WO2024002363A1 - Polypeptides d'il-2 et procédés d'utilisation - Google Patents

Polypeptides d'il-2 et procédés d'utilisation Download PDF

Info

Publication number
WO2024002363A1
WO2024002363A1 PCT/CN2023/105095 CN2023105095W WO2024002363A1 WO 2024002363 A1 WO2024002363 A1 WO 2024002363A1 CN 2023105095 W CN2023105095 W CN 2023105095W WO 2024002363 A1 WO2024002363 A1 WO 2024002363A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
amino acid
acid residue
wildtype
human
Prior art date
Application number
PCT/CN2023/105095
Other languages
English (en)
Inventor
Puwei YUAN
Junli Liu
Fei Zhang
Mingchen CHEN
Chengcheng GUAN
Hang Chen
Fan Liu
Original Assignee
Beijing Neox Biotech Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Neox Biotech Limited filed Critical Beijing Neox Biotech Limited
Publication of WO2024002363A1 publication Critical patent/WO2024002363A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • Interleukin-2 is a member of the IL-2 cytokine subfamily that includes IL-4, IL-7, IL-9, IL-15, IL-21, erythropoietin, and thrombopoietin.
  • IL-2 mediates key functions of the immune system, tolerance, and immunity.
  • IL-2 acts primarily as a T cell growth factor, which is essential for the proliferation and survival of T cells as well as the generation of effector and memory T cells.
  • IL-2 signals through the IL-2 receptor (IL-2R) , a heterotrimeric complex consisting of three chains: alpha (CD25) , beta (CD122) , and gamma (CD132) .
  • IL-2 The binds to the IL-2R ⁇ subunit with low affinity (K D ⁇ 10 -8 M) . However, binding to IL-2R ⁇ alone does not lead to signal transduction. Heterodimerization of the ⁇ and ⁇ subunits of IL-2R is required for signaling in T cells.
  • IL-2 can signalize either via intermediate-affinity dimeric IL-2R ⁇ (K D ⁇ 10 -9 M) or high-affinity trimeric IL-2R ⁇ (K D ⁇ 10 -11 M) .
  • Dimeric IL-2R is expressed by memory CD8+ T cells and natural killer (NK) cells, whereas regulatory T cells (Tregs) and activated T cells express high levels of trimeric IL-2R.
  • IL-2 polypeptides that exhibit reduced binding affinity to IL-2R ⁇ , while retaining significant binding affinity to IL-2R ⁇ relative to wildtype human IL-2.
  • IL-2 polypeptides that exhibit elevated binding affinity to IL-2R ⁇ relative to wildtype IL-2.
  • the IL-2 polypeptides of the disclosure can significantly activate CD8+ T cells and/or NK cells, and meanwhile can reduce or eliminate the activation of Tregs.
  • the IL-2 polypeptides of the disclosure can further exhibit improved thermostability relative to wildtype IL-2.
  • pharmaceutical compositions and kits of IL-2 polypeptides herein effective for treatment of a condition, such as cancer are provided herein.
  • One embodiment provides an isolated polypeptide comprising an IL-2 variant moiety with an amino acid residue mutation at K64 of a sequence of wildtype human IL-2.
  • One embodiment provides an isolated polypeptide comprising an IL-2 variant moiety with amino acid residue mutations at both K64 and P65 of a sequence of wildtype human IL-2.
  • One embodiment provides an isolated polypeptide comprising an IL-2 variant moiety having a sequence of any one of SEQ ID NOs: 3-41.
  • One embodiment provides a pharmaceutical composition comprising an isolated polypeptide described herein; and a pharmaceutically acceptable excipient.
  • One embodiment provides a method of treating a condition in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising an isolated polypeptide described herein; and a pharmaceutically acceptable excipient.
  • nucleic acid molecule comprising a nucleic acid sequence that encodes a polypeptide described herein.
  • the nucleic acid molecule is a DNA molecule.
  • nucleic acid molecule is an RNA molecule.
  • nucleic acid molecule is an mRNA molecule.
  • One embodiment provides a pharmaceutical composition
  • a pharmaceutical composition comprising a nucleic acid molecule encoding a polypeptide described herein; and a pharmaceutically acceptable excipient.
  • One embodiment provides a method of treating a condition in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a nucleic acid molecule encoding for a polypeptide described herein; and a pharmaceutically acceptable excipient.
  • One embodiment provides an expression vector comprising a nucleic acid sequence that encodes the polypeptide described herein.
  • One embodiment provides a host cell comprising expression vector comprising a nucleic acid sequence that encodes the polypeptide described herein.
  • One embodiment provides a method for preparing a polypeptide comprising an IL-2 variant moiety, the method comprising: performing recombinant expression with a nucleic acid sequence that encodes the polypeptide described herein, an expression vector described herein, or a host cell described herein.
  • FIG. 1 illustrates an example recombinant expression plasmid vector of wildtype IL-2.
  • FIG. 2 illustrates sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis results of wildtype IL-2 and three IL-2 mutants of the disclosure.
  • FIG. 3 illustrates size exclusion chromatography (SEC) analysis results of wildtype IL-2 and three IL-2 mutants of the disclosure.
  • Wildtype or “WT” or “wt” or “native” refers to an amino acid sequence that is found in nature, including allelic variations.
  • a wildtype protein or polypeptide has an amino acid sequence that has not been intentionally modified.
  • “Variants” of an amino acid sequence refers to amino acid insertion variants, amino acid addition variants, amino acid deletion variants, and/or amino acid substitution variants.
  • Amino acid insertion variants are characterized by insertion of one or more amino acids in a particular amino acid sequence, e.g., a wildtype IL-2 sequence or a functional variant thereof.
  • Amino acid addition variants include N-and/or C-terminal fusions of one or more amino acids.
  • Amino acid deletion variants are characterized by removal of one or more amino acids from the sequence. The deletions may be in any position of the protein sequence.
  • Amino acid deletion variants include deletion at the N-and/or C-terminus of the protein to produce N-and/or C-terminal truncation variants.
  • Amino acid substitution variants are characterized by removal of one or more amino acids from the sequence, and insertion of one or more another amino acids in place of the original amino acids.
  • IL-2 mutant or “IL-2 mutein” or “IL-2 variant” or “IL-2 variant moiety” herein refers to a polypeptide in which one or more amino acids of the wildtype human IL-2 or a functional variant thereof are mutated, preferably substituted with different amino acids.
  • an IL-2 mutant can be prolonged, truncated, or modified (such as glycosylated, methylated, or PEGylated) .
  • an IL-2 mutant has a sequence that is at least 80%identical to the sequence of wildtype human IL-2 or SEQ ID NO: 1.
  • an IL-2 mutant provided herein comprises a signal peptide of wildtype human IL-2.
  • an IL-2 mutant provided herein does not comprise a signal peptide of wildtype human IL-2.
  • wildtype human IL-2 refers to the 133-amino acid sequence of native human IL-2 (less the signal peptide, consisting of an additional 20 amino acids at the N-terminus) , whose amino acid sequence is described in Fujita, et. al, PNAS USA, 80, 7437-7441 (1983) , with or without an additional N-terminal methionine, which is necessarily included when the protein is expressed as an intracellular fraction in Escherichia coli.
  • wildtype human IL-2 comprises the amino acid sequence of SEQ ID NO: 1.
  • wildtype IL-2 or wildtype human IL-2 refers to an IL-2 polypeptide containing one or more amino acid mutations that do not affect the binding of the polypeptide to the IL-2 receptor, for example, substitution of alanine or serine for cysteine at a position corresponding to residue 125 of wildtype human IL-2 (C125A/S) .
  • CD25 or “ ⁇ subunit of IL-2 receptor” or “IL-2R ⁇ ” refers to any natural CD25 from any vertebrate sources, including mammals such as primates (e.g., human) and rodents (e.g., mouse and rat) , a “full-length” unprocessed CD25 and any processed forms of CD25 derived from cells, and naturally-occurring CD25 variants, such as splice variants or allelic variants.
  • CD25 is human CD25.
  • CD122 or “ ⁇ subunit of IL-2 receptor” or “IL-2R ⁇ ” refers to any natural CD122 from any vertebrate sources, including mammals such as primates (e.g., human) and rodents (e.g., mouse and rat) , a “full-length” unprocessed CD122 and any processed forms of CD122 derived from cells, and naturally-occurring CD122 variants, such as splice variants or allelic variants.
  • CD122 is human CD122.
  • CD132 or “ ⁇ subunit of IL-2 receptor” or “IL-2R ⁇ ” refers to any natural CD132 from any vertebrate sources, including mammals such as primates (e.g., human) and rodents (e.g., mouse and rat) , a “full-length” unprocessed CD132 and any processed forms of CD132 derived from cells, and naturally-occurring CD132 variants, such as splice variants or allelic variants. In certain embodiments, CD132 is human CD132.
  • Natural amino acid refers to an amino acid of the 20 naturally-occurring amino acids in protein. Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate) , basic (lysine, arginine, histidine) , non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan) , and uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are classified as aromatic amino acids.
  • “Unnatural amino acid” refers to an amino acid other than the 20 naturally-occurring amino acids in protein.
  • amino acid residue mutation refers to substitution, deletion, insertion, modification, or any combination thereof of one or more amino acid residues for obtaining a final construct, such that the final construct possesses desired characteristics, e.g., altered binding affinity or enhanced stability.
  • an amino acid residue mutation is an amino acid residue substitution.
  • amino acid residues can be substituted in which one amino acid residue is replaced with another amino acid residue having different structure and/or chemical property.
  • Percentage identity refers to a percentage of identical amino acid residues between two sequences being compared after an optimal alignment of sequences.
  • An optimal alignment of sequences may be produced manually or by means of computer programs which use a sequence alignment algorithm (e.g., ClustalW, T-coffee, COBALT, BestFit, FASTA, BLASTP, BLASTN, and TFastA) .
  • Percentage identity can be calculated by determining the number of identical positions between the two sequences being compared, dividing this number by the number of positions compared, and multiplying the result obtained by 100 so as to obtain the percentage identity between the two sequences.
  • subject or “patient” encompasses mammals.
  • mammals include, but are not limited to, any member of the Mammalia class: humans, non-human primates, such as chimpanzees, and other apes and monkey species; farm animals, such as cattle, horses, sheep, goats, swine; domestic animals, such as rabbits, dogs, and cats; laboratory animals, including rodents, such as rats, mice and guinea pigs, and the like.
  • the mammal is a human.
  • treatment or “treating” are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient is still afflicted with the underlying disorder.
  • the compositions are, in some embodiments, administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease has not been made.
  • IL-2 is a 15-kDa four- ⁇ -helix-bundle cytokine that contains a single disulfide bond, which is essential for its biological activity.
  • IL-2 is a member of a cytokine family that includes IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, which all share the common cytokine receptor ⁇ chain.
  • IL-2 is responsible for regulating the immune activity of white blood cells (usually lymphocytes) in the immune system.
  • IL-2 acts by binding to IL-2 receptors on the surface of immune cells and is a cytokine necessary for effector T cell expansion, survival, and function.
  • IL-2 mediates key functions of the immune system, tolerance, and immunity.
  • IL-2 acts primarily as a T cell growth factor, which is essential for the proliferation and survival of T cells as well as the generation of effector and memory T cells.
  • IL-2 signals through the IL-2 receptor (IL-2R) , a heterotrimeric complex consisting of three subunits: alpha ( ⁇ ; CD25) , beta ( ⁇ ; CD122) , and gamma ( ⁇ ; CD132) .
