US20220048965A1 - Interleukin-4 receptor-binding fusion proteins and uses thereof - Google Patents

Interleukin-4 receptor-binding fusion proteins and uses thereof Download PDF

Info

Publication number
US20220048965A1
US20220048965A1 US17/397,287 US202117397287A US2022048965A1 US 20220048965 A1 US20220048965 A1 US 20220048965A1 US 202117397287 A US202117397287 A US 202117397287A US 2022048965 A1 US2022048965 A1 US 2022048965A1
Authority
US
United States
Prior art keywords
fusion protein
cell
protein
nucleic acid
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/397,287
Inventor
Fahar Merchant
Raj K. Puri
Bharatkumar H. Joshi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medicenna Therapeutics Inc
US Department of Health and Human Services
Original Assignee
Medicenna Therapeutics Inc
US Department of Health and Human Services
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 Medicenna Therapeutics Inc, US Department of Health and Human Services filed Critical Medicenna Therapeutics Inc
Priority to US17/397,287 priority Critical patent/US20220048965A1/en
Publication of US20220048965A1 publication Critical patent/US20220048965A1/en
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PURI, RAJ K., JOSHI, BHARATKUMAR H.
Assigned to MEDICENNA THERAPEUTICS, INC. reassignment MEDICENNA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MERCHANT, FAHAR
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5406IL-4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1761Apoptosis related proteins, e.g. Apoptotic protease-activating factor-1 (APAF-1), Bax, Bax-inhibitory protein(s)(BI; bax-I), Myeloid cell leukemia associated protein (MCL-1), Inhibitor of apoptosis [IAP] or Bcl-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2026IL-4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4747Apoptosis related proteins
    • 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]
    • C07K14/5437IL-13
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/48Regulators of apoptosis

Definitions

  • the present invention relates to interleukin-4 receptor-binding protein fusions. More specifically, the invention provides, in part, fusion proteins that include an interleukin-4 or interleukin-13 protein moiety joined to a pro-apoptotic Bcl-2 family member protein moiety.
  • Interleukin-4 is a pleiotropic cytokine produced by activated T cells, and is the ligand for the IL-4 receptor (IL-4R), which can also bind to interleukin-13 (IL-13).
  • the IL-4/1L-4Ra complex can then bind the second component of the IL-4 receptor, yc (the “Type I” receptor) with relatively low affinity. Additionally, the IL-4/IL-4Ra complex can also bind the interleukin-13 (IL-13) receptor al (IL-13R al) (the “Type II receptor).
  • Type 1 and Type 11 receptor chains Different cell types express different amounts of the Type 1 and Type 11 receptor chains.
  • 1L-4Ra is present on most cells
  • yc is generally expressed on hematopoietic cells
  • IL-13R al is generally expressed on non-hematopoietic cells. Accordingly, yc, but not IL-13R al, is found on T cells, natural killer (NK) cells, basophils, mast cells, and most mouse B cells (most human B cells express both yc and IL-13R al)
  • Some bone marrow-derived cells including macrophages and dendritic cells, express both yc and IL-13R al and consequently respond to both IL-4 and IL-13.
  • IL-13R al but little or no yc, is found on most non-bone marrow-derived cells, including smooth muscle and epithelial cells.
  • Circularly permuted molecules are those in which the termini of a linear molecule (e.g., ligand) have been joined together, either directly or via a linker, to produce a circular molecule, after which the circular molecule is opened at another location to produce a new linear molecule with termini different from the termini of the original molecule.
  • Circularly permuted variants of IL-4 have been described in, for example, U.S. Pat. No. 6,011,002, issued Jan. 4, 2000, to Pastan el al.
  • apoptosis is a common phenomenon in the development of animal cells and is both positively and negatively regulated. In addition to its involvement in neuronal and lymphoid system development and overall cell population homeostasis, apoptosis also plays a significant role in various diseases and injuries resulting from aberrant regulation of apoptotic pathways. For example, aberrant activation of neuronal cell death by apoptosis has been implicated in many neurodegenerative diseases and conditions, such as Alzheimer disease (Barinaga, Science 281:1303-1304), Huntington's disease, spinal-muscular atrophy, neuronal damage caused during stroke (reviewed in Rubin, British Med.
  • transient ischemic neuronal injury e.g., spinal cord injury
  • aberrant suppression of apoptosis can result in hyperproliferation of cells, leading to cancer and other hyperproliferative disorders.
  • Bcl-2 was one of the first proteins identified as regulating apoptosis (Cleary et al., Cell 47:19-28, 1986; Tsujimoto and Croce, Proc. Natl. Acad. Sci. USA 83:5214-5218, 1986). Since its discovery, several Bcl-2-related proteins (“Bcl-2 family proteins” or “Bcl-2 family members”) have been identified as regulators of apoptosis (White, Genes Dev. 10:1-15, 1996; Yang et al., Cell 80:285-291, 1995; Lomonosova, E. and G. Chinnadurai, Oncogene 27, S2S19, 2009).
  • chemotherapeutic agents act by inducing apoptosis in proliferating neoplastic cells, but their therapeutic value is limited by the extent to which they are toxic to normal cells.
  • Treatment with standard apoptosis inhibitory molecules for instance peptide-type caspase inhibitors (e.g., DEVD-type), has proven unsatisfactory for clinical work due to low membrane permeability of these inhibitors.
  • Targeted immunotoxins genetic or biochemical fusions between a toxic molecule, for instance a bacterial toxin, and a targeting domain derived, typically from an antibody molecule
  • DT diphtheria toxin
  • Pseudomonas exotoxin (PE) fusion proteins have been investigated as potential cancer therapeutics (Kreitman and Pastan, Blood 90:252-259, 1997; Shimamura et al. Cancer Res. 67:9903-9912; 2007).
  • the present invention relates to interleukin-4 receptor binding fusion proteins. More specifically, the invention provides, in part, fusion proteins that include a interleukin-4 receptor-binding protein moiety joined to a pro-apoptotic Bcl-2 family member protein moiety and uses thereof
  • the invention provides a fusion protein including a interleukin-4 (IL-4) receptor binding protein and a pro-apoptotic Bcl-2 family polypeptide.
  • IL-4 receptor binding protein may be circularly permuted (cp).
  • the Bcl-2 family polypeptide may be a pro-apoptotic Bcl-2 family polypeptide comprising a BH3 domain (such as Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak or Bax).
  • the BH3 domain may further include a mutation that reduces phosphorylation.
  • the pro-apoptotic Bcl-2 family polypeptide including a BH3 domain that further includes a mutation that reduces phosphorylation may be a Bad polypeptide.
  • the fusion protein may be capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis of a target cell expressing an IL-4R.
  • the IL-4 receptor binding protein may be a mutant IL-4 or IL-13 selective for binding to a Type I or a Type TT IL-4 receptor (IL-4R)
  • the mutant IL-4 selective for binding to a Type II IL-4R may include a KFR variant or a KF variant.
  • the mutant IL-4 selective for binding to a Type I IL-4R may include an RGA variant.
  • the mutant IL-13 may be an All variant or a DN variant.
  • the fusion protein may further include a linker.
  • the linker may have the sequence GS or may be a ubiquitin or ubiquitin variant molecule.
  • the fusion protein may include the sequence set forth in SEQ ID NOs: 24-27.
  • nucleic acid molecule encoding a fusion protein as described herein, or a vector including the nucleic acid molecule, or a host cell including the vector.
  • nucleic acid molecule encoding a fusion protein as set forth in SEQ ID NOs: 24-27 or comprising SEQ ID NOs: 35-38.
  • a pharmaceutical composition including a fusion protein as described herein, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule, or a host cell including the vector.
  • a method of inducing cell death by administering a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule, or a host cell including the vector, to a subject in need thereof.
  • a method of inducing cell death by contacting a target cell that expresses an IL-4R with a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule.
  • a method of treating cancer by administering: a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule, or a host cell including the vector, to a subject in need thereof.
  • a method of treating cancer by contacting a neoplastic cell that expresses an IL-4R with a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule.
  • a method of treating cancer by contacting a non-malignant cell that expresses an IL-4R in a tumour microenvironment in a subject in need thereof with a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule.
  • the non-malignant cell is contacted prior to the subject starting a therapy.
  • a method of treating a hyperproliferative or differentiative disorder by administering a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule to a subject in need thereof.
  • the hyperproliferative or differentiative disorder may be a fibrosis or hyperplasia, an inflammatory conditions or an autoimmune condition.
  • the fibrosis or hyperplasia may be pulmonary fibrosis or hyperplasia (such as benign prostatic hyperplasia), cardiac fibrosis, or liver fibrosis;
  • the inflammatory condition may be prostatitis, vernal keratoconjunctivitis, artherosclerosis, or idiopathic pulmonary pneumonia; or the autoimmune condition may be Graves disease.
  • a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule for inducing cell death or treating cancer or treating a hyperproliferative or differentiative disorder in a subject in need thereof.
  • the subject may be a human.
  • FIG. 1 is an illustration of a cp1L-4BAD (cpIL-4g:s-BADaa) in a pET 24a expression vector;
  • FIG. 2 is a graph showing the effect of cp1L-4BAD (cpIL-4BADaa) on IL-4Ra positive tumor cell (U 251) viability;
  • FIG. 3 is a graph showing the effect of cp1L-4BAD (cpIL-4BADaa) (squares) on 1L-4Ra positive tumor cell (U 251) viability in the presence of excess IL-4 (triangles);
  • FIG. 4 is a graph showing the effect of cp1L-4BAD (cpIL-4BADaa) on IL-4Ra positive tumor cell (Daudi cell) viability;
  • FIG. 5 is a graph showing the effect of cp1L-4BAD (cp1L-4BADaa) (triangles) on IL-41ta positive tumor cell Daudi viability in the presence of excess IL-4 (squares);
  • FIG. 6 is a graph showing the effect of cp1L-4BAD (cp1L-4BADaa) on the colony number of IL-4Ra positive tumor cells (U 251);
  • FIG. 9 shows an illustration of a pGWO7 E. coli expression vector
  • FIGS. 10A-F show the nucleic acid (SEQ ID NOs: 29, 30 and 32) and amino sequences (SEQ ID NOs: 18, 19 and 21) of IL-4BAD ( FIGS. 10A and 10B ), cpIL-4BAD ( FIGS. 10C and 10D ) and cpS4-BAD ( FIGS. 10E and 10F ) fusion constructs;
  • FIGS. 11A-B show the nucleic acid ( FIG. 11A ; SEQ ID NO: 37) and amino sequence ( FIG. 11B ; SEQ ID NO: 26) of a pKFR4-BAD-H6 fusion construct.
