10202464B2 (en) 2011-04-29 2019-02-12 Roche Glycart Ag Immunoconjugates
US20220002672A1 (en) * 2011-12-16 2022-01-06 The Usa, As Represented By The Secretary, Department Of Health And Human Services. Methods of producing t cell populations enriched for stable regulatory t-cells
US11365232B2 (en) 2012-08-10 2022-06-21 Roche Glycart Ag Interleukin-2 fusion proteins and uses thereof
US10562949B2 (en) 2012-08-10 2020-02-18 Roche Glycart Ag Interleukin-2 fusion proteins and uses thereof
WO2014153111A2 (en) 2013-03-14 2014-09-25 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US11976102B2 (en) 2013-03-14 2024-05-07 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US9932380B2 (en) 2013-03-14 2018-04-03 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US9580486B2 (en) 2013-03-14 2017-02-28 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
EA034326B1 (en) * 2013-03-14 2020-01-28 Эмджен Инк. Human interleukin-2 mutein stimulating regulatory t cells and uses thereof
CN105143253A (en) * 2013-03-14 2015-12-09 美国安进公司 Interleukin-2 muteins for the expansion of t-regulatory cells
US9546203B2 (en) 2013-03-14 2017-01-17 Amgen Inc. Aglycosylated Fc-containing polypeptides with cysteine substitutions
KR20150127185A (en) * 2013-03-14 2015-11-16 암젠 인크 AGLYCOSYLATED Fc-CONTAINING POLYPEPTIDES
WO2014153111A3 (en) * 2013-03-14 2014-11-13 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US10093711B2 (en) 2013-03-14 2018-10-09 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
KR102219124B1 (en) 2013-03-14 2021-02-22 암젠 인크 AGLYCOSYLATED Fc-CONTAINING POLYPEPTIDES
WO2014153063A1 (en) 2013-03-14 2014-09-25 Amgen Inc. AGLYCOSYLATED Fc-CONTAINING POLYPEPTIDES
TWI709572B (en) * 2013-03-14 2020-11-11 美商安美基公司 Interleukin-2 muteins for the expansion of t-regulatory cells
US10829535B2 (en) 2013-03-14 2020-11-10 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
TWI687435B (en) * 2013-03-14 2020-03-11 美商安美基公司 Interleukin-2 muteins for the expansion of t-regulatory cells
US10562950B2 (en) 2013-03-14 2020-02-18 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US11680090B2 (en) 2013-09-24 2023-06-20 Medicenna Therapeutics, Inc. Interleukin-2 fusion proteins and uses thereof
AU2015215001B2 (en) * 2014-02-06 2019-08-15 F. Hoffmann-La Roche Ag Interleukin-2 fusion proteins and uses thereof
CN105980410B (en) * 2014-02-06 2020-02-28 豪夫迈·罗氏有限公司 Interleukin-2 fusion proteins and uses thereof
KR20160103134A (en) * 2014-02-06 2016-08-31 에프. 호프만-라 로슈 아게 Interleukin-2 fusion proteins and uses thereof
KR102042246B1 (en) * 2014-02-06 2019-11-28 에프. 호프만-라 로슈 아게 Interleukin-2 fusion proteins and uses thereof
TWI671318B (en) * 2014-02-06 2019-09-11 赫孚孟拉羅股份公司 Interleukin-2 fusion proteins and uses thereof
CN111423513A (en) * 2014-02-06 2020-07-17 豪夫迈·罗氏有限公司 Interleukin-2 fusion proteins and uses thereof
CN105980410A (en) * 2014-02-06 2016-09-28 豪夫迈·罗氏有限公司 Interleukin-2 fusion proteins and uses thereof
US11098099B2 (en) 2014-02-06 2021-08-24 Hoffmann-La Roche Inc. Interleukin-2 fusion proteins and uses thereof
EA034350B1 (en) * 2014-02-06 2020-01-30 Ф.Хоффманн-Ля Рош Аг Interleukin-2 fusion proteins and uses thereof
EP3508496A1 (en) 2014-02-06 2019-07-10 F. Hoffmann-La Roche AG Interleukin-2 fusion proteins and uses thereof
WO2015118016A1 (en) * 2014-02-06 2015-08-13 F. Hoffmann-La Roche Ag Interleukin-2 fusion proteins and uses thereof
US10654905B2 (en) 2014-04-24 2020-05-19 The Board Of Trustees Of The Leland Stanford Junior University Method of treating graft versus host disease with an interleukin-2 mutein
US11384131B2 (en) 2014-04-24 2022-07-12 The Board Of Trustees Of The Leland Stanford Junior University Superagonists, partial agonists and antagonists of interleukin-2
CN114133456A (en) * 2014-07-21 2022-03-04 德里尼亚公司 Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
US10035836B1 (en) 2014-08-11 2018-07-31 Delinia, Inc. Modified IL-2 variants that selectively activate regulatory T cells
WO2016164937A3 (en) * 2015-04-10 2017-01-12 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US11976103B2 (en) 2015-04-10 2024-05-07 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