  • the IL-2R ⁇ subunit binds IL-2 with low affinity (K D ⁇ 10 -8 M) .
  • K D ⁇ 10 -8 M low affinity
  • Heterodimerization of the ⁇ and ⁇ subunits of IL-2R is essential for signaling in T cells.
  • IL-2 can signalize either via intermediate-affinity dimeric IL-2R ⁇ (K D ⁇ 10 -9 M) or high-affinity trimeric IL-2R ⁇ (K D ⁇ 10 -11 M) .
  • Dimeric IL-2R is expressed by memory CD8+ T cells and NK cells, whereas regulatory T cells and activated T cells express high levels of trimeric IL-2R ⁇ .
  • IL-2 polypeptides described herein exhibit a reduced affinity for IL-2R ⁇ relative to the affinity for IL-2R ⁇ , the receptor complex that is predominant on and memory T cells and NK cells.
  • treatment using the IL-2 polypeptides described herein can promote activation of T cells such as CD8+ T cells over Tregs in the tumor microenvironment to elicit anti-tumor efficacy.
  • IL-2 receptors are expressed mainly by lymphoid cells. In the absence of stimulation, IL-2R ⁇ is mainly expressed on Treg cells and not expressed on T cells, but is potently induced on T cell activation. IL-2R ⁇ and ⁇ are constitutively expressed on some resting T cells, especially NK cells, CD8+ T cells, B cells, macrophages, monocytes, and DCs. Like IL-2R ⁇ , IL-2R ⁇ can be further induced on stimulation of these cells, either by an antigen or by IL-2.
  • IL-2 pleiotropic effects of IL-2 can be attributed to IL-2 signaling transduction via three different signaling pathways: JAK-STAT, PI3K/Akt/mTOR, and MAPK/ERK pathways.
  • JAK-STAT IL-2 signaling transduction via three different signaling pathways: JAK-STAT, PI3K/Akt/mTOR, and MAPK/ERK pathways.
  • IL-2R cytoplasmatic domains of the ⁇ and ⁇ subunits heterodimerize. This heterodimerization leads to the activation of Janus kinases, JAK1 and JAK3, which subsequently phosphorylate T338 on the ⁇ -subunit.
  • This phosphorylation leads to recruitment of STAT transcription factors, predominantly STAT5, which dimerize and migrate to the cell nucleus where the transcription factors bind to DNA.
  • IL-2 can prevent autoimmune diseases by promoting the differentiation of certain immature T cells into regulatory T cells, which suppress other T cells that would otherwise be primed to attack normal healthy cells in the body.
  • IL-2 can enhance activation-induced cell death.
  • IL-2 can also promote the differentiation of T cells into effector T cells and into memory T cells when the initial T cell is also stimulated by an antigen, thus helping the body fight off infections.
  • IL-2 stimulates naive CD4+ T cell differentiation into Th1 and Th2 lymphocytes while impeding differentiation into Th17 and follicular Th lymphocytes.
  • IL-2 stimulates proliferation and enhances function of T cells, NK cells, and B cells.
  • IL-2-activated B cells can generate secretory IgM rather than membrane-associated IgM.
  • Macrophages can gain maturity and elaborate transforming growth factor- ⁇ (TGF- ⁇ ) when stimulated with IL-2.
  • TGF- ⁇ transforming growth factor- ⁇
  • TGF- ⁇ transforming growth factor- ⁇
  • IL-2 toxicity hinders widespread implementation.
  • IL-2 toxicity includes, for example, anemia, thrombocytopenia, endothelial cell damage, and renal damage. Modulating IL-2 activity can be an effective treatment strategy for immunodeficiency, autoimmune disorders, infectious diseases, allergic conditions, and malignancies.
  • the polypeptide preferentially activates effector T cells and NK cells over regulatory T cells (e.g., CD4+CD25+ FoxP3+ T cells) .
  • regulatory T cells e.g., CD4+CD25+ FoxP3+ T cells
  • IL-2 can activate Tregs expressing IL-2R ⁇ trimer and non-targeted cells, such as eosinophils, which can inhibit the tumor suppressor activity of T cells, causing significant side effects.
  • CTLs cytotoxic T lymphocytes
  • IL-2 polypeptides comprising an IL-2 variant moiety having reduced affinity to IL-2R ⁇ relative to the affinity to IL-2R ⁇ .
  • An IL-2 mutein herein includes one or more amino acid substitutions, e.g., at positions K35, R38, F42, K43, F44, E61, E62, K64, P65, E68, L72, Y107, and/or C125, of wildtype human IL-2.
  • the affinity of IL-2 mutants of the present disclosure to IL-2R ⁇ is significantly reduced, e.g., such that these IL-2 mutants can reduce or block IL-2 activation of Tregs. Further, IL-2 mutants of the present disclosure can also significantly activate CD8+ T cells and/or NK cells.
  • the thermal stability of the IL-2 mutants of the present disclosure is significantly improved compared with the wildtype IL-2. The druggability of the IL-2 mutants with high thermal stability is significantly improved compared with the wildtype IL-2.
  • IL-2 polypeptides provided herein exhibit reduced binding affinity to IL-2 ⁇ and/or enhanced binding affinity to IL-2R ⁇ relative to a wildtype human IL-2.
  • the IL-2 polypeptides can include an IL-2 variant moiety having one or more amino acid residue mutations of a sequence of wildtype human IL-2.
  • IL-2 polypeptides having modulated binding affinity to IL-2 ⁇ are generated by making amino acid substitutions to the wildtype IL-2 protein at one or more positions on the binding interface between wildtype IL-2 and the ⁇ -subunit of the IL-2R.
  • IL-2 polypeptides with modulated binding affinity to IL-2 ⁇ are generated by making amino acid substitutions to the wildtype IL-2 protein at one or more positions on the binding interface between wildtype IL-2 and the ⁇ -subunit of IL-2R.
  • IL-2 polypeptides provided herein are generated based on the sequence of wildtype human IL-2:
  • wildtype human IL-2 comprises the amino acid sequence of SEQ ID NO: 1.
  • wildtype human IL-2 consists of the amino acid sequence of SEQ ID NO: 1.
  • an IL-2 variant moiety comprises an amino acid sequence that is at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, 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%, or at least 99%identical to SEQ ID NO: 1.
  • an IL-2 polypeptide comprises one, two, three, four, five, or more amino acid residue mutations at positions selected from the group consisting of K35, R38, F42, K43, F44, E61, E62, K64, P65, E68, L72, Y107, and C125 of wildtype human IL-2.
  • an IL-2 polypeptide comprises one, two, three, four, or more amino acid residue mutations at positions selected from the group consisting of K35, R38, F42, K43, F44, E61, E62, K64, P65, E68, L72, and Y107 of wildtype human IL-2.
  • an IL-2 polypeptide comprises an amino acid residue mutation at K64 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at K64 is K64P or K64E.
  • an IL-2 polypeptide comprises an amino acid residue mutation at P65 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at P65 is P65E, P65D, P65R, P65W, or P65K.
  • an IL-2 polypeptide further comprises one or more amino acid residue mutations at positions selected from the group consisting of K35, R38, K43, E61, E62, L72, and Y107 of wildtype human IL-2.
  • an IL-2 polypeptide further comprises one or more amino acid residue mutations at positions selected from the group consisting of K35, R38, K43, and Y107 of wildtype human IL-2.
  • an IL-2 polypeptide further comprises one or more amino acid residue mutations at positions selected from the group consisting of F42, F44, and E62 of wildtype human IL-2.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at R38 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at R38 is R38W or R38Q.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at K43 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at K43 is K43Q, K43R, or K43G.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at E61 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at E61 is E61T.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at L72 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at L72 is L72T.
  • an IL-2 polypeptide further comprises one, two, three, more amino acid residue mutations selected from the group consisting of F42, K43, F44, and E62 of wildtype human IL-2.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at F42 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at F42 is F42K, F42R, F42E, or F42P.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at K43 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at K43 is K43R, K43G, or K43Q.
  • an IL-2 polypeptide further comprises an amino acid residue mutation at E62 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at E62 is E62S, E62R, E62N, E62T, or E62A.
  • an IL-2 polypeptide comprises one, two, or more amino acid residue mutations at positions selected from the group consisting of K35, R38, and Y107 of wildtype human IL-2.
  • the amino acid residue mutation at K35 is K35Y, K35D, or K35Q.
  • the amino acid residue mutation at K35 is K35Y or K35Q.
  • the amino acid residue mutation at Y107 is Y107K or Y107R.
  • the amino acid residue mutation at R38 is R38Q or R38W.
  • an IL-2 polypeptide comprises amino acid residue mutations selected from the group consisting of: K35Y; K35Q and Y107K; and R38Q and Y107R.
  • an IL-2 polypeptide comprises amino acid residue mutations selected from the group consisting of:
  • an IL-2 polypeptide comprises amino acid residue mutations selected from the group consisting of:
  • an IL-2 polypeptide comprises amino acid residue mutations selected from the group consisting of:
  • an IL-2 polypeptide comprises amino acid residue mutations selected from the group consisting of:
  • an IL-2 polypeptide further comprises an amino acid residue mutation at C125 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at C125 is C125A or C125S.
  • an IL-2 polypeptide further comprises a sequence of wildtype human IL-2 having one, two, or more amino acid residues replaced with a cysteine.
  • the amino acid residues replaced by cysteine are not at positions selected from K64 and P65 corresponding to SEQ ID NO: 1.
  • the amino acid residues replaced by cysteine are not at positions selected K35, R38, K43, Y107, K64, and P65 corresponding to SEQ ID NO: 1.
  • at least two of the two or more cysteine residues form a disulfide linkage.
  • an IL-2 polypeptide comprises an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to any one of SEQ ID NOs: 3-41. In some embodiments, an IL-2 polypeptide comprises a sequence of any one of SEQ ID NOs: 3-41.
  • an amino acid residue described herein is mutated to a natural amino acid, e.g., proline, lysine, cysteine, histidine, arginine, aspartic acid, asparagine, glutamic acid, glutamine, methionine, serine, threonine, tryptophan, or tyrosine.
  • an amino acid residue described herein is mutated prior to binding to (or reacting with) a conjugating moiety.
  • an amino acid residue described herein (e.g., of IL-2) is mutated to an unnatural amino acid.
  • a polypeptide herein comprises an unnatural amino acid, wherein the cytokine is conjugated to the protein, wherein the point of attachment is not the unnatural amino acid.
  • an amino acid residue described herein (e.g., of IL-2) is mutated prior to binding to (or reacting with) a conjugating moiety.
  • mutation to an unnatural amino acid reduces the likelihood of or minimizes a self-antigen response of the immune system.
  • Non-limiting examples of unnatural amino acid include p-acetyl-L-phenylalanine, p-iodo-L-phenylalanine, p-methoxyphenylalanine, O-methyl-L-tyrosine, p-propargyloxyphenylalanine, p-propargyl-phenylalanine, L-3- (2-naphthyl) alanine, 3-methyl-phenylalanine, O-4-allyl-L-tyrosine, 4-propyl-L-tyrosine, tri-O-acetyl-GlcNAcp-serine, L-Dopa, fluorinated phenylalanine, isopropyl-L-phenylalanine, p-azido-L-phenylalanine, p-acyl-L-phenylalanine, p-benzoyl-L-phenylalanine, p-boronophenylalanine,
  • the unnatural amino acid comprises a selective reactive group, or a reactive group for site-selective labeling of a target polypeptide.
  • the chemistry is a biorthogonal reaction (e.g., biocompatible and selective reactions) .
  • the chemistry is a Cu (I) -catalyzed or “copper-free” alkyne-azide triazole-forming reaction, the Staudinger ligation, inverse-electron-demand Diels-Alder (IEDDA) reaction, “photo-click” chemistry, or a metal-mediated process such as olefin metathesis and Suzuki-Miyaura or Sonogashira cross-coupling.
  • the unnatural amino acid comprises a photoreactive group, which crosslinks, upon irradiation with, e.g., UV.
  • the unnatural amino acid comprises a photo-caged amino acid.
  • the unnatural amino acid is a para-substituted, meta-substituted, or an ortho-substituted amino acid derivative.