  • the present disclosure provides, in part, a fusion protein including an IL-4R binding protein joined to a pro-apoptotic Bcl-2 family protein and uses thereof.
  • IL-4R binding proteins include IL-4 and IL-13.
  • IL-4 proteins or IL-4 “protein moieties” include native IL-4 proteins, as well as variant IL-4 proteins.
  • a “native” or “wild type” IL-4 sequence refers to a human IL-4 sequence, whether purified from natural sources or made using recombinant techniques, and including the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • IL-4 sequences include the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • SEQ ID NO: 2 MHKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAASKDTTEKETFCRA ATVLRQFYSHHEKDTRCLGATAQQFH RH KQURFLKRLDRN LWGLAGLNSCPVKEANQSTLENFLERLKTEVIREKYSKCSS.
  • IL-4 proteins that can be used in the fusion proteins of the present disclosure are variant IL-4 proteins that have increased selectivity for yc (Type I receptor) relative to IL-13R al (Type II receptor) or vice versa as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012).
  • a variant IL-4 protein that has increased selectivity for yc is an IL-4 protein that includes the following mutations relative to the sequence of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus: R121Q/Y124W/S125F (the “RGA” or “super-4” or “S4” variant) as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012).
  • a variant IL-4 protein that has increased selectivity for IL-13R al is an IL-4 protein that includes the following mutations relative to the sequence of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus: R121K/Y124F/S125R (the “KFR” or “KFR4” variant) or R121K/Y124F (the “KF” variant).
  • IL-4 proteins that can be used in the fusion proteins of the present disclosure are circularly permuted (cp), as described in, for example, U.S. Pat. No. 6,011,002, issued Jan. 4, 2000, to Pastan et al.
  • a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, as follows:
  • a cp1L-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, in the context of an “RGA” or “super-4” or “S4” variant, as follows:
  • a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, in the context of a “KFR” variant, as follows:
  • a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, in the context of a “KF” variant, as follows:
  • a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 105-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-104 with a GGNGG linker and an initial methionine residue, as described in, for example, U.S. Pat. No. 6,011,002, issued Jan. 4, 2000, to Pastan et al.
  • Exemplary IL-4 proteins that can be used in the fusion proteins of the present disclosure include those described herein, as well as sequences having at least 80% sequence identity, at least 85%, at least 90%, at least 95%, at least 98% or even at least 99% sequence identity to native IL-4 (“variant IL-4 proteins”), as long as the variant IL-4 protein retains the ability to bind the IL-4 receptor, or retains increased selectivity for the yc (Type I receptor) relative to IL-13R al (Type II receptor) or vice versa as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012), or retains a desired biological activity.
  • variant IL-4 proteins include those described herein, as well as sequences having at least 80% sequence identity, at least 85%, at least 90%, at least 95%, at least 98% or even at least 99% sequence identity to native IL-4 (“variant IL-4 proteins”), as long as the variant IL-4 protein retains the ability to bind
  • IL-4 proteins include fragments that can be smaller than the native 129 amino acid IL-4 protein, as long as the IL-4 protein fragment retains the ability to bind the IL-4 receptor, or retains increased selectivity for the yc (Type I receptor) relative to IL-13R al (Type II receptor) or vice versa as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012), or retains a desired biological activity, whether as a fragment of the native sequence, or in a cp form or fragment thereof
  • nucleic acid molecules that encode an IL-4 protein as described herein or known in the art, including but not limited to RNA sequences corresponding to the DNA sequences described herein.
  • Exemplary IL-4 nucleic acid molecules include:
  • IL-13 proteins or IL-13 “protein moieties” include native IL-13 proteins, as well as variant IL-13 proteins.
  • a “native” or “wild type” IL-13 sequence refers to a human IL-13 sequence, whether purified from natural sources or made using recombinant techniques, and including the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • IL-13 proteins that can be used in the fusion proteins of the present disclosure are variant IL-13 proteins that have increased selectivity for IL-13Ra1 (type II receptor) relative wild-type IL-13 protein.
  • the IL-13 variant sequence may include the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • a variant IL-13 protein that has increased selectivity for IL-13Ra1 (type II receptor) relative wild-type IL-13 protein is an IL-13 protein that includes the following mutations relative to the sequence of native human IL-13 (SEQ ID NO: 7), the numbering excluding the methionine at the N-terminus:
  • the IL-13 variant sequence may include the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • IL-13 proteins that can be used in the fusion proteins of the present disclosure are circularly permuted (cp).
  • a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure includes an IL-13 protein in which residues 44-114 of native human IL-13 (SEQ ID NO: 7) are joined to residues 1-43 with a linker and an initial methionine residue, as follows:
  • a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure is as follows:
  • a variant cp1L-13 protein that can be used in the fusion proteins of the present disclosure includes an IL-13 protein in which residues 44-114 of native human IL-13 (SEQ ID NO: 7) are joined to residues 1-43 with a linker and an initial methionine residue, in the context of the “Al 1” variant, as follows:
  • a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure is as follows:
  • a variant cp1L-13 protein that can be used in the fusion proteins of the present disclosure includes an IL-13 protein in which residues 44-114 of native human IL-13 (SEQ ID NO: 7) are joined to residues 1-43 with a linker and an initial methionine residue, in the context of the “DN” variant, as follows:
  • a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure is as follows:
  • Exemplary IL-13 proteins that can be used in the fusion proteins of the present disclosure include those described herein, as well as sequences having at least 80% sequence identity, at least 85%, at least 90%, at least 95%, at least 98% or even at least 99% sequence identity to native IL-13 (“variant IL-13 proteins”), as long as the variant IL-13 protein retains the ability to bind the IL-13 receptor, or retains increased selectivity for the IL-13Ra1 (type II receptor) relative to wild-type IL-13 protein, or retains a desired biological activity.
  • IL-13 proteins include fragments that can be smaller than the native 114 amino acid IL-13 protein, as long as the IL-13 protein fragment retains the ability to bind the IL-13 receptor, or retains increased selectivity for the IL-13Ra1 (type II receptor) relative to wild-type IL-13 protein, or retains a desired biological activity.
  • nucleic acid molecules including but not limited to RNA sequences or DNA sequences
  • IL-13 protein as described herein or known in the art.
  • Bcl-2-related proteins or polypeptides (“Bcl-2 family proteins” or “Bcl-2 family members”) are involved in regulation of apoptosis.
  • Bcl-2 family proteins fall into two distinct categories: those that inhibit cell death (the “anti-apoptotic” Bcl-2 family proteins) and those that enhance cell death (the “pro-apoptotic” Bcl-2 family proteins).
  • Bcl-2 family proteins share one to four conserved Bcl-2 homology (BH) domains, designated BH1, BH2, BH3, and BH4.
  • Pro-apoptotic Bcl-2 family proteins include those having a BH3 domain, such as Bad (e.g., Accession no: NP116784, CAG46757 or Q92934), Bik/Nbk (e.g., Accession no: CAG30276 or Q13323), Bid (e.g., Accession no: CAG28531 or P55957), Bim/Bod (e.g., Accession no: NP619527), Hrk (Accession no: 000198), Bak, or Bax.
  • Bad e.g., Accession no: NP116784, CAG46757 or Q92934
  • Bik/Nbk e.g., Accession no: CAG30276 or Q13323
  • Bid e.g., Accession no: CAG28531 or P55957
  • Bim/Bod e.g., Accession no: NP619527
  • Hrk Accession no: 000198
  • pro-apoptotic Bcl-2 family proteins that can be used in the fusion proteins according to the present disclosure are mutated (for example at serine residues e.g., serine to alanine mutations) to prevent phosphorylation.
  • Bad, Bcl-2-associated agonist of cell death is a regulator of programmed cell death (apoptosis).
  • Bad positively regulates cell apoptosis by forming heterodimers with Bcl-xL and Bcl-2, and reversing their death repressor activity.
  • Pro-apoptotic activity of Bad is regulated through its phosphorylation.
  • Exemplary Bad proteins that can be used in the fusion proteins of the present disclosure include those in GenBank Accession Nos. CAG46757; AAH01901.1; and CAG46733.1, as well as those sequences provided in U.S. Pat. No.
  • a Bad protein that can be used in the fusion proteins according to the present disclosure contains serine mutations at positions 112 and/or 136 to reduce phosphorylation.
  • a Bad protein that can be used in the fusion proteins according to the present disclosure contains serine to alanine mutations at positions 112 and/or 136 to reduce phosphorylation.
  • a Bad protein that can be used in the fusion proteins according to the present disclosure includes a sequence as follows, or fragment thereof:
  • a Bad protein that can be used in the fusion proteins according to the present disclosure includes a variant sequence as follows, or fragment thereof:
  • An exemplary Bik/Nbk protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows, or fragment thereof:
  • An exemplary Bid protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows, or fragment thereof:
  • An exemplary Bim/Bod protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows:
  • Hrk protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows:
  • a pro-apoptotic Bcl-2 family protein includes at least a fragment of a Bcl-2 family member, where the pro-apoptotic Bcl-2 family protein or fragment is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis.
  • inhibiting cell survival is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will survive.
  • inhibitting cell proliferation is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the growth or proliferation of a cell.