WO2016164937A2 (en) 2015-04-10 2016-10-13 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
AU2019264645B2 (en) * 2015-04-10 2021-08-12 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US10851144B2 (en) 2015-04-10 2020-12-01 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
IL283764B1 (en) * 2015-04-10 2023-09-01 Amgen Inc Interleukin-2 muteins for the expansion of t-regulatory cells
US10766938B2 (en) 2016-01-20 2020-09-08 Delinia, Inc. Nucleic acid encoding human IL-2 variant
US10875901B2 (en) 2016-01-20 2020-12-29 Delinia, Inc. Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
US11535657B2 (en) 2016-01-20 2022-12-27 Delinia, Inc. Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
US10774126B2 (en) 2016-01-20 2020-09-15 Delinia, Inc. Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
US10294287B2 (en) 2016-01-20 2019-05-21 Delinia, Inc. Molecules that selectively activate regulatory T cells for the treatment of autoimmune diseases
CN109071623B (en) * 2016-05-04 2022-05-27 美国安进公司 Interleukin-2 muteins for expansion of T regulatory cells
CN109071623A (en) * 2016-05-04 2018-12-21 美国安进公司 For expanding the interleukin 2 mutain of T regulatory cell
EP3808764A1 (en) 2016-05-04 2021-04-21 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US11505591B2 (en) 2016-05-18 2022-11-22 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11077172B2 (en) 2016-11-08 2021-08-03 Delinia, Inc. IL-2 variants for the treatment of psoriasis
US11905320B2 (en) 2016-12-22 2024-02-20 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11708400B2 (en) 2016-12-22 2023-07-25 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11505588B2 (en) 2016-12-22 2022-11-22 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11739133B2 (en) 2016-12-22 2023-08-29 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11987610B2 (en) 2016-12-22 2024-05-21 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11851467B2 (en) 2016-12-22 2023-12-26 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11530248B2 (en) 2016-12-22 2022-12-20 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11851471B2 (en) 2017-01-09 2023-12-26 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11767355B2 (en) 2017-03-15 2023-09-26 Cue Biopharma, Inc. Methods for modulating an immune response
US11993641B2 (en) 2017-03-15 2024-05-28 Cue Biopharma, Inc. Methods for modulating an immune response
US11479595B2 (en) 2017-03-15 2022-10-25 Cue Biopharma, Inc. Methods for modulating an immune response
US10961310B2 (en) 2017-03-15 2021-03-30 Pandion Operations, Inc. Targeted immunotolerance
US11958893B2 (en) 2017-03-15 2024-04-16 Cue Biopharma, Inc. Methods for modulating an immune response
US11466068B2 (en) 2017-05-24 2022-10-11 Pandion Operations, Inc. Targeted immunotolerance
US11542312B2 (en) 2017-06-19 2023-01-03 Medicenna Therapeutics, Inc. IL-2 superagonists in combination with anti-PD-1 antibodies
US11834689B2 (en) 2017-07-11 2023-12-05 The Scripps Research Institute Incorporation of unnatural nucleotides and methods thereof
US10610571B2 (en) 2017-08-03 2020-04-07 Synthorx, Inc. Cytokine conjugates for the treatment of proliferative and infectious diseases
AU2018309166B2 (en) * 2017-08-03 2022-12-08 Synthorx, Inc. Cytokine conjugates for the treatment of proliferative and infectious diseases
US11701407B2 (en) 2017-08-03 2023-07-18 Synthorx, Inc. Cytokine conjugates for the treatment of proliferative and infectious diseases
US11622993B2 (en) 2017-08-03 2023-04-11 Synthorx, Inc. Cytokine conjugates for the treatment of autoimmune diseases
EP3661956A4 (en) * 2017-08-03 2021-04-28 Synthorx, Inc. Cytokine conjugates for the treatment of proliferative and infectious diseases
US11091526B2 (en) 2017-12-06 2021-08-17 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11779632B2 (en) 2017-12-06 2023-10-10 Pandion Operation, Inc. IL-2 muteins and uses thereof
US11965008B2 (en) 2017-12-06 2024-04-23 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11091527B2 (en) 2017-12-06 2021-08-17 Pandion Operations, Inc. IL-2 muteins and uses thereof