  • the unnatural amino acid comprises N6-azidoethoxy-L-lysine (AzK) , N6-propargylethoxy-L-lysine (PraK) , BCN-L-lysine, norbornene lysine, TCO-lysine, methyltetrazine lysine, allyloxycarbonyllysine, m-acetylphenylalanine, 2-amino-8-oxononanoic acid, 2-amino-8-oxooctanoic acid, p-acetyl-L-phenylalanine, p-azidomethyl-L-phenylalanine (pAMF) , p-iodo-L-phenylalanine, p-methoxyphenylalanine, p-propargyloxyphenylalanine, p-propargyl-phenylalanine, L-3- (2-naphthy
  • the unnatural amino acid is 3-aminotyrosine, 3-nitrotyrosine, 3, 4-dihydroxy-phenylalanine, or 3- iodotyrosine.
  • the unnatural amino acid is phenylselenocysteine.
  • the unnatural amino acid is a benzophenone, ketone, iodide, methoxy, acetyl, benzoyl, or azide-containing phenylalanine derivative.
  • the unnatural amino acid is a benzophenone, ketone, iodide, methoxy, acetyl, benzoyl, or azide-containing lysine derivative.
  • an IL-2 polypeptide comprises an amino acid sequence that is heterologous to wildtype human IL-2 or a functional variant thereof.
  • the amino acid sequence can be a conjugating moiety, a half-life extension moiety, an albumin, an immunoglobulin or a fragment thereof, a transferrin, or a PEG.
  • the amino acid sequence can be linked to an N-terminus of an IL-2 variant moiety. In some embodiments, the amino acid sequence can be linked to a C-terminus of an IL-2 variant moiety.
  • a nucleic acid molecule can encode a polypeptide and/or IL-2 variant moiety described herein.
  • the nucleic acid molecule can be codon-optimized to encode a polypeptide and/or IL-2 variant moiety described herein.
  • Such a nucleic acid molecule can be inserted in an expression vector, which can then be transformed or incorporated into a host cell.
  • the expression vector can be a plasmid.
  • the host cell comprising the expression vector can be used to express an IL-2 mutant described herein.
  • the host cell can be E. coli or other suitable bacterial cell.
  • the preparation of the present invention can be performed using standard procedures known to one skilled in the art, for example, in Michael R. Green and Joseph Sambrook, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (2012) ; Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (1986) ; Current Protocols in Molecular Biology (CPMB) (Fred M. Ausubel, et al. ed., John Wiley and Sons, Inc. ) , Current Protocols in Immunology (CPI) (John E. Coligan, et. al., ed. John Wiley and Sons, Inc.
  • IL-2 polypeptides provided herein include an IL-2 variant moiety and a conjugating moiety that is bound to one or more cytokines (e.g., interleukins, IFNs, or TNFs) .
  • a conjugating moiety is a molecule that modulates the interaction of a cytokine with its receptor, e.g., IL-2 with IL-2R subunits.
  • the conjugating moiety is a molecule that when bound to the cytokine, enables the cytokine conjugate to modulate an immune response.
  • the conjugating moiety is bound to the cytokine through a covalent bond.
  • a cytokine described herein is attached to a conjugating moiety with a triazole group. In some embodiments, a cytokine described herein is attached to a conjugating moiety with a dihydropyridazine or pyridazine group. In some embodiments, the conjugating moiety comprises a water-soluble polymer. In other embodiments, the conjugating moiety comprises a protein or a binding fragment thereof. In additional embodiments, the conjugating moiety comprises a peptide. In additional embodiments, the conjugating moiety comprises a nucleic acid. In additional embodiments, the conjugating moiety comprises a small molecule.
  • the conjugating moiety comprises a bioconjugate (e.g., a TLR agonist such as a TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, or TLR9 agonist; or a synthetic ligand such as Pam3Cys, CFA, MALP2, Pam2Cys, FSL-1, Hib-OMPC, Poly I: C, Poly A: U, AGP, MPLA, RC-529, MDF2p, CFA, or flagellin) .
  • a conjugating moiety increases serum half-life and/or improves stability in vivo.
  • the conjugating moiety reduces cytokine interaction with one or more cytokine receptor domains or subunits. In some embodiments, the conjugating moiety blocks cytokine interaction with one or more cytokine domains or subunits with its cognate receptor (s) . In some embodiments, cytokine conjugates described herein comprise multiple conjugating moieties. In some embodiments, a conjugating moiety is attached to an unnatural or natural amino acid in the cytokine peptide. In some embodiments, an IL-2 peptide comprises a conjugating moiety attached to a natural amino acid in the IL-2 cytokine peptide.
  • a cytokine conjugate is attached to an unnatural amino acid in the cytokine peptide.
  • a conjugating moiety is attached to the N-or C-terminus amino acid of the cytokine peptide.
  • Various combinations sites are disclosed herein, for example, a first conjugating moiety is attached to an amino acid in the cytokine peptide, and a second conjugating moiety is attached to the N-or C-terminus of the cytokine peptide.
  • a single conjugating moiety is attached to multiple residues of the cytokine peptide (e.g., a staple) .
  • a conjugating moiety is attached to both the N-and C-terminus amino acids of the cytokine peptide.
  • a conjugating moiety is a masking moiety.
  • a masking moiety can block, occlude, inhibit, decrease, or otherwise reduce (i.e., masks) the activity or binding of the cytokine to its cognate receptor or protein.
  • an IL-2 cytokine can be activatable by a protease at a target site, such as in a tumor microenvironment, by inclusion of a proteolytically cleavable linker.
  • a proteolytically cleavable linker links the cytokine to the masking moiety. Upon proteolytic cleavage of the cleavable linker at the target site, the cytokine can become activated, thereby rendering the cytokine capable of binding to its cognate receptor or protein with increased affinity.
  • a masking moiety can include a cleavable peptide and/or linker.
  • the cleavable linker comprises a cleavable peptide.
  • a cleavable peptide is a polypeptide that includes a cleavage site, such as a protease cleavage site.
  • the cleavage site is a site recognizable for cleavage of a portion of the cleavable peptide of a cleavable linker described herein.
  • the cleavable peptide comprises more than one cleavage site.
  • the cleavage site is an amino acid sequence that is recognized and cleaved by a cleaving agent.
  • Exemplary cleaving agents include proteins, enzymes, DNAzymes, RNAzymes, metals, acids, and bases.
  • the protease cleavage site is a matrix metalloprotease (MMP) cleavage site, a disintegrin and metalloprotease domain-containing (ADAM) metalloprotease cleavage site, a prostate specific antigen (PSA) protease cleavage site, a urokinase-type plasminogen activator (uPA) protease cleavage site, a membrane type serine protease 1 (MT-SP1) protease cleavage site, a matriptase protease cleavage site (ST14) or a legumain protease cleavage site.
  • MMP matrix metalloprotease
  • ADAM disintegrin and metalloprotease domain-containing
  • PSA prostate specific antigen
  • uPA urokinase-type plasminogen activator
  • ST14 matriptase protease cleavage site
  • ST14 legumain protease cle
  • the matrix metalloprotease (MMP) cleavage site is a MMP9 cleavage site, a MMP13 cleavage site or a MMP2 cleavage site.
  • the disintegrin and metalloprotease domain-containing (ADAM) metalloprotease cleavage site is an ADAM9 metalloprotease cleavage site, an ADAM10 metalloprotease cleavage site or an ADAM17 metalloprotease cleavage site.
  • Protease cleavage sites can be designated by a specific amino acid sequence.
  • a conjugating moiety is a half-life extension domain that increases serum half-life and/or improves stability of an IL-2 polypeptide.
  • the half-life extension domain can be fused to the N-or C-terminal of the cytokine herein.
  • a proteolytically cleavable linker links the cytokine to the masking moiety, links the cytokine to a half-life extension domain, and/or links the masking moiety to a half-life extension domain.
  • the half-life extension domain is an albumin polypeptide or a functional fragment thereof.
  • Albumin is a natural carrier protein that has an extended serum half-life of approximately three weeks due to its size and its susceptibility to FcRn-mediated recycling, which reduces the likelihood of intracellular degradation.
  • linking an IL-2 polypeptide to albumin can significantly extend the serum half-life of the IL-2 polypeptide.
  • the masked or unmasked cytokine comprises a half-life extension domain that comprises an albumin polypeptide, or a fragment or a variant thereof.
  • the albumin polypeptide is mouse serum albumin. In some embodiments, the albumin polypeptide is human serum albumin.
  • the masked or unmasked cytokine comprises a half-life extension domain that comprises an immunoglobulin Fc domain, or a fragment or a variant thereof.
  • the immunoglobulin Fc domain is mouse immunoglobulin Fc domain.
  • the immunoglobulin Fc domain is human immunoglobulin Fc domain.
  • the human immunoglobulin Fc domain is IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM.
  • the albumin polypeptide or Fc domain is linked to a masking moiety.
  • a masking moiety is linked to the N-or C-terminus of the albumin polypeptide or Fc domain.
  • the albumin polypeptide or Fc domain is linked to a masking moiety via a linker.
  • a linker is linked to the amino-terminus or the carboxy-terminus of the albumin polypeptide or Fc domain.
  • an N-or C-terminal spacer domain of the linker is linked to the N-or C-terminus of the albumin polypeptide or Fc domain.
  • a cleavable peptide of the linker is linked to the N-or C-terminus of the albumin polypeptide or Fc domain.
  • the albumin polypeptide or Fc domain is linked to a cytokine or functional fragment thereof (e.g., IL-2 or a mutein thereof) .
  • a cytokine or functional fragment thereof is linked to the N-or C-terminus of the albumin polypeptide or Fc domain.
  • the albumin polypeptide or Fc domain is linked to a cytokine or functional fragment thereof via a linker.
  • a linker is linked to the amino-terminus or the carboxy-terminus of the albumin polypeptide or Fc domain.
  • a conjugating moiety is a water-soluble polymer.
  • the water-soluble polymer is a non-peptidic, non-toxic, and biocompatible.
  • a substance can be considered biocompatible if the beneficial effects associated with use of the substance alone or with another substance (e.g., an active agent such as a cytokine moiety) in connection with living tissues (e.g., administration to a subject) outweighs any deleterious effects as evaluated by a clinician, e.g., a physician, a toxicologist, or a clinical development specialist.
  • a water-soluble polymer is non-immunogenic.
  • a substance can be considered non-immunogenic if the intended use of the substance in vivo does not produce an undesired immune response (e.g., the formation of antibodies) , or if an immune response is produced, that such a response is not deemed clinically significant or important as evaluated by a clinician, e.g., a physician, a toxicologist, or a clinical development specialist.
  • the weight-average molecular weight of a water-soluble polymer is from about 100 Daltons (Da) to about 150,000 Da.
  • Weight-average molecular weight ranges include from about 5,000 Da to about 100,000 Da, from about 6,000 Da to about 90,000 Da, from about 10,000 Da to about 85,000 Da, from about 10,000 Da to about 85,000 Da, from about 20,000 Da to about 85,000 Da, from about 53,000 Da to about 85,000 Da, from about 25,000 Da to about 120,000 Da, from about 29,000 Da to about 120,000 Da, from about 35,000 Da to about 120,000 Da, and from about 40,000 Da to about 120,000 Da.
  • Non-limiting examples of weight-average molecular weights for a water-soluble polymer include about 100 Da, about 200 Da, about 300 Da, about 400 Da, about 500 Da, about 600 Da, about 700 Da, about 750 Da, about 800 Da, about 900 Da, about 1,000 Da, about 1,500 Da, about 2,000 Da, about 2,200 Da, about 2,500 Da, about 3,000 Da, about 4,000 Da, about 4,400 Da, about 4,500 Da, about 5,000 Da, about 5,500 Da, about 6,000 Da, about 7,000 Da, about 7,500 Da, about 8,000 Da, about 9,000 Da, about 10,000 Da, about 11,000 Da, about 12,000 Da, about 13,000 Da, about 14,000 Da, about 15,000 Da, about 20,000 Da, about 22,500 Da, about 25,000 Da, about 30,000 Da, about 35,000 Da, about 40,000 Da, about 45,000 Da, about 50,000 Da, about 55,000 Da, about 60,000 Da, about 65,000 Da, about 70,000 Da, and about 75,000 Da.