  • enhancing cell death or apoptosis is meant increasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will undergo apoptotic, necrotic, or any other form of cell death.
  • Suitable assays for measuring the inhibition of cell survival, inhibition of cell proliferation, or enhancement of cell death or apoptosis are described herein or known in the art.
  • nucleic acid molecules e.g., RNA sequences or DNA sequences
  • RNA sequences or DNA sequences that encode a pro-apoptotic Bcl-2 family member as described herein, including but not limited to RNA sequences corresponding to the DNA sequences described herein.
  • An exemplary pro-apoptotic Bcl-2 family member nucleic acid molecule includes:
  • Fusion proteins include IL-4R binding proteins, such as IL-4 and IL-13, joined to a pro-apoptotic Bcl-2 family member, with optional additional sequences or moieties (such as linkers), as described herein, as well as nucleic acid molecules encoding such fusion proteins. Also encompassed are recombinant nucleic acid molecules in which a nucleic acid sequence encoding a fusion protein is operably linked to a promoter, vectors containing such a molecule, and transgenic cells comprising such a molecule.
  • IL-4 (including cp1L-4 and IL-4 fragments and variants) can be linked to pro-apoptotic Bcl-2 family polypeptides comprising a BH3 domain as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak, or Bax or combinations thereof, or fragments or variants thereof, as long as pro-apoptotic activity is retained.
  • Any form or derivative of IL-4 can be used.
  • IL-4 or fragments of IL-4 that bind to the IL-4 receptor can be used.
  • pro-apoptotic Bcl-2 family proteins or fragments or variants thereof can be joined to 1L-4 or fragments or variants thereof or multiple 1L-4 proteins or fragments or variants thereof can be joined to pro-apoptotic Bcl-2 family proteins or fragments or variants thereof.
  • IL-13 (including IL-13 fragments or variants) can be linked to pro-apoptotic Bcl-2 family polypeptides, for example those comprising a BH3 domain, as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, or Hrk, or combinations thereof, as long as the combination or fragments or variants thereof retains pro-apoptotic activity.
  • pro-apoptotic Bcl-2 family polypeptides for example those comprising a BH3 domain, as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, or Hrk, or combinations thereof, as long as the combination or fragments or variants thereof retains pro-apoptotic activity.
  • Any form or derivative of IL-13 can be used.
  • IL-13 or fragments of IL-13 that bind to the IL-13 receptor can be used.
  • pro-apoptotic Bcl-2 family proteins or fragments or variants thereof can be joined to IL-13 or fragments or variants thereof or multiple IL-13 proteins or fragments or variants thereof can be joined to pro-apoptotic Bcl-2 family proteins or fragments or variants thereof.
  • a cp1L-4 can be linked to pro-apoptotic Bcl-2 family polypeptides, such as those comprising a BH3 domain as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak, or Bax or combinations thereof, or fragments or variants thereof, as long as pro-apoptotic activity is retained.
  • pro-apoptotic Bcl-2 family polypeptides such as those comprising a BH3 domain as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak, or Bax or combinations thereof, or fragments or variants thereof, as long as pro-apoptotic activity is retained.
  • Any form or derivative of cpIL-4 can be used.
  • multiple cpIL-4 proteins or fragments or variants thereof can be joined to a pro-apoptotic Bcl-2 family protein or fragments or variants thereof, or multiple pro-apoptotic Bcl-2 family proteins or fragments or variants thereof, can be joined to cp1L-4 proteins or fragments or variants thereof.
  • Exemplary fusion proteins are listed in Table 1.
  • IL-4/Bcl-2 Family Fusion Proteins Circularly permuted Bcl-2 Family Name
  • IL-4 Linker Protein Description IL4- MHKCDITLQEIIKTLN GS FQIPEFEPSEQED Human IL-4 Bad SLTEQKTLCTELTVT SSSAERGLGPSP fused to human DIFAASK D TTEKETF AGDGPSGSGKHH Bad variant CRAATVLRQFYSHH RQAPGLLWDASH via a GS EKDTRCLGATAQQF QQEQPTSSSHHG linker HRHKQLIRFLKRLDR GAGAVEIRSRHSA NLWGLAGLNSCPVK YPAGTEDDEGMG EANQSTLENFLERLK EEPSPFRGRSRA TIIVIREKYSKCSS APPNLWAAQRYG (SEQ ID NO: 2).
  • the joining or “fusion” of an IL-4R binding protein, such as IL-4 or IL-13, to a pro-apoptotic Bcl-2 family member may be direct, such that one portion of the IL-4R binding protein is directly attached to a portion of the pro-apoptotic Bcl-2 family member.
  • one end of the amino acid sequence of an IL-4R binding protein can be directly attached to an end of the amino acid sequence of the pro-apoptotic Bcl-2 family member.
  • the C-terminus of the IL-4R binding protein can be linked to the N-terminus of the pro-apoptotic Bcl-2 family member, or the C-terminus of the pro-apoptotic Bcl-2 family member can be linked to the N-terminus of the IL-4R binding protein.
  • Methods of generating such fusion proteins are routine in the art, for example using recombinant molecular biology methods.
  • an IL-4R binding protein moiety can be linked to the pro-apoptotic Bcl-2 family member moiety indirectly through a linker.
  • the linker can serve, for example, simply as a convenient way to link the two moieties, as a means to spatially separate the two moieties, to provide an additional functionality to the IL-4R binding protein or the pro-apoptotic Bcl-2 family member, or a combination thereof
  • the linker joining the IL-4R binding protein moiety and the pro-apoptotic Bcl-2 family member moiety can be designed to (1) allow the two molecules to fold and act independently of each other, (2) not have a propensity for developing an ordered secondary structure which could interfere with the functional domains of the two moieties, (3) have minimal hydrophobic or charged characteristics which could interact with the functional protein domains and/or (4) provide steric separation of the two regions.
  • the linker can also be used to provide, for example, lability to the connection between the IL-4R binding protein and the pro-apoptotic Bcl-2 family member, an enzyme cleavage site (for example, a cleavage site for a protease), a stability sequence, a molecular tag, a detectable label, or various combinations thereof.
  • a linker can be present between two domains of an IL-4R binding protein (such as in a cp molecule) or pro-apoptotic Bcl-2 family member.
  • the linker can be bifunctional or polyfunctional, i.e., contain at least about a first reactive functionality at, or proximal to, a first end of the linker that is capable of bonding to, or being modified to bond to, the IL-4R binding protein and a second reactive functionality at, or proximal to, the opposite end of the linker that is capable of bonding to, or being modified to bond to, the pro-apoptotic Bcl-2 family member being modified.
  • the two or more reactive functionalities can be the same (i.e. the linker is homobifunctional) or they can be different (i.e. the linker is heterobifunctional).
  • the length and composition of a linker can be varied considerably.
  • the length and composition of the linker are generally selected taking into consideration the intended function of the linker, and optionally other factors such as ease of synthesis, stability, resistance to certain chemical and/or temperature parameters, and biocompatibility.
  • the linker should not significantly interfere with the activity of the IL-4R binding protein and/or pro-apoptotic Bcl-2 family member.
  • Linkers suitable for use in a fusion protein according to the present disclosure include peptides.
  • the linker can be attached to the IL-4R binding moiety and/or the pro-apoptotic Bcl-2 family member moiety using recombinant DNA technology.
  • recombinant DNA technology Such methods are well-known in the art and details of this technology can be found, for example, in Sambrook, et al. Molecular Cloning: A Laboratory Manual. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989 or Ausubel et al. Current Protocols in Molecular Biology, John Wiley & Sons, 1994) or updates thereto.
  • the linker peptides can have a chain length of 1 to 500 amino acid residues (such as 1 to 100, 1 to 50, 6 to 30, 1 to 40, 1 to 20, or less than 30 amino acids or 5 to 10 amino acids).
  • a linker can be 2, 3, 4, 5, 6, 7, or 8 amino acids in length, or can be about 10, 20, 30, 40 or 50 amino acids in length.
  • a linker may for instance include the amino acid sequence Gly-Ser (GS) or may be the amino acid sequence Gly-Ser (GS) or may include a ubiquitin sequence:
  • GGGSM QI FVRTLTGRTITLEVEPSDTI EN VRAR I QDREGI PPDQQRLI FAGRQLEDGRTLS DYNIQRESTLHLVLRLRGGGS (SEQ ID NO: 28) or variant thereof.
  • Ubiquitin molecules suitable for use as linkers are described in, for example, Bachran, C. el al. “Anthrax toxin-mediated delivery of the Pseudomonas exotoxin A enzymatic domain to the cytosol of tumor cells via cleavable ubiquitin fusions MBio. 2013 Apr. 30; 4(3):e00201-13, or in PCT publication WO/2012/139112.
  • Peptide linkers that are susceptible to cleavage by enzymes of the complement system, urokinase, tissue plasminogen activator, trypsin, plasmin, or another enzyme having proteolytic activity may be used in one example.
  • the IL-4R binding protein can be attached via a linker susceptible to cleavage by enzymes having a proteolytic activity such as a urokinase, a tissue plasminogen activator, plasmin, thrombin or trypsin.
  • the IL-4R binding protein can be attached to the pro-apoptotic Bcl-2 family member via disulfide bonds (for example, the disulfide bonds on a cysteine molecule).
  • a fusion protein according to the present disclosure may include a pro-apoptotic Bcl-2 family member and an IL-4R binding protein linked by a cleavable linker region.
  • the cleavable linker region can be a protease-cleavable linker, although other linkers, cleavable for example by small molecules, may be used.
  • protease cleavage sites include those cleaved by factor Xa, thrombin and collagenase.
  • the protease cleavage site include those cleaved by a protease that is associated with a disease.
  • the protease cleavage site is one that is cleaved by a protease that is up-regulated or associated with cancers in general.
  • proteases are uPA, the matrix metalloproteinase (MiMP) family, the caspases, elastase, prostate specific antigen (PSA, a serine protease), and the plasminogen activator family, as well as fibroblast activation protein.