US10946068B2 (en) 2017-12-06 2021-03-16 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11945852B2 (en) 2017-12-06 2024-04-02 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11981717B2 (en) 2017-12-19 2024-05-14 Xencor, Inc. Engineered IL-2 Fc fusion proteins
KR20200103681A (en) 2017-12-27 2020-09-02 쿄와 기린 가부시키가이샤 IL-2 variant
AU2018394189B2 (en) * 2017-12-27 2023-12-21 Kyowa Kirin Co., Ltd. IL-2 variant
WO2019131964A1 (en) 2017-12-27 2019-07-04 協和発酵キリン株式会社 Il-2 variant
US11702461B2 (en) 2018-01-09 2023-07-18 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides comprising reduced-affinity immunomodulatory polypeptides
WO2019158764A1 (en) 2018-02-16 2019-08-22 Iltoo Pharma Use of interleukin 2 for treating sjögren's syndrome
WO2020007937A1 (en) 2018-07-03 2020-01-09 Iltoo Pharma Use of interleukin-2 for treating systemic sclerosis
WO2020035482A1 (en) 2018-08-13 2020-02-20 Iltoo Pharma Combination of interleukin-2 with an interleukin 1 inhibitor, conjugates and therapeutic uses thereof
US11077195B2 (en) 2019-02-06 2021-08-03 Synthorx, Inc. IL-2 conjugates and methods of use thereof
US11248046B2 (en) 2019-02-15 2022-02-15 Integral Molecular, Inc. Claudin 6 antibodies and uses thereof
US11254736B2 (en) 2019-02-15 2022-02-22 Integral Molecular, Inc. Antibodies comprising a common light chain and uses thereof
WO2020201095A1 (en) 2019-03-29 2020-10-08 Institut Curie Interleukin-2 variants with modified biological activity
US11739146B2 (en) 2019-05-20 2023-08-29 Pandion Operations, Inc. MAdCAM targeted immunotolerance
EP4385573A2 (en) 2019-08-13 2024-06-19 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
WO2021030602A1 (en) 2019-08-13 2021-02-18 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
US11692020B2 (en) 2019-11-20 2023-07-04 Anwita Biosciences, Inc. Cytokine fusion proteins, and their pharmaceutical compositions and therapeutic applications
WO2021116444A1 (en) 2019-12-12 2021-06-17 Iltoo Pharma Interleukin 2 chimeric constructs
WO2021127262A1 (en) 2019-12-17 2021-06-24 Amgen Inc. Dual interleukin-2 /tnf receptor agonist for use in therapy
US11725034B2 (en) 2019-12-20 2023-08-15 Regeneron Pharmaceuticals, Inc. IL2 agonists and methods of use thereof
US11981715B2 (en) 2020-02-21 2024-05-14 Pandion Operations, Inc. Tissue targeted immunotolerance with a CD39 effector
US11827684B2 (en) 2020-04-22 2023-11-28 Merck Sharp & Dohme Llc Human interleukin-2 conjugates biased for the interleukin-2 receptor beta GAMMAc dimer and conjugated to a nonpeptidic, water-soluble polymer
US11897930B2 (en) 2020-04-28 2024-02-13 Anwita Biosciences, Inc. Interleukin-2 polypeptides and fusion proteins thereof, and their pharmaceutical compositions and therapeutic applications
US11878062B2 (en) 2020-05-12 2024-01-23 Cue Biopharma, Inc. Multimeric T-cell modulatory polypeptides and methods of use thereof
US11879001B2 (en) 2020-06-03 2024-01-23 Ascendis Pharma Oncology Division A/S Conjugate comprising an IL-2 moiety
WO2022013696A3 (en) * 2020-07-14 2022-02-24 Pfizer Inc. Recombinant vaccinia virus
WO2022043493A1 (en) * 2020-08-28 2022-03-03 Ascendis Pharma Oncology Division A/S Glycosylated il-2 proteins and uses thereof
US12029782B2 (en) 2020-09-09 2024-07-09 Cue Biopharma, Inc. MHC class II T-cell modulatory multimeric polypeptides for treating type 1 diabetes mellitus (T1D) and methods of use thereof
US12024559B2 (en) 2020-10-23 2024-07-02 Asher Biotherapeutics, Inc. Fusions with CD8 antigen binding molecules for modulating immune cell function
CN112279906A (en) * 2020-10-30 2021-01-29 浙江新码生物医药有限公司 Human interleukin 2-polyethylene glycol conjugate, preparation method and application thereof
WO2022120224A1 (en) * 2020-12-04 2022-06-09 Visterra, Inc. Methods of using interleukin-2 agents
WO2023057588A1 (en) 2021-10-06 2023-04-13 Iltoo Pharma Interleukin 2 chimeric constructs with targeting specificy to inflamed tissues
WO2024056154A1 (en) 2022-09-12 2024-03-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Interleukin-2 for use in treating autism spectrum disorder
WO2024151515A2 (en) 2023-01-09 2024-07-18 Odyssey Therapeutics, Inc. Anti-tnfr2 antigen-binding proteins and uses thereof
US12049502B2 (en) 2023-11-29 2024-07-30 Integral Molecular, Inc. Antibodies directed to claudin 6, including bispecific formats thereof