  • Branched versions of the water-soluble polymer e.g., a branched 40,000 Da water-soluble polymer comprised of two 20,000 Da polymers
  • the conjugate does not have any PEG moieties attached, either directly or indirectly, with a PEG having a weight average molecular weight of less than about 6,000 Da.
  • PEGs can comprise a number of (OCH2CH2) monomers or (CH2CH2O) monomers.
  • the number of repeating units can be identified by the subscript “n” in “ (OCH2CH2) n. ”
  • the value of “n” typically can fall within one or more of the following ranges: from 2 to about 3400, from about 100 to about 2300, from about 100 to about 2270, from about 136 to about 2050, from about 225 to about 1930, from about 450 to about 1930, from about 1200 to about 1930, from about 568 to about 2727, from about 660 to about 2730, from about 795 to about 2730, from about 795 to about 2730, from about 909 to about 2730, and from about 1,200 to about 1, 900.
  • the number of repeating units (i.e., “n” ) can be determined by dividing the total weight-average molecular weight of the polymer by the molecular weight of the repeating monomer.
  • the water-soluble polymer is an end-capped polymer, i.e., having at least one terminus capped with a relatively inert group, such as a lower C1-C6 alkoxy group, or a hydroxyl group.
  • the water-soluble polymer can be a methoxy-PEG (mPEG) , which is a linear form of PEG in which one terminus of the PEG is a methoxy (-OCH3) group, while the other terminus is a hydroxyl or other functional group that can be optionally chemically modified.
  • mPEG methoxy-PEG
  • Non-limiting examples of water-soluble polymers include linear or branched discrete PEG (dPEG) ; linear, branched, or forked PEGs; and Y-shaped PEG derivatives.
  • an IL-2 polypeptide described herein is conjugated to a water-soluble polymer selected from poly (alkylene glycols) , such as polyethylene glycol (PEG) , poly (propylene glycol) (PPG) , copolymers of ethylene glycol and propylene glycol and the like, poly (oxyethylated polyol) , poly (olefinic alcohol) , poly (vinylpyrrolidone) , poly (hydroxyalkylmethacrylamide) , poly (hydroxyalkylmethacrylate) , poly (saccharides) , poly (a- hydroxy acid) , poly (vinyl alcohol) (PVA) , polyacrylamide (PAAm) , poly (N- (2-hydroxypropyl) methacrylamide) (PHPMA) , polydimethylacrylamide (PDAAm) , polyphosphazene, polyoxazolines (POZ) , poly (N-acryloylmorpholine)
  • a water-soluble polymer comprises a polyglycerol (PG) , e.g., a HPG, a LPG, a midfunctional PG, a linear-block-hyperbranched PG, or a side-chain functional PG.
  • PG polyglycerol
  • the polyglycerol is a hyperbranched PG (HPG) .
  • a water-soluble polymer is a degradable synthetic PEG alternative.
  • degradable synthetic PEG alternatives include poly [oligo (ethylene glycol) methyl methacrylate] (POEGMA) ; backbone modified PEG derivatives generated by polymerization of telechelic, or di-end-functionalized PEG-based macromonomers; PEG derivatives comprising comonomers comprising degradable linkage such as poly [ (ethylene oxide) -co- (methylene ethylene oxide) ] [P (EO-co-MEO) ] , cyclic ketene acetals such as 5, 6-benzo-2-methylene-1, 3-dioxepane (BMDO) , 2-methylene-1, 3-dioxepane (MDO) , and 2-methylene-4-phenyl-1, 3-dioxolane (MPDL) copolymerized with OEGMA, or poly- (s-caprolactone) -graft-poly (ethylene oxide), or poly- (s-
  • a water-soluble polymer comprises a poly (zwitterions) .
  • poly (zwitterions) include poly (sulfobetaine methacrylate) (PSBMA) , poly (carboxybetaine methacrylate) (PCBMA) , and poly (2-methyacryloyloxyethyl phosphorylcholine) (PMPC) .
  • a water-soluble polymer comprises a polycarbonate.
  • polycarbonates include pentafluorophenyl 5-methyl-2-oxo-1, 3-dioxane-5-carboxylate (MTC-OC6F5) .
  • a water-soluble polymer comprises a polymer hybrid, such as for example, a polycarbonate/PEG polymer hybrid, a peptide/protein-polymer conjugate, or a hydroxyl-containing and/or zwitterionic derivatized polymer (e.g., a hydroxyl-containing and/or zwitterionic derivatized PEG polymer) .
  • a polymer hybrid such as for example, a polycarbonate/PEG polymer hybrid, a peptide/protein-polymer conjugate, or a hydroxyl-containing and/or zwitterionic derivatized polymer (e.g., a hydroxyl-containing and/or zwitterionic derivatized PEG polymer) .
  • a water-soluble polymer comprises a polysaccharide.
  • polysaccharides include dextran, polysialic acid (PSA) , hyaluronic acid (HA) , amylose, heparin, heparan sulfate (HS) , dextrin, or hydroxyethyl starch (HES) .
  • a water-soluble polymer comprises a glycan.
  • glycans include N-linked glycans, O-linked glycans, glycolipids, O-GlcNAc, and glycosaminoglycans.
  • a water-soluble polymer comprises a polyoxazoline polymer.
  • a polyoxazoline polymer is a linear synthetic polymer, and similar to PEG, comprises a low polydispersity.
  • a polyoxazoline polymer is a polydispersed polyoxazoline polymer, characterized with an average molecule weight.
  • the average molecule weight of a polyoxazoline polymer includes, for example, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 10,000, 12,000, 20,000, 35,000, 40,000, 50,000, 60,000, 100,000, 200,000, 300,000, 400,000, or 500,000 Da.
  • a polyoxazoline polymer comprises poly (2-methyl 2-oxazoline) (PMOZ) , poly (2-ethyl 2-oxazoline) (PEOZ) , or poly (2-propyl 2-oxazoline) (PPOZ) .
  • a cytokine e.g., an interleukin, IFN, or TNF
  • a cytokine e.g., an interleukin, IFN, or TNF
  • a water-soluble polymer is a polyacrylic acid polymer.
  • a water-soluble polymer comprises polyamine.
  • Polyamine is an organic polymer comprising two or more primary amino groups.
  • a polyamine includes a branched polyamine, a linear polyamine, or cyclic polyamine.
  • a polyamine is a low-molecular-weight linear polyamine.
  • Exemplary polyamines include putrescine, cadaverine, spermidine, spermine, ethylene diamine, 1, 3-diaminopropane, hexamethylenediamine, tetraethylmethylenediamine, and piperazine.
  • a conjugating moiety is a lipid.
  • the lipid can be a fatty acid, e.g., a saturated fatty acid or an unsaturated fatty acid.
  • fatty acids can have from 6 to 26 carbon atoms, from 6 to 24 carbon atoms, from 6 to 22 carbon atoms, from 6 to 20 carbon atoms, from 6 to 18 carbon atoms, from 20 to 26 carbon atoms, from 12 to 26 carbon atoms, from 12 to 24 carbon atoms, from 12 to 22 carbon atoms, from 12 to 20 carbon atoms, or from 12 to 18 carbon atoms in length.
  • the fatty acid has 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26 carbon atoms in length.
  • Non-limiting examples of fatty acids include caproic acid (hexanoic acid) , enanthic acid (heptanoic acid) , caprylic acid (octanoic acid) , pelargonic acid (nonanoic acid) , capric acid (decanoic acid) , undecylic acid (undecanoic acid) , lauric acid (dodecanoic acid) , tridecylic acid (tridecanoic acid) , myristic acid (tetradecanoic acid) , pentadecylic acid (pentadecanoic acid) , palmitic acid (hexadecanoic acid) , margaric acid (heptadecanoic acid) , stearic acid (octadecanoic acid) , nonadecyl
  • a conjugating moiety described herein is a protein or a binding fragment thereof.
  • proteins include albumin, transferrin, or transthyretin.
  • the protein or a binding fragment thereof comprises an antibody or a binding fragments thereof.
  • the conjugating moiety is albumin or a functional fragment thereof.
  • Albumin is a family of water-soluble globular proteins. Albumin is commonly found in blood plasma, comprising about 55-60%of all plasma proteins.
  • Human serum albumin (HSA) is a 585 amino acid polypeptide comprising three domains: domain I (amino acid residues 1-195) , domain II (amino acid residues 196-383) , and domain III (amino acid residues 384-585) .
  • Each domain further includes a binding site, which can interact either reversibly or irreversibly with endogenous ligands such as fatty acids, bilirubin, or hemin, or exogenous compounds, such as heterocyclic or aromatic compounds.
  • the conjugating moiety is transferrin.
  • Transferrin is a 679 amino acid polypeptide that is about 80 kDa in size and comprises two Fe 3+ binding sites with one at the N-terminal domain and the other at the C-terminal domain. Human transferrin has a half-life of about 7-12 days.
  • the conjugating moiety is transthyretin (TTR) .
  • TTR transthyretin
  • Transthyretin is a transport protein located in the serum and cerebrospinal fluid which transports the thyroid hormone thyroxine (T4) and retinol-binding protein bound to retinol.
  • a conjugating moiety is an antibody or a binding fragment thereof.
  • the antibody or a binding fragment thereof can be a humanized antibody or a binding fragment thereof, murine antibody or a binding fragment thereof, chimeric antibody or a binding fragment thereof, monoclonal antibody or a binding fragment thereof, monovalent Fab', divalent Fab2, F (ab) '3 fragments, single-chain variable fragment (scFv) , bis-scFv, (scFv) 2, diabody, minibody, nanobody, triabody, tetrabody, humabody, disulfide stabilized Fv protein (dsFv) , single-domain antibody (sdAb) , IgNAR, camelid antibody or a binding fragment thereof, bispecific antibody or a binding fragment thereof, or a chemically modified derivative thereof.
  • a conjugating moiety is a Fragment crystallizable domain (Fc domain) of an antibody, e.g., of IgG, IgD, IgA, IgE, or IgM.
  • a polypeptide herein comprises a fusion to an Fc portion of IgG (e.g., IgG1, IgG2, IgG3, or IgG4) .
  • a polypeptide herein is fused to the Fc portion is further conjugated to one or more conjugation moieties described below.
  • the Fc domain can be fused to the N-or C-terminus of an IL-2 mutein.
  • an IL-2 polypeptide herein exhibits a decreased binding affinity to the IL-2R ⁇ subunit relative to a wildtype IL-2 polypeptide.
  • the decreased binding affinity is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to the binding affinity of wildtype IL-2 polypeptide to IL-2R ⁇ .
  • the decreased binding affinity is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or more relative to a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein exhibits about the same binding affinity ( ⁇ 10%) to the IL-2R ⁇ subunit relative to a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein increases activation of CD8+ T cells relative to a wildtype IL-2 polypeptide.
  • the increased activation of CD8+T cells is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to activation of CD8+ T cells by a wildtype IL-2 polypeptide.
  • the increased activation of CD8+ T cells is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or greater relative to activation of CD8+T cells by a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein increases STAT5 phosphorylation in human CD8+ T cells relative to a wildtype IL-2 polypeptide.
  • the increased STAT5 phosphorylation in human CD8+ T cells is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to STAT5 phosphorylation in human CD8+ T cells by a wildtype IL-2 polypeptide.
  • the increased STAT5 phosphorylation in human CD8+ T cells is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or greater relative to STAT5 phosphorylation in human CD8+ T cells by a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein increases activation of NK cells relative to a wildtype IL-2 polypeptide.
  • the increased activation of NK cells is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to activation of NK cells by a wildtype IL-2 polypeptide.
  • the increased activation of NK cells is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or greater relative to activation of NK cells by a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein increases STAT5 phosphorylation in human NK cells relative to a wildtype IL-2 polypeptide.
  • the increased STAT5 phosphorylation in human NK cells is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to STAT5 phosphorylation in human NK cells by a wildtype IL-2 polypeptide.
  • the increased STAT5 phosphorylation in human NK cells is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or greater relative to STAT5 phosphorylation in human NK cells by a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein decreases activation of Tregs relative to a wildtype IL-2 polypeptide.
  • the decreased activation of Tregs is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to activation of Tregs by a wildtype IL-2 polypeptide.
  • the decreased activation of Tregs is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or greater relative to activation of Tregs by a wildtype IL-2 polypeptide.