  • the cleavage site is cleaved by a protease secreted by cancer-associated cells. Examples of these proteases include matrixmetalloproteases, elastase, plasmin, thrombin, and uPA.
  • the protease cleavage site is one that is up-regulated or associated with a specific cancer.
  • the precise sequences are available in the art and the skilled person will have no difficulty in selecting a suitable cleavage site.
  • the protease cleavage region targeted by Factor Xa is IEGR.
  • the protease cleavage region targeted by enterokinase is DDDDK.
  • the protease cleavage region targeted by thrombin is LVPRG.
  • the cleavable linker region is one which is targeted by endocellular proteases.
  • the linker can be attached to the IL-4R binding protein moiety and/or pro-apoptotic Bcl-2 family member moiety using routine techniques as known in the art.
  • Fusion proteins can be prepared using routine methods as known in the art. Fusion proteins, as well as modifications thereto, can be made, for example, by engineering the nucleic acid encoding the fusion protein using recombinant DNA technology or by peptide synthesis. Modifications to the fusion protein may be made, for example, by modifying the fusion protein polypeptide itself, using chemical modifications and/or limited proteolysis. Combinations of these methods may also be used to prepare the fusion proteins.
  • fusion proteins can be produced in a prokaryotic host (e.g., E. coli, A. salmonicida or B.
  • subtilis or in a eukaryotic host (e.g., Saccharomyces or Pichia ; mammalian cells, e.g., COS, NIH 3T3, CHO, BHK, 293, or HeLa cells; or insect cells (baculovirus)).
  • a eukaryotic host e.g., Saccharomyces or Pichia ; mammalian cells, e.g., COS, NIH 3T3, CHO, BHK, 293, or HeLa cells; or insect cells (baculovirus)
  • the fusion proteins can be purified from the host cells using standard techniques known in the art.
  • Sequences for various exemplary fusion proteins are provided in Table 1. Variants and homologs of these sequences can be cloned, if an alternative sequence is desired, using standard techniques (see, for example, Ausubel et al., Current Protocols in Molecular Biology, Wiley & Sons, NY (1997 and updates); Sambrook et al., Sambrook, et al. Molecular Cloning: A Laboratory Manual. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989 or updates thereto).
  • the nucleic acid sequence can be obtained directly from a suitable organism, such as Aeromonas hydrophila , by extracting mRNA and then synthesizing cDNA from the mRNA template (for example by RT-PCR) or by PCR-amplifying the gene from genomic DNA.
  • a suitable organism such as Aeromonas hydrophila
  • the nucleic acid sequence encoding either the IL-4R binding moiety or the pro-apoptotic Bcl-2 family moiety can be obtained from an appropriate cDNA library by standard procedures.
  • the isolated cDNA is then inserted into a suitable vector, such as a cloning vector or an expression vector.
  • Mutations can be introduced at specific, pre-selected locations by in vitro site-directed mutagenesis techniques well-known in the art. Mutations can be introduced by deletion, insertion, substitution, inversion, or a combination thereof, of one or more of the appropriate nucleotides making up the coding sequence.
  • the expression vector can further include regulatory elements, such as transcriptional elements, required for efficient transcription of the fusion protein-encoding sequences.
  • regulatory elements such as transcriptional elements
  • Examples of regulatory elements that can be incorporated into the vector include, but are not limited to, promoters, enhancers, terminators, and polyadenylation signals.
  • Vectors that include a regulatory element operatively linked to a nucleic acid sequence encoding a genetically engineered fusion protein can be used to produce the fusion protein.
  • the expression vector may additionally contain heterologous nucleic acid sequences that facilitate the purification of the expressed fusion protein, such as affinity tags such (e.g., metal-affinity tags, histidine tags, avidin/streptavidin encoding sequences, glutathione-S-transferase (GST) encoding sequences, maltose binding protein (MBP) encoding sequences or biotin encoding sequences).
  • affinity tags such as metal-affinity tags, histidine tags, avidin/streptavidin encoding sequences, glutathione-S-transferase (GST) encoding sequences, maltose binding protein (MBP) encoding sequences or biotin encoding sequences.
  • GST glutathione-S-transferase
  • MBP maltose binding protein
  • biotin encoding sequences biotin encoding sequences
  • such tags are attached to the N- or C-terminus of a fusion protein
  • the fusion protein can include one or more linkers, as well as other moieties, as desired and/or as discussed herein. These can include a binding region, such as avidin or an epitope, or a tag such as a polyhistidine tag, which can be useful for purification and processing of the fusion protein, as well as other linkers as described herein.
  • detectable markers can be attached to the fusion protein, so that the traffic of the fusion protein through a body or cell can be monitored conveniently. Such markers include radionuclides, enzymes, fluorophores, chromophores, and the like.
  • DNA can be altered in numerous ways without affecting the biological activity of the encoded protein.
  • PCR can be used to produce variations in the DNA sequence which encodes a fusion protein.
  • variations in the DNA sequence encoding a fusion protein can be used to optimize for codon preference in a host cell used to express the protein, or may contain other sequence changes that facilitate expression.
  • a covalent linkage of an IL-4R binding protein directly to a pro-apoptotic Bcl-2 family member or via a linker may take various forms as is known in the art.
  • the covalent linkage may be in the form of a disulfide bond.
  • the DNA encoding one of the components can be engineered to contain a unique cysteine codon.
  • the second component can be derivatized with a sulfhydryl group reactive with the cysteine of the first component.
  • a sulfhydryl group either by itself or as part of a cysteine residue, can be introduced using solid phase polypeptide techniques. For example, the introduction of sulfhydryl groups into peptides is described by Hiskey (Peptides 3:137, 1981).
  • Fusion proteins can be assayed using standard techniques known in the art or described herein.
  • the ability of the fusion proteins to kill or inhibit growth of cells can be assayed in vitro using suitable cells, typically a cell line expressing the target or a cancer cell.
  • suitable cells typically a cell line expressing the target or a cancer cell.
  • cells of the selected test cell line are grown to an appropriate density and the candidate fusion protein is added.
  • the fusion protein can be added to the culture at around at least 1 ng/mL, at least 1 ug/mL, or at least 1 mg/mL, such as from about 0.01 ug/mL to about 1 mg/mL, from about 0.10 ug/mL to about 0.5 mg/mL, from about 1 ug/mL to about 0.4 mg/mL.
  • serial dilutions are tested.
  • cell survival or growth is assessed.
  • Methods of determining cell survival include, but are not limited to, the resazurin reduction test (see Fields & Lancaster Am. Biotechnol. Lab., 11:48-50, 1993; O'Brien et al., Eur. J. Biochem., 267:5421-5426, 2000 or U.S. Pat. No. 5,501,959), the sulforhodamine assay (Rubinstein et al., J. Natl. Cancer Inst., 82:113-118, 1999) or the neutral red dye test (Kitano et al., Euro. J. Clin.
  • Cytotoxicity is determined by comparison of cell survival in the treated culture with cell survival in one or more control cultures, for example, untreated cultures and/or cultures pre-treated with a control compound (typically a known therapeutic), or other appropriate control.
  • Cell viability can be assayed using a variety of methods, including MTT (3-(4,5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide) (Barltrop, Bioorg. & Med. Chem. Lett. 1: 611, 1991; Cory et al., Cancer Comm. 3, 207-12, 1991; Paull J. Heterocyclic Chem. 25, 911, 1988). Assays for cell viability are also available commercially.
  • MTT 3-(4,5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide
  • These assays include but are not limited to CELLTITER-GLO® Luminescent Cell Viability Assay (Promega), which uses luciferase technology to detect ATP and quantify the health or number of cells in culture, and the Cell Titer-Glo® Luminescent Cell Viability Assay, which is a lactate dehyrodgenase (LDH) cytotoxicity assay (Promega).
  • CELLTITER-GLO® Luminescent Cell Viability Assay Promega
  • LDH lactate dehyrodgenase
  • Fusion proteins that confer selectivity for a specific type of cancer may be tested for their ability to target that specific cancer cell type.
  • a fusion protein comprising a specific IL-4 that targets cells displaying IL-4R Type I or Type II can be assessed for its ability to selectively target such cells by comparing the ability of the fusion protein to kill cancer cells to its ability to kill a normal cell, or a different type of cancer cell (e.g., one that does not express IL-4R Type I or Type II).
  • flow cytometric methods as are known in the art, may be used to determine if a fusion protein comprising a Type I or Type II receptor-specific IL-4 is able to selectively target a specific type of cell. Binding of a labeled antibody to the bound fusion protein will indicate binding of the fusion protein to the target.
  • assays for measuring cell apoptosis are known in the art.
  • Apoptotic cells are characterized by characteristic morphological changes, including chromatin condensation, cell shrinkage and membrane blebbing, which can be clearly observed using light microscopy.
  • the biochemical features of apoptosis include DNA fragmentation, protein cleavage at specific locations, increased mitochondria] membrane permeability, and the appearance of phosphatidylserine on the cell membrane surface.
  • Assays for apoptosis are known in the art.
  • Exemplary assays include TUNEL (Terminal deoxynucleotidyl Transferase Biotin-dUTP Nick End Labeling) assays, caspase activity (specifically caspase-3) assays, and assays for fas-ligand and annexin V.
  • a variety of cell lines suitable for testing the candidate fusion proteins are known in the art and many are commercially available (for example, from the American Type Culture Collection, Manassas, Va.). Similarly, animal models are known in the art and many are commercially available.
  • the fusion proteins including IL-4R binding protein and a pro-apoptotic Bcl-2 family member, as described herein, can be used for a variety of therapeutic purposes.
  • the fusion proteins described herein can be used in the treatment or prophylaxis of any disease, disorder or condition which involves cells which express an IL-4R, and which would be benefited by inhibiting cell proliferation or enhancing cell death.
  • the fusion proteins described herein can be used in the treatment or prophylaxis of any disease, disorder or condition which involves cells which express a Type I or Type II IL-4R, and in which selection of one type of receptor over the other is useful, and which would be benefited by inhibiting cell proliferation or enhancing cell death.