Also Published As

Publication number Publication date
US20170313753A1 (en) 2017-11-02
JP2014133756A (en) 2014-07-24
EP2382228B1 (en) 2020-08-26
US20230139534A1 (en) 2023-05-04
US20130195795A1 (en) 2013-08-01
JP2012515778A (en) 2012-07-12
EP2382228A1 (en) 2011-11-02
ES2825173T3 (en) 2021-05-14
AU2010206840A1 (en) 2011-07-21
JP2018087244A (en) 2018-06-07
CA2749539C (en) 2022-07-19
JP2021185191A (en) 2021-12-09
JP2016138140A (en) 2016-08-04
JP2020023550A (en) 2020-02-13
US9732134B2 (en) 2017-08-15
JP5766124B2 (en) 2015-08-19
EP2382228A4 (en) 2013-01-09
CA2749539A1 (en) 2010-07-29
US11560415B2 (en) 2023-01-24
MX2011007647A (en) 2011-09-01
AU2010206840B2 (en) 2015-02-05
US20110274650A1 (en) 2011-11-10

Similar Documents

Publication Publication Date Title
US20230139534A1 (en) Methods of treating graft versus host disease using il-2 muteins
JP7272663B2 (en) Interleukin-2/interleukin-2 receptor alpha fusion proteins and methods of use
KR100607609B1 (en) IL-2 Selective Agonists And Antagonists
JP4502580B2 (en) Use of IL-18 inhibitors for the treatment or prevention of sepsis
Gillespie et al. Interleukin-18: perspectives on the newest interleukin
JP2011132248A (en) THERAPY FOR CHRONIC INFLAMMATORY DEMYELINATING POLYNEUROPATHY USING INTERFERON-beta
KR20240099117A (en) Novel conjugate of immune stimulating IL-2 analog and a preparation method thereof
US20020150555A1 (en) Osteoclastgenic inhibitory agent
KR20230060514A (en) Interleukin-2 muteins and uses thereof
JP2009165483A (en) Ifnar2 mutant, its production and use thereof
EP1648932A2 (en) Il-11 muteins
AU2002309887B2 (en) Use of IL-18 inhibitors for the treatment or prevention of sepsis
AU2002309887A1 (en) Use of IL-18 inhibitors for the treatment or prevention of sepsis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10733814

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010206840

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2749539

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/007647

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2011548074

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2010206840

Country of ref document: AU

Date of ref document: 20100120

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010733814

Country of ref document: EP