  • an IL-2 polypeptide herein decreases STAT5 phosphorylation in human Tregs relative to a wildtype IL-2 polypeptide.
  • the decreased STAT5 phosphorylation in human Tregs is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or greater relative to STAT5 phosphorylation in human Tregs by a wildtype IL-2 polypeptide.
  • the decreased STAT5 phosphorylation in human Tregs is about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 30-fold, 50-fold, 100-fold, 200-fold, 300-fold, 500-fold, 1000-fold, or greater relative to STAT5 phosphorylation in human Tregs by a wildtype IL-2 polypeptide.
  • the wildtype IL-2 comprises an amino acid residue mutation at C125 of a sequence of wildtype human IL-2.
  • the amino acid residue mutation at C125 is C125A or C125S.
  • an IL-2 polypeptide herein comprises an IL-2 mutein is fused to a heterologous polypeptide (i.e., a polypeptide that is not IL-2 and preferably is not a variant of IL-2, e.g., a conjugated moiety described herein.
  • the heterologous polypeptide can increase the circulating half-life of the IL-2 polypeptide.
  • the polypeptide that increases the circulating half-life can be serum albumin, e.g., HSA.
  • an IL-2 polypeptide provided herein includes an amino acid sequence that is heterologous to wildtype human IL-2 or a functional variant thereof.
  • the amino acid sequence can be a half-life extension moiety that increases stability of the polypeptide by increasing in vivo half-life of the polypeptide, e.g., when administered to a subject.
  • the half-life extension moiety can be a protein, an antibody, an albumin, an immunoglobulin or a fragment thereof, a transferrin, or a PEG.
  • the antibody can be an anti-albumin antibody.
  • the albumin can be a serum albumin, e.g., a mouse serum albumin or a human serum albumin.
  • the immunoglobulin fragment is the Fc domain of a human immunoglobulin, e.g., IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM.
  • a human immunoglobulin e.g., IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM.
  • a polypeptide provided herein is an antibody fusion protein.
  • a polypeptide provided herein can be an immunoglobulin Fc fusion protein.
  • the polypeptide can include an immunoglobulin Fc domain.
  • the immunoglobulin Fc domain is a human immunoglobulin Fc domain, e.g., IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, IgM, or a fragment thereof.
  • Binding affinity of an IL-2 polypeptide provided herein to a target receptor or a subunit thereof can be assessed by measuring the dissociation constant (K D ) .
  • an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D that is at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 100-fold, or at least 1000-fold, or at least 10,000-fold, or at least 100,000-fold higher than the K D of wildtype IL-2.
  • an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D of at least 10 -9 M, at least 10 -8 M, or greater. In some embodiments, an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D of 10 -10 M, 10 -9 M, 10 -8 M, or greater. In some embodiments, an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D of from 10 -10 M to 10 -9 M, from 10 -9 M to 10 -8 M, from 10 -8 M to 10 -7 M.
  • an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D of about 3 x 10 -9 M, about 6 x 10 -9 M, about 1 x 10 -8 M, about 2 x 10 -8 M, or about 3 x 10 -8 M.
  • an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D that is approximately the K D of wildtype IL-2 binding to IL-2R ⁇ . In some embodiments, an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D that is at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold lower than the K D of wildtype IL-2. In some embodiments, an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D of about 10 -9 M or less. In some embodiments, an IL-2 polypeptide provided herein binds to IL-2R ⁇ with a K D of 10 -10 M, or less.
  • IL-2 signaling activity by an IL-2 mutant provided herein can be assessed by IL-2-mediated phosphorylation of STAT5.
  • Human peripheral blood mononuclear cells (PBMCs) can be treated with an IL-2 polypeptide provided herein and phosphorylation of STAT5 can be subsequently analyzed in NK cells and different T cell subsets by flow cytometry.
  • STAT5 phosphorylation can be used to calculate the half-maximal effect concentration or EC 50 .
  • Relative activity of an IL-2 mutant can be determined by comparing EC 50 values of mutant to wildtype.
  • IL-2 polypeptide comprises an isolated and purified IL-2 mutein, wherein the IL-2 polypeptide has a decreased affinity to IL-2R ⁇ relative to a wildtype IL-2.
  • the IL-2 polypeptide comprises an isolated and purified IL-2 mutein having one or more amino acid substitutions at positions K35, R38, F42, K43, F44, E61, E62, K64, P65, E68, L72, Y107, and/or C125 of wildtype human IL-2 (SEQ ID NO: 1) .
  • the IL-2 polypeptide preferentially interacts with the IL-2R ⁇ and IL-2R ⁇ subunits to form an IL-2/IL-2R ⁇ complex. Formation of the IL-2/IL-2R ⁇ complex can thereby stimulate or enhance expansion of CD4+ helper cells, CD8+ effector naive and memory T cells, NK cells, and/or NKT cells. In some cases, the expansion of Teff cells skews the Teff/Treg ratio toward the Teff population.
  • the cell proliferative disease or condition is a neoplastic disease, such as cancer.
  • the cancer is a metastatic cancer.
  • the cancer is a relapsed or refractory cancer.
  • the cancer is a cancer.
  • the treatment-naive cancer can be a cancer that has not been treated by a therapy.
  • the cancer is leukemia, lymphoma, sarcoma, myeloma, glioma, glioblastoma, glioblastoma multiforme, glioma, head and neck cancer, colorectal cancer, colon cancer, prostate cancer, castration-resistant prostate cancer, pancreatic cancer, melanoma, breast cancer (e.g., triple negative, ER positive, ER negative, chemotherapy resistant, trastuzumab-resistant, HER2 positive, doxorubicin resistant, tamoxifen resistant, ductal carcinoma, lobular carcinoma, primary, metastatic) , neuroblastoma, lung cancer (e.g., non-small cell lung carcinoma, squamous cell lung carcinoma (e.g., head, neck, or esophagus) , adenocarcinoma, large cell lung carcinoma, small cell lung carcinoma, carcinoid, sarcoma) , ovarian cancer, bone cancer (e.g.,
  • the cancer is a solid tumor.
  • solid tumors include bladder cancer, bone cancer, brain cancer, breast cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, kidney cancer, lung cancer, melanoma, ovarian cancer, pancreatic cancer, and prostate cancer.
  • the cancer is a hematologic malignancy, such as a leukemia, a lymphoma, or a myeloma.
  • the hematologic malignancy is a T cell malignancy.
  • the hematological malignancy is a B cell malignancy.
  • Non-limiting examples of hematologic malignancies include chronic lymphocytic leukemia (CLL) , small lymphocytic lymphoma (SLL) , follicular lymphoma (FL) , diffuse large B-cell lymphoma (DLBCL) , mantle cell lymphoma (MCL) , Waldenstrom’s macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt’s lymphoma, non-Burkitt high grade B cell lymphoma, primary mediastinal B cell lymphoma (PMBL) , immunoblastic large cell lymphoma, precursor B lymphoblastic lymphoma, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell
  • the inflammatory or autoimmune disease is atherosclerosis, obesity, inflammatory bowel disease (IBD) , rheumatoid arthritis, allergic encephalitis, psoriasis, atopic skin disease, osteoporosis, peritonitis, hepatitis, lupus, celiac disease, syndrome, polymyalgia rheumatica, multiple sclerosis (MS) , ankylosing spondylitis, type 1 diabetes mellitus, alopecia areata, vasculitis, and temporal arteritis, graft versus host disease (GVHD) , asthma, COPD, a paraneoplastic autoimmune disease, cartilage inflammation, juvenile arthritis, juvenile rheumatoid arthritis, pa
  • a pharmaceutical composition of the disclosure can be a combination of a compound, e.g., an IL-2 polypeptide described herein, with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • compositions for administration can include aqueous solutions of the active compounds in water-soluble form.
  • Suspensions of the active compounds can be prepared as oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension can also contain suitable stabilizers or agents which increase the solubility of a compound to allow for the preparation of highly concentrated solutions.
  • the active ingredient can be in powder form for constitution with a suitable vehicle, for example, sterile pyrogen-free water, before use.
  • therapeutically effective amounts of a compound described herein is administered in a pharmaceutical composition to a subject having a disease or condition to be treated.
  • the subject is a mammal such as a human.
  • a therapeutically effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors.
  • compositions can be formulated using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations that can be used pharmaceutically. Formulation can be modified depending upon the route of administration chosen.
  • Pharmaceutical compositions comprising compounds described herein can be manufactured, for example, by mixing, dissolving, emulsifying, encapsulating, entrapping, or compression processes.
  • compositions can include at least one pharmaceutically acceptable carrier, diluent, or excipient and compounds described herein as free-base or pharmaceutically acceptable salt form.
  • Pharmaceutical compositions can contain solubilizers, stabilizers, tonicity enhancing agents, buffers, and preservatives.
  • compositions comprising the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid, or liquid composition.
  • Solid compositions include, for example, powders, tablets, dispersible granules, capsules, and cachets.
  • Liquid compositions include, for example, solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein.
  • Semi-solid compositions include, for example, gels, suspensions, and creams.
  • compositions can be in liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions can also contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and other pharmaceutically acceptable additives.
  • Non-limiting examples of dosage forms suitable for use in the disclosure include liquid, powder, gel, nanosuspension, nanoparticle, microgel, aqueous or oily suspensions, emulsion, and any combination thereof.
  • Non-limiting examples of pharmaceutically acceptable excipients suitable for use in the disclosure include binding agents, disintegrating agents, anti-adherents, anti-static agents, surfactants, anti-oxidants, coating agents, coloring agents, plasticizers, preservatives, suspending agents, emulsifying agents, anti-microbial agents, spheronization agents, and any combination thereof.
  • a composition of the disclosure can be, for example, an immediate release form or a controlled release formulation.
  • An immediate release formulation can be formulated to allow the compounds to act rapidly following administration.
  • Non-limiting examples of immediate release formulations include readily dissolvable formulations.
  • a controlled release formulation can be a pharmaceutical formulation that has been adapted such that release rates and release profiles of the active agent can be matched to physiological and chronotherapeutic requirements, or has been formulated to effect release of an active agent at a programmed rate.
  • Non-limiting examples of controlled release formulations include granules, delayed release granules, hydrogels (e.g., of synthetic or natural origin) , other gelling agents (e.g., gel-forming dietary fibers) , matrix-based formulations (e.g., formulations comprising a polymeric material having at least one active ingredient dispersed through) , granules within a matrix, polymeric mixtures, and granular masses.
  • a controlled release formulation is a delayed release form.
  • a delayed release form can be formulated to delay a compound’s action for an extended period of time.
  • a delayed release form can be formulated to delay the release of an effective dose of one or more compounds, for example, for about 4, about 8, about 12, about 16, or about 24 hours.
  • a controlled release formulation can be a sustained release form.
  • a sustained release form can be formulated to sustain, for example, the compound’s action over an extended period of time.
  • a sustained release form can be formulated to provide an effective dose of any compound described herein (e.g., provide a physiologically effective blood profile) over about 4, about 8, about 12, about 16, or about 24 hours.