  • a fusion protein including a pro-apoptotic Bcl-2 family member can be used to induce apoptosis or cell death or to treat a disorder associated with abnormal apoptosis or cell proliferation, such as cancer.
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • hyperproliferative and neoplastic disease states may be categorized as pathologic (e.g., as a deviation from normal but not associated with a disease state).
  • a “cancer” or “neoplasm” is meant any unwanted growth of cells serving no physiological function.
  • a cell of a neoplasm has been released from its normal cell division control, i.e., a cell whose growth is not regulated by the ordinary biochemical and physical influences in the cellular environment.
  • a neoplastic cell proliferates to form a clone of cells which are either benign or malignant.
  • cancers or neoplasms include, without limitation, transformed and immortalized cells, tumours, and carcinomas such as breast cell carcinomas and prostate carcinomas.
  • the term cancer includes cell growths that are technically benign but which carry the risk of becoming malignant.
  • malignancy is meant an abnormal growth of any cell type or tissue.
  • the term malignancy includes cell growths that are technically benign but which carry the risk of becoming malignant. This term also includes any cancer, carcinoma, neoplasm, neoplasia, or tumor. The terms are therefore meant to include all types of cancerous growths or oncogenic processes, metastatic tissue or malignantly transformed cells, tissues or organs, irrespective of hi stopathologic type or stage of invasiveness.
  • a fusion protein including a pro-apoptotic Bcl-2 family member is not used in connection with a cancer affecting a stem cell.
  • carcinomas which are the predominant cancers and are cancers of epithelial cells or cells covering the external or internal surfaces of organs, glands, or other body structures (e.g., skin, uterus, lung, breast, prostate, stomach, bowel), and which tend to metastasize; sarcomas, which are derived from connective or supportive tissue (e.g., bone, cartilage, tendons, ligaments, fat, muscle); and hematologic tumors, which are derived from bone marrow and lymphatic tissue.
  • carcinomas sarcomas, and hematopoietic neoplastic disorders e.g., leukemia.
  • Carcinomas may be adenocarcinomas (which generally develop in organs or glands capable of secretion, such as breast, lung, colon, prostate or bladder) or may be squamous cell carcinomas (which originate in the squamous epithelium and generally develop in most areas of the body).
  • adenocarcinomas which generally develop in organs or glands capable of secretion, such as breast, lung, colon, prostate or bladder
  • squamous cell carcinomas which originate in the squamous epithelium and generally develop in most areas of the body.
  • Sarcomas may be osteosarcomas or osteogenic sarcomas (bone), chondrosarcomas (cartilage), leiomyosarcomas (smooth muscle), rhabdomyosarcomas (skeletal muscle), mesothelial sarcomas or mesotheliomas (membranous lining of body cavities), fibrosarcomas (fibrous tissue), angiosarcomas or hemangioendotheliomas (blood vessels), liposarcomas (adipose tissue), gliomas or astrocytomas (neurogenic connective tissue found in the brain), myxosarcomas (primitive embryonic connective tissue), or mesenchymous or mixed mesodermal tumors (mixed connective tissue types).
  • Hematopoietic neoplastic disorders include diseases involving hyperplastic/neoplastic cells of hematopoietic origin e.g., arising from myeloid, lymphoid or erythroid lineages or precursor cells thereof.
  • the diseases arise from poorly differentiated acute leukemias (e.g., erythroblastic leukemia and acute megakaryoblastic leukemia).
  • Additional exemplary myeloid disorders include, but are not limited to, acute promyeloid leukemia (APML), acute myeloenous leukemia (AML) and chronic myeloenous leukemia (CML); lymphoid malignancies include but are not limited to acute lymphoblastic leukemia (ALL), which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia, and Waldenstrom's macroglobulinemia.
  • ALL acute lymphoblastic leukemia
  • ALL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • hairy cell leukemia and Waldenstrom's macroglobulinemia.
  • malignant lymphomas include, but are not limited to non-Hodgkin lymphoma and variants thereof, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Stemberg disases.
  • ATL adult T cell leukemia/lymphoma
  • CCL cutaneous T cell lymphoma
  • LGF large granular lymphocytic leukemia
  • Hodgkin's disease Reed-Stemberg disases.
  • Cancers may also be named based on the organ in which they originate i.e., the “primary site,” for example, cancer of the breast, brain, lung, liver, skin, prostate, testicle, bladder, colon and rectum, cervix, uterus, etc. This naming persists even if the cancer metastasizes to another part of the body, that is different from the primary site. Cancers named based on primary site may be correlated with histological classifications. For example, lung cancers are generally small cell lung cancers or non-small cell lung cancers, which may be squamous cell carcinoma, adenocarcinoma, or large cell carcinoma; skin cancers are generally basal cell cancers, squamous cell cancers, or melanomas.
  • lung cancers are generally small cell lung cancers or non-small cell lung cancers, which may be squamous cell carcinoma, adenocarcinoma, or large cell carcinoma
  • skin cancers are generally basal cell cancers, squamous cell cancer
  • Lymphomas may arise in the lymph nodes associated with the head, neck and chest, as well as in the abdominal lymph nodes or in the axillary or inguinal lymph nodes. Identification and classification of types and stages of cancers may be performed by using for example information provided by the Surveillance, Epidemiology, and End Results (SEER) Program of the National Cancer Institute.
  • a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof can be used to treat cancers such as gastric carcinoma, invasive pituitary adenomas, biliary tract carcinoma, cervical cancer, lymphoma, melanoma, chronic lymphocytic leukemia, non-hodgkins lymphoma, follicular lymphoma, pancreatic cancer, colorectal cancer, colon cancer, thyroid cancer, liver cancer, ovarian cancer, prostate cancer, bladder cancer, renal cell carcinoma, mesothelioma, rhabdomyosarcoma, breast cancer, non-small cell lung cancer, head and neck cancers, or Kaposi's carcinoma.
  • cancers such as gastric carcinoma, invasive pituitary adenomas, biliary tract carcinoma, cervical cancer, lymphoma, melanoma, chronic lymphocytic leukemia, non-hodgkins lymphoma, follicular lymphom
  • a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof can be used to treat CNS cancers such as gliomas, meningeal tumours, diffuse intrinsic pontine glioma, medulloblastoma, neuroblastoma, anaplastic astrocytoma, glioblastoma multiforme, metastatic brain cancer, or CNS lymphoma.
  • CNS cancers such as gliomas, meningeal tumours, diffuse intrinsic pontine glioma, medulloblastoma, neuroblastoma, anaplastic astrocytoma, glioblastoma multiforme, metastatic brain cancer, or CNS lymphoma.
  • the fusion proteins can be used to treat, stabilize or prevent cancer. Fusion proteins can also be used in the treatment of indolent cancers, recurrent cancers including locally recurrent, distantly recurrent and/or refractory cancers (i.e. cancers that have not responded to other anti-cancer treatments), metastatic cancers, locally advanced cancers and aggressive cancers.
  • the fusion proteins may exert either a cytotoxic or cytostatic effect resulting in, for example, a reduction in the number or growth of cancer cells, a reduction in the size of a tumor, the slowing or prevention of an increase in the size of a tumor, an increase in the disease-free survival time between the disappearance or removal of a tumor and its reappearance, prevention of an initial or subsequent occurrence of a tumor (e.g. metastasis), an increase in the time to progression, reduction of one or more adverse symptoms associated with a tumor, or an increase in the overall survival time of a subject having cancer.
  • a cytotoxic or cytostatic effect resulting in, for example, a reduction in the number or growth of cancer cells, a reduction in the size of a tumor, the slowing or prevention of an increase in the size of a tumor, an increase in the disease-free survival time between the disappearance or removal of a tumor and its reappearance, prevention of an initial or subsequent occurrence of a tumor (e.g. metastasis), an
  • proliferative and/or differentiative disorders that can be treated using a fusion protein including a pro-apoptotic Bcl-2 family member include proliferative non-malignant diseases such as pulmonary fibrosis or hyperplasia (such as benign prostatic hyperplasia), cardiac fibrosis, or liver fibrosis; inflammatory conditions such as prostatitis, vernal keratoconjunctivitis, artherosclerosis, idiopathic pulmonary pneumonia; or autoimmune conditions such as Graves disease.
  • proliferative non-malignant diseases such as pulmonary fibrosis or hyperplasia (such as benign prostatic hyperplasia), cardiac fibrosis, or liver fibrosis
  • inflammatory conditions such as prostatitis, vernal keratoconjunctivitis, artherosclerosis, idiopathic pulmonary pneumonia
  • autoimmune conditions such as Graves disease.
  • a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis.
  • the IL-4R binding protein-pro-apoptotic Bcl-2 family fusion protein is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis, when compared to a suitable control, such as IL-4 alone, IL-4 joined to a non-pro-apoptotic Bcl-2 family protein, etc.
  • a suitable control may also include a previously-established standard.
  • any test or assay for determining the activity or efficacy of an IL-4R binding protein-pro-apoptotic Bcl-2 family fusion protein may be compared to the established standard and it may not be necessary to include a control for comparison each time.
  • inhibiting cell survival is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will survive.
  • inhibitting cell proliferation is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the growth or proliferation of a cell.
  • enhancing cell death or apoptosis is meant increasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will undergo apoptotic, necrotic, or any other form of cell death.
  • a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis by at least 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more, when compared to a cell cultured under similar conditions but not contacted with the fusion protein.
  • Suitable assays for measuring the inhibition of cell survival, inhibition of cell proliferation, or enhancement of cell death or apoptosis are described herein or known in the art.