  • a compound described herein can be conveniently formulated into pharmaceutical compositions composed of one or more pharmaceutically acceptable carriers. See e.g., Remington’s Pharmaceutical Sciences, latest edition, by E.W. Martin Mack Pub. Co., Easton, PA, herein incorporated by reference in its entirety, which discloses pharmaceutically acceptable excipients and carriers, and methods of preparing pharmaceutical compositions.
  • Such carriers can be carriers for administration of compositions to humans and non-humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH.
  • Pharmaceutical compositions can also include one or more additional active ingredients such as antimicrobial agents, anti-inflammatory agents, and anesthetics.
  • compositions can include additional carriers, as well as thickeners, diluents, buffers, preservatives, and surface active agents in addition to the agents disclosed herein.
  • a pharmaceutical composition disclosed herein can be administered in a therapeutically effective amount by various forms and routes including, for example, oral, topical, parenteral, intravenous injection, intravenous infusion, subcutaneous injection, subcutaneous infusion, intramuscular injection, intramuscular infusion, intradermal injection, intradermal infusion, intraperitoneal injection, intraperitoneal infusion, intracerebral injection, intracerebral infusion, subarachnoid injection, subarachnoid infusion, intraocular injection, intraspinal injection, intrasternal injection, ophthalmic administration, endothelial administration, local administration, intranasal administration, intrapulmonary administration, rectal administration, intraarterial administration, intrathecal administration, inhalation, intralesional administration, intradermal administration, epidural administration, absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa) , intracapsular administration, subcapsular administration, intracardiac administration, transtracheal
  • a pharmaceutical composition can be administered in a local manner, for example, via injection of the compound directly into an organ, optionally in a depot or sustained release formulation or implant.
  • a pharmaceutical composition can be provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation.
  • a rapid release form can provide an immediate release.
  • An extended release formulation can provide a controlled release or a sustained delayed release.
  • a compound herein may be administered in combination with one or more therapeutic agents, for example, a cytokine, antiviral agent, or antifungal agent.
  • therapeutic agent encompasses any agent administered to treat a symptom or disease in an animal in need of such treatment.
  • the compound can also be administered as a component of a vaccine, i.e., combined with essentially any preparation intended for active immunological prophylaxis.
  • Toxicity and therapeutic efficacy of a compound herein can be determined by standard pharmaceutical procedures in cell culture or experimental animals.
  • Cell culture assays and animal studies can be used to determine the LD50 (the dose lethal to 50%of a population) and the ED50 (the dose therapeutically effective in 50%of a population) .
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50) .
  • a compound herein that exhibits large therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like.
  • a therapeutically effective dose can be estimated initially from cell culture assays by determining an IC50.
  • a dose can then be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by HPLC. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition.
  • An attending physician for patients treated with a compound herein would know how and when to terminate, interrupt, or adjust administration due to toxicity, organ dysfunction, and the like. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity) .
  • the magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.
  • a compound herein can be administered to an individual alone as a pharmaceutical preparation appropriately formulated for the route of delivery and for the condition being treated.
  • Suitable routes may include oral, rectal, transdermal, vaginal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections, and the like.
  • parenteral delivery including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections, and the like.
  • penetrants appropriate to the barrier to be permeated can be used in the formulation.
  • a compound herein can be formulated as a liquid with carriers that may include a buffer and or salt such as phosphate buffered saline.
  • a compound herein may be formulated as a solid with carriers or fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the formulated end product may be a tablet, pill, capsule, dragee, liquid, gel, syrup, slurry, suspension, and the like.
  • push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol may be used.
  • the push-fit capsules can contain the active ingredients in admixture with fillers as above while in soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • Formulation for oral delivery can involve conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, lyophilizing processes, and the like.
  • a compound herein also may be mixed with a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, sorbitol, and the like; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone (PVP) , and the like, as well as mixtures of any two or more thereof.
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, alginic acid or a salt thereof such as sodium alginate, and the like.
  • a compound herein can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank’s solution, Ringer’s solution, or physiological saline buffer.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • kits or article of manufacture comprising a polypeptide described herein, including a pharmaceutical composition thereof.
  • the kit can include instructions for use of a polypeptide such as in the methods provided herein.
  • the kit includes instructions for the use of a polypeptide in methods for treating a disorder described herein (e.g., a cancer) in a subject in need thereof by administering to the subject a therapeutically effective amount of the polypeptide.
  • the subject is a human.
  • the disorder is a cancer.
  • the polypeptide is an IL-2 polypeptide provided herein.
  • the kit can further include a container.
  • suitable containers include bottles, vials (e.g., dual chamber vials) , syringes (such as single or dual chamber syringes) , test tubes, and intravenous (IV) bags.
  • the container can be formed from a variety of materials such as glass or plastic.
  • the container can hold a formulation of an IL-2 polypeptide.
  • the formulation is a lyophilized formulation.
  • the formulation is a frozen formulation.
  • the formulation is a liquid formulation.
  • the kit may further comprise a label or a package insert, which is on or associated with the container, may indicate directions for reconstitution and/or use of the formulation.
  • the label or package insert may further indicate that the formulation is useful or intended for intravenous, subcutaneous, or other modes of administration for treating a disorder (e.g., a cancer) in a subject.
  • the container holding the formulation can be a single-use vial or a multi-use vial, which can allow for repeat administrations of the reconstituted formulation.
  • the kit can further include a second container comprising a suitable diluent.
  • the kit can further include other materials desirable from a commercial, therapeutic, and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • kits for a single dose-administration unit includes a container of an aqueous formulation of a therapeutic IL-2 polypeptide, including single or multi-chambered pre-filled syringes.
  • an article of manufacture or kit comprising the formulations described herein for administration in an auto-injector device.
  • An auto-injector can be described as an injection device that upon activation, will deliver its contents without additional necessary action from the patient or administrator. They are particularly suited for self-medication of therapeutic formulations when the delivery rate must be constant, and the time of delivery is greater than a few moments.
  • Example 1 Recombinant expression and preparation of mutant IL-2 proteins.
  • the numbering scheme of the mutation positions in TABLE 1 corresponds to a mature human IL-2 protein as in SEQ ID NO: 1.
  • a mature human IL-2 protein does not include methionine as the first amino acid residue at the N-terminus.
  • the numbering scheme starts with alanine as the first residue of the 133 amino acid sequence of SEQ ID NO: 1.
  • Some variants described herein also include a C125A mutation, which reduces the likelihood of the formation of dimeric IL-2.
  • N-terminal 18-amino acid signal peptide (from N-to C-terminus) :
  • An exemplary amino acid sequence of encoded IL-2 construct is (from N-to C-terminus) :
  • the bold segment represents the 18-amino acid signal peptide; the underlined segment represents the mature 133-amino acid IL-2 sequence; the italicized segment represents the G 4 S linker and 6X-His-tag.
  • An example DNA expression sequence of an IL-2 construct is (from N-to C-terminus) :
  • Segment (i) represents a NotI endonuclease cleavable site; Segment (ii) represents a Okaxaki fragment element; the Segment (iii) in bold represents the 18-amino acid signal peptide encoding sequence; the underlined Segment (iv) represents the mature IL-2 encoding sequence; the italicized Segment (v) represents the human G 4 S linker and 6X-His-tag encoding sequence; Segment (vi) represents a stop codon; and Segment (vii) represents a XbaI endonuclease cleavable site.
  • the confirmed DNA sequence was constructed into the expression vector pcDNA3.1 (Thermo Fisher, catalog no. V79020) .
  • the plasmid containing the IL-2 gene (FIG. 1) was transformed into E. coli DH5 ⁇ cells.
  • a large amount of both wildtype and mutant plasmids was obtained by culturing the E. coli DH5 ⁇ cells for amplification and plasmid purification.
  • Cells were inoculated in 300 mL of medium (Gibco TM FreeStyle TM 293 expression medium, catalog no. 12338018) in a 1-L shake flask at an inoculation volume of 0.5x10 6 cells/ml. The cells were incubated for 48-54 hr in a shaker incubator at 37 °C, 120 rpm, and 5%carbon dioxide concentration. The cells were then incubated for 24 hr in a bed incubator until the cell density reached 1x10 6 cells/ml. Then, 300 ⁇ g of wildtype or mutant expression plasmid was added to 30 ml of medium. The mixture was vortexed for 3s to thoroughly mix.
  • medium Gibco TM FreeStyle TM 293 expression medium, catalog no. 12338018
  • transfection reagent PEI MAX 40K (Polysciences, catalog no. 24765) was added to the transfection reagent /plasmid mixture. The mixture was held static for 20 min, and then added to the HEK293F cells. After transfection, the cells were incubated for 48-54 hr in a shaker incubator at 37 °C, 120 rpm, and 5%carbon dioxide concentration. After incubation, the supernatant of the culture medium was isolated by centrifugation and prepared for protein purification.
  • the wildtype IL-2 and mutant IL-2 proteins were purified using Ni-NTA agarose microspheres (Thermo Fisher, catalog no. R901) .
  • An appropriate amount of Ni-NTA agarose microspheres were added to the supernatant in the previous step, and incubated at 4 °C for 30 min.
  • An appropriate volume of wash buffer 50 mM PBS, pH 7.4, 10 mM imidazole
  • an appropriate volume of elution buffer 50 mM PBS, pH 7.4, 250 mM imidazole was added to elute the wildtype or mutant protein-bound microspheres and obtain the purified wildtype or mutant proteins.
  • the purified wildtype or mutant proteins were detected by SDS-PAGE protein gel.
  • the results of SDS-PAGE protein gel detection showed that the molecular weights of wildtype IL-2 protein, m21, m24, and m52 mutant proteins were consistent with the theoretical molecular weights, confirming that the correct proteins were expressed (FIG. 2) .
  • Size exclusion chromatography (SEC) was used to detect the purity and protein aggregation of the wildtype or mutant proteins.
  • the SEC (at 280 nm) results showed that the peaks of wildtype IL-2 protein, m21, m24, and m52 mutant proteins were all around 20 min, indicating that these proteins were monomeric proteins and did not produce aggregated proteins (FIG. 3) .
  • Example 2 Recombinant expression and preparation of IL-2-Fc fusion proteins.
  • Mutant IL-2-Fc fusion proteins may be generated based on the amino acid sequences of wildtype human IL-2 and IL-2 mutant proteins shown in TABLE 2.
  • the DNA sequence may be synthesized and confirmed by gene sequencing.
  • the confirmed DNA sequence may be constructed into the expression vector pcDNA3.1 (Thermo Fisher, catalog no. V79020) .
  • the plasmid containing the IL-2 gene may be transformed into E. coli DH5 ⁇ cells.
  • a large amount of both wildtype and mutant IL-2-Fc plasmid may be obtained by culturing the E. coli DH5 ⁇ cells for amplification and plasmid purification.
  • Cells may be inoculated in 300 mL of medium (Gibco TM FreeStyle TM 293 expression medium, catalog no. 12338018) in a 1-L shake flask at an inoculation volume of 0.5x10 6 cells/ml.
  • the cells may be incubated for 48-54 hr in a shaker incubator at 37 °C, 120 rpm, and 5%carbon dioxide concentration.
  • the cells may then be incubated for 24 hr in a bed incubator until the cell density reaches 1x10 6 cells/ml.
  • 300 ⁇ g of wildtype or mutant IL2-Fc expression plasmid may be added to 30 ml of medium. The mixture may then be vortexed for 3s to thoroughly mix.
  • transfection reagent PEI MAX 40K (Polysciences, catalog no. 24765) may be added to the transfection reagent /plasmid mixture. The mixture may be held static for 20 min, and then added to the HEK293F cells. After transfection, the cells may be incubated for 48-54 hrs in a shaker incubator at 37 °C, 120 rpm, and 5%carbon dioxide concentration. After incubation, the supernatant of the culture medium may be isolated by centrifugation and prepared for protein purification.
  • PEI MAX 40K Polysciences, catalog no. 24765
  • the wildtype IL-2-Fc and mutant IL-2-Fc proteins may be purified using Protein A agarose microspheres.
  • An appropriate amount of Protein A agarose microspheres may be added to the supernatant in the previous step, and incubated at 4 °C for 10 min. The mixture may then be subjected to low-speed centrifugation at 1000 rpm for 3 min to collect the IL-2-Fc fusion protein-bound microspheres.
  • An appropriate volume of wash buffer 50 mM PBS, pH 7.4
  • an appropriate volume of elution buffer (Glycine-HCl, pH 3) may be added to elute the wildtype or mutant protein-bound microspheres and obtain purified wildtype or mutant fusion proteins.
  • Example 3 Recombinant expression and preparation of IL-2-antibody fusion proteins.
  • Mutant IL-2-antibody fusion proteins may be generated based on the amino acid sequences of wildtype human IL-2 and IL-2 mutant proteins shown in TABLE 2.
  • the IL-2 mutant-antibody fusion proteins comprise IL-2 mutant sequences and complete antibodies or antigen-binding moiety thereof (such as PD-1 antibody) .
  • the IL-2 mutant may be linked to the N-terminus or C-terminus of the light chain or heavy chain of the antibody.
  • the DNA sequence may be synthesized and confirmed by gene sequencing.
  • the confirmed DNA sequence may be constructed into the expression vector pcDNA3.1 (Thermo Fisher, catalog no. V79020) .
  • the plasmid containing the IL-2 gene may be transformed into E. coli DH5 ⁇ cells.
  • a large amount of both wildtype and mutant-antibody plasmids may be obtained by culturing the E. coli DH5 ⁇ cells for amplification and plasmid purification.
  • Cells may be inoculated in 300 mL of medium (Gibco TM FreeStyle TM 293 expression medium, catalog no. 12338018) in a 1-L shake flask at an inoculation volume of 0.5x10 6 cells/ml.
  • the cells may be incubated for 48-54 hr in a shaker incubator at 37 °C, 120 rpm, and 5%carbon dioxide concentration.
  • the cells may then be incubated for 24 hr in a bed incubator until the cell density reached 1x10 6 cells/ml.
  • 300 ⁇ g of wildtype or mutant IL-2-Ab fusion expression plasmid may be added to 30 ml of medium.
  • the mixture may be vortexed for 3 s to thoroughly mix.
  • transfection reagent PEI MAX 40K (Polysciences, catalog no. 24765) may be added to the transfection reagent /plasmid mixture. The mixture may be held static for 20 min, and then added to the HEK293F cells. After transfection, the cells may be incubated for 48-54 hr in a shaker incubator at 37 °C, 120 rpm, and 5%carbon dioxide concentration. After incubation, the supernatant of the culture medium may be isolated by centrifugation and prepared for protein purification.
  • PEI MAX 40K Polysciences, catalog no. 24765
  • the wildtype IL-2-antibody and mutant IL-2-antibody fusion proteins may be purified using Protein A agarose microspheres.
  • An appropriate amount of Protein A agarose microspheres may be added to the supernatant in the previous step, and incubated at 4 °C for 10 min. The mixture may then be subjected to low-speed centrifugation at 1000 rpm for 3 min to collect the IL-2-antibody fusion protein-bound microspheres.
  • An appropriate volume of wash buffer (50 mM PBS, pH 7.4) may then be added to remove non-specific proteins.
  • an appropriate volume of elution buffer (Glycine-HCl, pH 3) may be added to elute the wildtype or mutant protein-bound microspheres and obtain purified wildtype or mutant fusion proteins.
  • Example 4 Determination of thermal stability of IL-2 mutant proteins.
  • the melting point T m (°C) values which indicate the structural stability of the protein samples, were obtained by monitoring the intrinsic tryptophan and tyrosine fluorescence at the emission wavelengths of 330 nm and 350 nm.
  • IL-2 mutant protein stability was assessed by detecting small changes in tryptophan and tyrosine fluorescence in the mutant IL-2 protein using the label-free nano differential scanning fluorimetry (nanoDSF) assay (Prometheus, PR NT. 48) .
  • nanoDSF nano differential scanning fluorimetry
  • wt (C145A) IL-2 and IL-2 mutant proteins were prepared as described in Example 1.
  • the wt (C145A) or mutant IL-2 protein samples were then diluted in PBS to a concentration of 0.5 mg/ml.
  • About 10 ⁇ l of protein sample (wt (C125A) or mutant IL-2 proteins) were loaded in the detection capillaries and then placed on the sample holder.
  • a temperature gradient of 1 °C/min from 25-90 °C was then applied, and the intrinsic protein fluorescence at 330 and 350 nm was recorded for each sample.
  • the data were analyzed using the data analysis software provided by the NT. 48 instrument.
  • Example 5 Determination of the affinity of mutant IL-2 protein to human IL-2R ⁇ receptor.
  • the IL-2R ⁇ -Fc fusion protein was diluted with buffer (10 mM PBS + 0.02%tween-20 + 0.1%BSA, pH 7.4) from the initial concentration of 1000 nM to 15.6 nM in a 4-fold gradient to yield a total of 5 concentrations: 1000 nM, 250 nM, 62.5 nM, and 15.6 nM, respectively.
  • NTA sensor was soaked in buffer (10 mM PBS + 0.02%tween-20 + 0.1%BSA, pH 7.4) for 10 min to equilibrate.
  • the IL-2 mutant protein was loaded onto the NTA sensor. The loading time was 90s. The mutant protein was then eluted and then equilibrated for 30 s. The mutant protein was then combined with 4 different concentrations of IL-2R ⁇ -Fc fusion protein. The binding time was 120 s. Finally, the bound IL-2R ⁇ -Fc fusion protein was eluted with buffer. The dissociation time was 180 s.
  • the BLI affinity test results of wildtype IL-2 and mutant IL-2 to IL-2R ⁇ are shown in TABLE 3.
  • the test results include the dissociation constant (K D ) , the rate constant of association of the mutant to the receptor (k on ) , and the rate constant of dissociation of the mutant from the receptor (k dis ) .
  • ND undetectable. Since the affinity of some mutants to IL-2R ⁇ was too low, the affinity could not be determined by fitting the data in the concentration range used in the experiment.
  • Example 6 Determination of the affinity of mutant IL-2 protein and human IL-2R ⁇ .
  • mutant IL-2 protein to human IL-2R ⁇ -Fc fusion protein (ACRO, Catalog: ILG-H5254) was determined by label-free biolayer interferometry ( R8, BLI) as follows:
  • the IL-2R ⁇ -Fc fusion protein was diluted with buffer (10 mM PBS + 0.02%tween-20 + 0.1%BSA, pH 7.4) from the initial concentration of 1000 nM to 15.6 nM in a 4-fold gradient to yield a total of 5 concentrations: 1000 nM, 250 nM, 62.5 nM, and 15.6 nM, respectively.
  • NTA sensor was soaked in buffer (10 mM PBS + 0.02%tween-20 + 0.1%BSA, pH 7.4) for 10 min to equilibrate.
  • the mutant IL-2 protein was loaded onto the NTA sensor. The loading time was 90s. The mutant protein was then eluted and then equilibrated for 30 s. The mutant protein was then combined with 4 different concentrations of IL-2R ⁇ -Fc fusion protein. The binding time was 120 s. Finally, the bound IL-2R ⁇ -Fc fusion protein was eluted with buffer. The dissociation time was 180 s.
  • Example 7 Determination of pSTAT5 phosphorylation in human peripheral blood by IL-2 mutants.
  • IL-2 biological activity of wildtype IL-2 or mutant IL-2 proteins was assessed by measuring the phosphorylation level of STAT5 in various cell types (including CD8+T cells, NK cells, and Tregs) in human peripheral blood mononuclear cells (PMBCs) .
  • Wildtype IL-2 or mutant IL-2 was diluted in basal medium to 1000 nM, 100 nM, 10 nM, 1 nM, 0.1 nM, 0.01 nM, 0.001 nM, and 0.0001 nM to produce a total of eight concentrations.
  • Phosflow TM Perm Buffer III (BD, Cat No. 558050) was added to the PBMCs. The cells were lysed by incubation for 30 min on ice. The cells were then washed twice with staining buffer (BD, Cat No. 554655) .
  • CD8+ T cells are defined as CD3+CD4-CD8+ cells
  • NK cells are defined as CD3-CD56+ cells
  • Treg cells are defined as CD3+CD4+CD25+Foxp3+ cells.
  • the pSTAT5 fluorescence values (MFI) of CD8+ T cells, NK cells, and Treg cells under different IL-2 concentrations were calculated and fitted by computer program or four-parameter regression calculation method.
  • Detection results of wildtype IL-2 and IL-2 mutants activating STAT5 phosphorylation in human CD8+ T cells are shown in TABLE 5.
  • the biological activities of m24 and m27 on CD8+ T cells increased by 470.89%and 455.32%, respectively.
  • Detection results of wildtype IL-2 and IL-2 mutants activating STAT5 phosphorylation in human NK cells are shown in TABLE 6.
  • the biological activities of m24 and m27 on NK cells increased by 578.08%and 612.56%, respectively.
  • Detection results of wildtype IL-2 and IL-2 mutants activating STAT5 phosphorylation in human Treg cells are shown in TABLE 7.
  • the IL-2 mutants all reduced or effectively eliminated activating STAT5 phosphorylation of Tregs.
  • Example 8 Evaluation of the therapeutic effect of IL-2 mutants in the C57BL/6 mouse B16-F10 cutaneous melanoma model.
  • B16-F10 melanoma cancer cells (about 5 x 10 5 cells) were implanted subcutaneously into the flanks of C57BL/6-IL2RB tm2 (IL2RB) IL2RG tm2 (IL2RG) /Bcgen mice (Beijing Biocytogen Co., Ltd, stock No.: 111850) . At around the 6 th day post-inoculation, the average tumor volume was about 100 mm 3 . The tumor-bearing mice were divided into 3 groups with 8 mice per group by random block method. Mice bearing B16-F10 cells were treated with either PBS, wt (C125A) , or m24 protein once daily for 10 days (10 doses) .
  • Tumor measurements were collected every 3 or 4 days throughout the study. Tumor measurements (length (L) and width (W) ) were collected three times per week using digital calipers, and the tumor volume was calculated using the formula: (LxWxW) /2. Finally, tumors were collected from mice treated with wt (C125A) or m24 protein to detect tumor infiltrating lymphocytes by flow cytometry.
  • the results showed comparable tumor growth inhibition in response to treatment with wt(C125A) and m24.
  • the flow cytometry analysis showed that the CD3+CD4-CD8+ /CD3+CD4+CD25+FOXP3+ Treg ratio was higher in m24 treated tumors than in wt (C125A) treated tumors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des polypeptides mutants d'IL-2 et des compositions de ceux-ci qui présentent une affinité de liaison réduite à l'IL-2Rα et/ou une affinité de liaison améliorée à l'IL-2 βγ. L'invention concerne en outre des procédés d'administration desdits polypeptides mutants d'IL-2 et des compositions de ceux-ci pour le traitement d'un état, tel que le cancer et des troubles immunitaires.
PCT/CN2023/105095 2022-07-01 2023-06-30 Polypeptides d'il-2 et procédés d'utilisation WO2024002363A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2022/103352 2022-07-01
CN2022103352 2022-07-01