  • the IC5o of a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof, in inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis can be in the range from about 0.1 ng/mL to about 10,000 ng/mL, or any value therebetween, such as about 0.5 ng/mL, 1 ng/mL, 5 ng/mL, 10 ng/mL, 25 ng/mL, 50 ng/mL, 75 ng/mL, 100 ng/mL, 150 ng/mL, 200 ng/mL, 250 ng/mL, 300 ng/mL, 350 ng/mL, 400 ng/mL, 450 ng/mL, 500 ng/mL, 550 ng/mL, 600 ng/mL, 650 ng/mL, 700 ng/mL, 750 ng/mL, 800 ng/mL,
  • Target cells include, without limitation, neurons, lymphocytes, stem cells, epithelial cells, cancer cells, neoplasm cells, immune cells, non-malignant cells of the tumour microenvironment, hyper-proliferative cells, etc.
  • the target cell chosen will depend on the disease or injury or condition the fusion protein is intended to treat.
  • compositions according to the present disclosure can include one or more fusion proteins and one or more non-toxic, pharmaceutically-acceptable carriers, diluents, excipients and/or adjuvants. Such compositions can be suitable for use in treatment of therapeutic indications as described herein.
  • a fusion protein including a pro-apoptotic Bcl-2 family member can be administered in therapeutically-effective amounts together with one or more anti-cancer or other therapeutics.
  • the fusion protein(s) can be administered before, during or after treatment with the anti-cancer or other therapeutic.
  • An “anti-cancer therapeutic” is a compound, composition, or treatment (e.g., surgery) that prevents or delays the growth and/or metastasis of cancer cells.
  • anti-cancer therapeutics include, but are not limited to, surgery (e.g., removal of all or part of a tumor), chemotherapeutic drug treatment, radiation, gene therapy, hormonal manipulation, immunotherapy (e.g., therapeutic antibodies and cancer vaccines) and antisense or RNAi oligonucleotide therapy.
  • chemotherapeutic drugs include, but are not limited to, hydroxyurea, busulphan, cisplatin, carboplatin, chlorambucil, melphalan, cyclophosphamide, Ifosfamide, danorubicin, doxorubicin, epirubicin, mitoxantrone, vincristine, vinblastine, vinorelbine, etoposide, teniposide, paclitaxel, docetaxel, gemcitabine, cytosine, arabinoside, bleomycin, neocarcinostatin, suramin, taxol, mitomycin C, Avastin, Herceptin®, fluorouracil, temozolamide, etc.
  • the fusion protein(s) are also suitable for use with standard combination therapies employing two or more chemotherapeutic agents. It is to be understood that anti-cancer therapeutics includes novel compounds or treatments developed in the future.
  • the fusion protein can also be administered in combination with a sensitizing agent, such as a radio-sensitizer (see for example Diehn et al., J. Natl. Cancer Inst. 98:1755-7, 2006).
  • a sensitizing agent is any agent that increases the activity of a fusion protein.
  • a sensitizing agent will increase the ability of a fusion protein to inhibit cancer cell growth or kill cancer cells.
  • exemplary sensitizing agents include antibodies to IL-10, bone morphogenic proteins and HDAC inhibitors (see for example Sakariassen et al., Neoplasia 9(11):882-92, 2007). These sensitizing agents can be administered before or during treatment with the fusion protein.
  • Exemplary dosages of such sensitizing agents include at least 1 ug/mL, such as at least 10 ug/mL, at least 100 ug/mL, for example 5-100 ug/mL or 10-90 ug/mL.
  • the sensitizing agents can be administered daily, three times a week, twice a week, once a week or once every two weeks. Sensitizing agents can also be administered after treatment with the fusion protein is finished.
  • the fusion proteins may be used as part of a neo-adjuvant therapy (to primary therapy), as part of an adjuvant therapy regimen, where the intention is to cure the cancer in a subject.
  • the fusion proteins can also be administered at various stages in tumor development and progression, including in the treatment of advanced and/or aggressive neoplasias (e.g., overt disease in a subject that is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy), metastatic disease, locally advanced disease and/or refractory tumors (e.g., a cancer or tumor that has not responded to treatment).
  • Primary therapy refers to a first line of treatment upon the initial diagnosis of cancer in a subject.
  • Exemplary primary therapies may involve surgery, a wide range of chemotherapies and radiotherapy.
  • adjuvant therapy refers to a therapy that follows a primary therapy and that is administered to subjects at risk of relapsing. Adjuvant systemic therapy is begun soon after primary therapy, for example 2, 3, 4, 5, or 6 weeks after the last primary therapy treatment to delay recurrence, prolong survival or cure a subject.
  • the fusion proteins can be used alone or in combination with one or more other chemotherapeutic agents as part of an adjuvant therapy. Combinations of the fusion proteins and standard chemotherapeutics may act to improve the efficacy of the chemotherapeutic and, therefore, can be used to improve standard cancer therapies. This application can be particularly important in the treatment of drug-resistant cancers which are not responsive to standard treatment.
  • the microenvironment of a tumor contains both malignant and non-malignant cells.
  • the tumor microenvironment can be identified using one or more of the following criteria: (a) a region comprising non-malignant cells which share the same physiological environment, or which are directly adjacent to malignant cells; (b) the extended tumor region; (c) an area of inflammation surrounding or proximal to a tumor; (d) an area in which the number or rate of proliferation of regulatory T cells is elevated; and (e) an area in which tumor-associated macrophages, dendritic cells, myeloid-derived suppressor cells, Th2 cells or fibrocytes are elevated.
  • the tumor microenvironment may also be determined by the local cell-cell interactions between malignant cells and between malignant cells and any adjacent or nearby non-malignant cells. Such interactions may include, for example, cell adhesion events and/or paracrine effects of soluble mediators produced by one cell (malignant or non-malignant) on another cell (malignant or non-malignant) in the tumor microenvironment.
  • the non-malignant cells in the tumor microenvironment can be important for tumor initiation and progression (Reynolds et al., Cancer Res., 1996, 56(24):5754-5757).
  • the non-malignant cells also called stromal cells, occupy or accumulate in the same cellular space as malignant cells, or the cellular space adjacent or proximal to malignant cells, which modulate tumor cell growth or survival.
  • non-malignant cells that normally function to support inflammatory and immune response can be capable of contributing to tumor initiation or progression.
  • a fusion protein including a pro-apoptotic Bcl-2 family member can be used for inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis of a non-malignant cell that expresses a Type I or Type II IL-4R in a tumour microenvironment.
  • Such non-malignant cells can be immunoregulatory or inflammatory cells such as antigen presenting cells (e.g., macrophages, dendritic cells, B cells) or myeloid-derived suppressor cells (e.g., myeloid-derived monocytes and tie-2-expressing monocytes) present within the tumor microenvironment, and inhibition of T cell subsets that function to support tumor progression (e.g., regulatory T cells and Th2 helper cells) and/or suppressing production of one or more inflammatory cytokines in a tumor microenvironment.
  • antigen presenting cells e.g., macrophages, dendritic cells, B cells
  • myeloid-derived suppressor cells e.g., myeloid-derived monocytes and tie-2-expressing monocytes
  • non-malignant cells of a tumor microenvironment are regulatory T cells, which are observed in higher frequencies in a number of tumors, including Hodgkin's lymphoma, non-Hodgkin's lymphoma (Shi et al., Ai Zheng., 2004, 23(5):597-601 (abstract only)), malignant melanoma (Viguier et al., J. Immunol., 2004, 173(2):1444-53; Javia et al., J.
  • the regulatory T cells are recruited to the tumor site in response to chemokines secreted by the tumor cells. See e.g., Curiel et al., Nat. Med., 2004, 10:942-949. An increase in the number of regulatory T cells may also correlate with poor prognosis (Curiel et al., Nat. Med 2004, 10:942-949; Sasada et al., Cancer, 2003, 98:1089-1099). Conversely, regulatory T cells are observed to decrease following chemotherapy (Beyer et al., Blood, 2005, 106:2018-2025). The tumour micro-environment can also have a higher proportion of Th2 cells, when compared to Th1 cells, which is associated with poor prognosis and survival.
  • a fusion protein including a pro-apoptotic Bcl-2 family member according to the invention is useful for restoring the Th1>>Th2 balance by, for example, depleting Th2 cells.
  • a fusion protein including a pro-apoptotic Bcl-2 family member according to the invention, such as cpIL-4-Bad may be administered to a subject prior to, or during, chemotherapy, radiation therapy, immunotherapy, etc. to disable or downregulate the tumor microenvironment.
  • Non-malignant cells can also be fibroblasts, myofibroblasts, glial cells, epithelial cells, adipocytes, vascular cells (including blood and lymphatic vascular endothelial cells and pericytes), resident and/or recruited inflammatory and immune (e.g., macrophages, dendritic cells, myeloid suppressor cells, granulocytes, lymphocytes, etc.), resident and/or recruited cells that are capable of giving rise to or differentiating into any of the above-noted non-malignant cells, and any functionally distinct subtypes of the above-noted cells as known in the art.
  • fibroblasts e.g., myofibroblasts, glial cells, epithelial cells, adipocytes, vascular cells (including blood and lymphatic vascular endothelial cells and pericytes), resident and/or recruited inflammatory and immune (e.g., macrophages, dendritic cells, myeloid suppressor cells, granulocytes, lymphocyte
  • a “subject” can be a mammal in need of treatment, such as a human or veterinary patient (e.g., rodent, such as a mouse or rat, a cat, dog, cow, horse, sheep, goat, or other livestock).
  • a “subject” may be a clinical patient, a clinical trial volunteer, an experimental animal, etc.
  • the subject may be suspected of having or at risk for having a condition characterized by cell proliferation, be diagnosed with a condition characterized by cell proliferation, or be a control subject that is confirmed to not have a condition characterized by cell proliferation, as described herein. Diagnostic methods for conditions characterized by cell proliferation and the clinical delineation of such diagnoses are known to those of ordinary skill in the art.
  • the composition can be a liquid solution, suspension, emulsion, sustained release formulation, or powder, and can be formulated with a pharmaceutically acceptable carrier.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • pharmaceutically-acceptable carrier refers to a carrier medium or vehicle which does not interfere with the effectiveness of the biological activity of the active ingredients and which is not toxic to the host or subject.