Publications (1)

Publication Number Publication Date
WO2024002363A1 true WO2024002363A1 (fr) 2024-01-04

Family

ID=89383369

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/105095 WO2024002363A1 (fr) 2022-07-01 2023-06-30 Polypeptides d'il-2 et procédés d'utilisation

Country Status (1)

Country Link
WO (1) WO2024002363A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101426916A (zh) * 2004-03-05 2009-05-06 诺华疫苗和诊断公司 改进的白介素-2突变蛋白
CN105440123A (zh) * 2011-02-10 2016-03-30 罗切格利卡特公司 突变体白介素-2多肽
CN109952311A (zh) * 2016-11-15 2019-06-28 分子免疫中心 用于增加白介素-2及其衍生蛋白质的分泌水平的方法
WO2021030374A1 (fr) * 2019-08-15 2021-02-18 Cytimm Therapeutics, Inc. Polypeptides d'interleukine 2 (il-2) modifiés, conjugués et utilisations de ceux-ci
CN112771072A (zh) * 2018-07-24 2021-05-07 生物技术Rna制药有限公司 Il2激动剂
US20210324030A1 (en) * 2020-04-21 2021-10-21 Regeneron Pharmaceuticals, Inc. Il-2 variants with reduced binding to il-2 receptor alpha and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101426916A (zh) * 2004-03-05 2009-05-06 诺华疫苗和诊断公司 改进的白介素-2突变蛋白
CN105440123A (zh) * 2011-02-10 2016-03-30 罗切格利卡特公司 突变体白介素-2多肽
CN109952311A (zh) * 2016-11-15 2019-06-28 分子免疫中心 用于增加白介素-2及其衍生蛋白质的分泌水平的方法
CN112771072A (zh) * 2018-07-24 2021-05-07 生物技术Rna制药有限公司 Il2激动剂
WO2021030374A1 (fr) * 2019-08-15 2021-02-18 Cytimm Therapeutics, Inc. Polypeptides d'interleukine 2 (il-2) modifiés, conjugués et utilisations de ceux-ci
US20210324030A1 (en) * 2020-04-21 2021-10-21 Regeneron Pharmaceuticals, Inc. Il-2 variants with reduced binding to il-2 receptor alpha and uses thereof

Similar Documents

Publication Publication Date Title
AU2018309172B2 (en) Cytokine conjugates for the treatment of autoimmune diseases
US11919934B2 (en) IL-15 conjugates and uses thereof
JP7415047B2 (ja) インターロイキン-2受容体βγc二量体について偏向され、非ペプチド性水溶性ポリマーにコンジュゲートされたヒトインターロイキン-2コンジュゲート
US20210070827A1 (en) Il-2 conjugates and methods of use to treat autoimmune diseases
WO1996010089A1 (fr) Modification d'un peptide et d'une proteine
JP2022512746A (ja) インターロイキン-10ポリペプチド複合体、その二量体、およびそれらの使用
US20220273767A1 (en) Interleukin 10 Conjugates and Uses Thereof
JP2022069441A (ja) アデノシンおよび/またはメチルチオアデノシンの酵素媒介枯渇方法
KR20220151202A (ko) 인터류킨-2 폴리펩타이드 접합체 및 그의 사용 방법
WO2024002363A1 (fr) Polypeptides d'il-2 et procédés d'utilisation
KR20220008323A (ko) 면역 세포를 증식하는 il-2 돌연변이 단백질
WO2024002170A1 (fr) Polypeptides d'il-21 et méthodes d'utilisation
TW202409069A (zh) Il-2多肽和使用方法
TW202409068A (zh) Il—21多肽和使用方法
US20240132563A1 (en) Bifunctional cytokine compositions
RU2793754C2 (ru) Конъюгаты IL-15 и пути их применения
TWI839476B (zh) 介白素-2多肽共軛物及其用途
RU2787201C2 (ru) Конъюгаты цитокинов для лечения аутоиммунных заболеваний
TW202132331A (zh) 介白素—2多肽共軛物及其用途

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: 23830525

Country of ref document: EP

Kind code of ref document: A1