  • Fusion proteins can be delivered along with a pharmaceutically-acceptable vehicle.
  • the vehicle may enhance the stability and/or delivery properties.
  • the disclosure also provides for formulation of the fusion protein with a suitable vehicle, such as an artificial membrane vesicle (including a liposome, noisome, nanosome and the like), microparticle or microcapsule, or as a colloidal formulation that comprises a pharmaceutically acceptable polymer.
  • a suitable vehicle such as an artificial membrane vesicle (including a liposome, noisome, nanosome and the like), microparticle or microcapsule, or as a colloidal formulation that comprises a pharmaceutically acceptable polymer.
  • the fusion protein formulations can include additives to stabilize the protein in vivo, such as human serum albumin, or other stabilizers for protein therapeutics known in the art.
  • Fusion protein formulations can also include one or more viscosity enhancing agents which act to prevent backflow of the formulation when it is administered, for example by injection or via catheter.
  • viscosity enhancing agents include, but are not limited to, biocompatible glycols and sucrose.
  • compositions containing one or more fusion proteins can be formulated as a sterile injectable aqueous or oleaginous suspension according to methods known in the art and using suitable one or more dispersing or wetting agents and/or suspending agents, such as those mentioned above.
  • the sterile injectable preparation can be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • Acceptable vehicles and solvents that can be employed include, but are not limited to, water, Ringer's solution, lactated Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils which are conventionally employed as a solvent or suspending medium
  • a variety of bland fixed oils including, for example, synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid can also be used in the preparation of injectables.
  • the fusion protein is conjugated to a water-soluble polymer, e.g., to increase stability or circulating half-life or reduce immunogenicity.
  • water-soluble polymers include, but are not limited to, polyethylene glycol (PEG), polyethylene glycol propionaldehyde, carboxymethylcellulose, dextran, polyvinyl alcohol (PVA), polyvinylpyrrolidone (PVP), polypropylene glycol homopolymers (PPG), polyoxyethylated polyols (POG) (e.g., glycerol) and other polyoxyethylated polyols, polyoxyethylated sorbitol, or polyoxyethylated glucose, and other carbohydrate polymers.
  • PEG polyethylene glycol
  • PVA polyethylene glycol propionaldehyde
  • PVP polyvinylpyrrolidone
  • PPG polypropylene glycol homopolymers
  • POG polyoxyethylated polyols
  • polymers such as PEG are described, e.g., in U.S. patent Pub. No. 20050106148 and references cited therein.
  • the polymer is a pH-sensitive polymers designed to enhance the release of drugs from the acidic endosomal compartment to the cytoplasm (see for example, Henry et al., Biomacromolecules 7(8):2407-14, 2006).
  • vaccines are prepared in an injectable form, either as a liquid solution or as a suspension.
  • Solid forms suitable for injection may also be prepared as emulsions, or with the polypeptides encapsulated in liposomes.
  • the cells are injected in any suitable carrier known in the art.
  • Suitable carriers typically comprise large macromolecules that are slowly metabolized, such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates, and inactive virus particles. Such carriers are well known to those skilled in the art. These carriers may also function as adjuvants.
  • Adjuvants are immunostimulating agents that enhance vaccine effectiveness.
  • Effective adjuvants include, but are not limited to, aluminum salts such as aluminum hydroxide and aluminum phosphate, muramyl peptides, bacterial cell wall components, saponin adjuvants, and other substances that act as immunostimulating agents to enhance the effectiveness of the composition.
  • Vaccines are administered in a manner compatible with the dose formulation.
  • an effective amount is meant a single dose, or a vaccine administered in a multiple dose schedule, that is effective for the treatment or prevention of a disease or disorder.
  • the dose is effective to inhibit the growth of a neoplasm.
  • the dose administered will vary, depending on the subject to be treated, the subject's health and physical condition, the capacity of the subject's immune system to produce antibodies, the degree of protection desired, and other relevant factors. Precise amounts of the active ingredient required will depend on the judgement of the practitioner.
  • compositions described herein include one or more fusion proteins in an amount effective to achieve the intended purpose.
  • compositions including a fusion protein containing a pro-apoptotic Bcl-2 family member are administered to a patient already suffering from a disease, disorder or condition characterized by cell proliferation, or at risk for such a disease, disorder or condition, in an amount sufficient to cure or at least partially arrest a symptom associated with cell proliferation or reduce cell growth.
  • the dosage to be administered is not subject to defined limits.
  • the dosage of the fusion protein may need to be modified or adapted for the particular purpose, for example the concentration of fusion protein needed for whole body administration may differ from that used for local administration.
  • the toxicity of the therapeutic may change depending upon the mode of administration and overall composition being used (e.g., buffer, diluent, additional chemotherapeutic, etc.).
  • an “effective amount” of a pharmaceutical composition according to the invention includes a therapeutically effective amount or a prophylactically effective amount.
  • a “therapeutically effective amount” refers to an amount of the fusion protein effective, at dosages and for periods of time necessary, that ameliorates the symptoms of the disease, disorder or condition to be treated.
  • a therapeutically effective amount of a compound may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the compound to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the fusion protein are outweighed by the therapeutically beneficial effects.
  • a therapeutically effective dose of a compound is well within the capability of those skilled in the art.
  • the therapeutically effective dose can be estimated initially either in cell culture assays, or in animal models, such as those described herein.
  • a “prophylactically effective amount” refers to to an amount of the fusion protein effective, at dosages and for periods of time necessary, that achieves the desired prophylactic result, such as delay in onset of symptoms of a neurological disorder or continued remission of a cancer.
  • Animal models can also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in other animals, including humans, using standard methods known in those of ordinary skill in the art.
  • Concentration of the fusion protein in the final formulation can be at least 0.1 mg/mL, such as at least 1 ng/mL or at least 1 ug/mL or at least 1 mg/mL.
  • concentration in the final formulation can be between about 0.01 ug/mL and about 1,000 ug/mL. In one example, the concentration in the final formulation is between about 0.01 mg/mL and about 100 mg/mL.
  • a fusion protein including an anti-apoptotic Bcl-2 family protein, or fragment thereof is administered at concentrations ranging from about 10 ng/mL to about 10,000 ng/mL, or any value therebetween, such as about 25 ng/mL, 50 ng/mL, 75 ng/mL, 100 ng/mL, 150 ng/mL, 200 ng/mL, 250 ng/mL, 300 ng/mL, 350 ng/mL, 400 ng/mL, 450 ng/mL, 500 ng/mL, 550 ng/mL, 600 ng/mL, 650 ng/mL, 700 ng/mL, 750 ng/mL, 800 ng/mL, 850 ng/mL, 900 ng/mL, 950 ng/mL, 1000 ng/mL, 1500 ng/mL, 2000 ng/mL, 2500 ng/mL, 3000 ng/mL, 3500
  • a fusion protein including an pro-apoptotic Bcl-2 family protein, or fragment thereof is administered at concentrations ranging from about 0.1 ng/mL to about 10,000 ng/mL
  • the actual amount of the compound(s) to be administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms.
  • the above dosage range is given by way of example only and is not intended to limit the scope in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing harmful side effects, for example, by first dividing the larger dose into several smaller doses for administration throughout the day.
  • the dosage will depend, among other things, upon the type of fusion protein being used and the type of disorder or condition being treated.
  • the fusion proteins according to the present disclosure contain substantially human sequences and are therefore less antigenic than, for example, immunotoxins or other molecules that contain non-human sequences.
  • the fusion proteins according to the present disclosure contain at least 80%, for example, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% human sequences.
  • the fusion proteins according to the present disclosure can be administered at substantially lower doses than for example, immunotoxins, or native IL-4R binding protein, such as IL-4 or IL-13.
  • the fusion proteins may elicit some level of antibody response when administered to a subject, which in some cases may lead to undesirable side effects. Therefore, if necessary, the antigenicity of the fusion proteins can be assessed as known in the art and/or described herein.
  • in vivo toxic effects of the fusion proteins can be evaluated by measuring their effect on animal body weight during treatment and by performing hematological profiles and liver enzyme analysis after the animal has been killed.
  • the general toxicity of the fusion proteins can be tested according to methods known in the art.
  • the overall systemic toxicity of the fusion proteins can be tested by determining the dose that kills 100% of mice (i.e. LDioo) or kills 50% of mice (i.e. LD50) following a single intravenous injection. Doses that are at least about 2, 5, or 10-fold less than the LDioo or LD50 can be selected for administration into other mammals, such as a human.
  • the kinetics and magnitude of the antibody response to the fusion proteins described herein can be determined, for example, in immunocompetent mice and can be used to facilitate the development of a dosing regimen that can be used in an immunocompetent human.
  • Immunocompetent mice such as the strain C57-BL6 are administered intravenous doses of fusion protein.
  • the mice are killed at varying intervals (e.g. following single dose, following multiple doses) and serum obtained.
  • An ELISA-based assay can be used to detect the presence of anti-fusion protein antibodies.
  • Serum samples from mice can be assessed for the presence of anti-fusion protein antibodies as known in the art.
  • epitope mapping can also be used to determine antigenicity of proteins as described in Stickler, et al., J. Immunotherapy, 23:654-660, 2000. Briefly, immune cells known as dendritic cells and CD4+ T cells are isolated from the blood of community donors who have not been exposed to the protein of interest. Small synthetic peptides spanning the length of the protein are then added to the cells in culture. Proliferation in response to the presence of a particular peptide suggests that a T cell epitope is encompassed in the sequence. This peptide sequence can subsequently be deleted or modified in the fusion protein thereby reducing its antigenicity.
  • Therapeutic efficacy and toxicity can also be determined by standard pharmaceutical procedures such as, for example, by determination of the median effective dose, or ED50 (i.e. the dose therapeutically effective in 50% of the population) and the median lethal dose, or LD50 (i.e. the dose lethal to 50% of the population).
  • the dose ratio between therapeutic and toxic effects is known as the “therapeutic index,” which can be expressed as the ratio, LD50/ED50.
  • the data obtained from cell culture assays and animal studies can be used to formulate a range of dosage for human or animal use.
  • the dosage contained in such compositions is usually within a range of concentrations that include the ED50 and demonstrate little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the subject, and the route of administration and the like.
  • Administration of the fusion proteins can be intralesionally, for instance by direct injection directly into the apoptotic tissue site; into a site that requires cell growth; into a site where a cell, tissue or organ is at risk of cell death; or into a site of hyperproliferati on or into a tumor.
  • the fusion protein can be administered systemically.
  • combination therapy comprising administration of a fusion protein in combination with a chemotherapeutic agent, the order in which the compositions are administered is interchangeable. Concomitant administration is also envisioned.
  • fusion proteins are administered systemically to patients, for example, by bolus injection or continuous infusion into a patient's bloodstream.
  • the fusion proteins may be administered locally, at the site of a tumor (intratumorally).
  • the administration can be via any route, e.g., locally, regionally, focally, systemic, convection enhanced delivery or combinations thereof.
  • the compounds When used in conjunction with one or more known chemotherapeutic agents, the compounds can be administered prior to, or after, administration of the chemotherapeutic agents, or they can be administered concomitantly.
  • the one or more chemotherapeutics may be administered systemically, for example, by bolus injection or continuous infusion, or they may be administered orally.
  • the pharmaceutical compositions can be formulated for administration by a variety of routes.
  • the compositions can be formulated for topical, rectal or parenteral administration or for administration by inhalation or spray.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrathecal, intrasternal injection or infusion techniques. Direct injection or infusion into a tumor is also contemplated. Convection enhanced delivery can also be used to administer the fusion protein.
  • the fusion protein can be injected into a subject having cancer, using an administration approach similar to the multiple injection approach of brachytherapy.
  • multiple aliquots of the purified fusion protein in the form of a pharmaceutical composition or formulation and in the appropriate dosage units may be injected using a needle.
  • Alternative methods of administration of the fusion proteins will be evident to one of ordinary skill in the art. Such methods include, for example, the use of catheters, or implantable pumps to provide continuous infusion of the fusion protein to the subject in need of therapy.
  • brachytherapy treatment planning program can be used in combination with brachytherapy treatment and ultrasound, for example, for placement of catheters for infusing fusion proteins to treat, for example, brain tumors or other localized tumors.
  • the positioning and placement of the needle can generally be achieved under ultrasound guidance.
  • the total volume, and therefore the number of injections and deposits administered to a patient can be adjusted, for example, according to the volume or area of the organ to be treated.
  • An example of a suitable software planning program is the brachytherapy treatment planning program Variseed 7.1 (Varian Medical Systems, Palo Alto, Calif.) or iPlan (BrainLab, Kunststoff, Germany) for convection enhanced delivery to the brain.
  • brachytherapy treatment planning program Variseed 7.1 (Varian Medical Systems, Palo Alto, Calif.) or iPlan (BrainLab, Kunststoff, Germany) for convection enhanced delivery to the brain.
  • Fusion proteins can be used in inhibiting cell survival or inhibiting cell proliferation in the central nervous system (CNS).
  • CNS central nervous system
  • the fusion protein When the site of delivery is the brain, the fusion protein must be capable of being delivered to the brain.
  • the blood-brain barrier limits the uptake of many therapeutic agents into the brain and spinal cord from the general circulation. Molecules which cross the blood-brain barrier use two main mechanisms: free diffusion and facilitated transport. Because of the presence of the blood-brain barrier, attaining beneficial concentrations of a given fusion protein in the CNS may require the use of specific drug delivery strategies. Delivery of fusion proteins to the CNS can be achieved by several methods.
  • fusion proteins can be delivered by direct physical introduction into the CNS, such as intraventricular, intralesional, or intrathecal injection.
  • Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Methods of introduction are also provided by rechargeable or biodegradable devices.
  • Another approach is the disruption of the blood-brain barrier by substances which increase the permeability of the blood-brain barrier. Examples include intra-arterial infusion of poorly diffusible agents such as mannitol, pharmaceuticals which increase cerebrovascular permeability such as etoposide, or vasoactive agents, such as leukotrienes or by convention enhanced delivery by catheter (CED).
  • compositions may be desirable to administer the compositions locally to the area in need of treatment; this can be achieved, for example, by local infusion during surgery, by injection, by means of a catheter, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including membranes, such as silastic membranes, or fibers.
  • a suitable membrane is Gliadel® (Eisai Inc.).
  • Non-viral approaches can also be employed for the introduction of a therapeutic to a cell requiring modulation of cell death (e.g., a cell of a patient).
  • a nucleic acid molecule can be introduced into a cell by administering the nucleic acid molecule in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:7413, 1987; Ono et al., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci.
  • nucleic acids are administered in combination with a liposome and protamine.
  • Gene transfer can also be achieved using non-viral means involving transfection in vitro. Such methods include the use of calcium phosphate, DEAE dextran, electroporation, and protoplast fusion. Liposomes can also be potentially beneficial for delivery of DNA into a cell. Transplantation of a fusion protein into the affected tissues of a patient can also be accomplished by transferring a normal nucleic acid into a cultivatable cell type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue.
  • a cultivatable cell type ex vivo e.g., an autologous or heterologous primary cell or progeny thereof
  • cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters), and regulated by any appropriate mammalian regulatory element.
  • CMV human cytomegalovirus
  • SV40 simian virus 40
  • metallothionein promoters e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters
  • enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid.
  • the enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers.
  • regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
  • This example describes making circularly permuted IL-4 proteins.
  • the coding sequence of IL-4 is designed to be reorganized creating a new amino terminus and a new carboxy terminus.
  • the site of reorganization is selected and coding regions are developed synthetically or using the native sequence as a template.
  • PCR can be used to amplify the separate coding regions and the 5′ and 3′ ends of the separate fragments are designed to overlap, thus allowing for the formation of a new coding sequence in which the newly generated peptide can for example encode a first amino acid that in the native protein may have been the 40th amino acid.
  • Specific examples of making circularly permuted ligands are provided in U.S. Pat. No. 6,011,002.
  • cpIL-4-BAD was prepared using standard techniques and tested on IL-4Ra-expressing cancer cells in vitro and on IL-4Ra positive xenograft tumors in an athymic nude mouse model, using cpIL4-PE as a reference. The results indicated that cpIL-4-BAD was effective at killing the cancer cells and at regressing the tumors.
  • cpIL-4-BAD fusion protein was prepared by transducing BL21(DE3)pLysS bacteria with chimeric plasmid cpIL-4BAD in a pET 24a expression vector ( FIG. 1 ).
  • the freshly transformed bacteria were plated on LB with kanamycin. Colonies were picked and transferred into 1 litre of superbroth enriched with glucose, MgCl2 and kanamycin and the cells were induced with 1 mM IPTG. The growth of the cells was monitored for 6 hours.
  • IL-4-BAD fusion proteins were refolded and purified using a HiTrap Column followed by two rounds of a MonoS column, after which cp1L-4-BAD fractions were evaluated by SDS-PAGE electrophoresis. This strategy yielded about 0.980 mg/L cpIL-4BAD at pH 6.5, and about 2.930 mg/L cp1L-4BAD at pH 6.8.
  • IL-4Ra positive tumor cells U 251 cells and Daudi cells
  • IL-4Ra positive tumor cells U 251 cells and Daudi cells
  • 0.1-1000 ng/mL of cpIL-4BAD was added and the plates were incubated for five days in a CO2 incubator at 37° C. On day 5, the plates were washed with 1 ⁇ PBS and the cells were detached with trypsin treatment. After trypsin inactivation, viable cells were counted using the trypan blue exclusion technique.
  • the cpIL-4BAD fusion protein killed Daudi cells in a concentration-dependant manner in vitro, with an IC50 value of about 0.1 ng/mL ( FIG. 4 ). 100 ⁇ excess of IL-4 also blocked the ability of cpIL-4BAD to kill the Daudi cells, demonstrating that the cpIL-4BAD activity was specific ( FIG. 5 ). The IC50 value rose from about 0.3 ng/mL to >1000 ng/mL in 5-day cell viability assays.
  • the cp1L-4BAD fusion protein was also found to decrease the colony numbers of IL-4Ra positive tumor cells in a colony formation assay. More specifcally, 5000 U 251 cells were plated in 10 cm culture plates (6 plates in total), cp1L-4BAD was added and the cells were incubated for 10 days. Colonies were stained with Crystal Blue and counted. The results indicated that cp1L-4BAD decreased U 251 colony numbers in a concentration dependent manner in vitro, with an IC50 value of about 5 ng/mL ( FIG. 6 ).
  • cp1L-4BAD The effectiveness of the cp1L-4BAD fusion protein was assessed in athymic mice after the development of subcutaneous glioma tumors with U 251 tumor cells. More specifically, cpIL-4BAD was injected intratumourally (IT) at 100 ⁇ g/kg (6 injections) and intraperitoneally (IP) 100 ⁇ g/kg (6 injections). 0.2% HAS/PBS was used as a vehicle control. The assessed end points were tumor size and survival. The results indicated that IT administration of cp1L-4BAD regressed the tumor growth significantly compared to the placebo control-treated animals after 6 injections. IP administration regressed the tumors dramatically compared to IT treated mice.
  • mice in both groups of mice recurred beyond day 40, when the mice in control groups were euthanized for ethical reasons, as the tumors were of 2000 mm3 on day 36 ( FIG. 7 ).
  • cpIL-4BAD-treated mice also survived longer than placebo treated mice ( FIG. 8 ).
  • IL-4Ra positive cell lines Six additional IL-4Ra positive cell lines were tested for their sensitivity to the cpIL-4BAD fusion protein and cpIL-4PE under identical experimental conditions. ICso values were determined by counting the cells using the trypan blue exclusion technique (Table 3).
  • IC 50 (ng/mL) Cell Line Origin cpIL-4BAD cpIL-4PE YCUT891 Tongue 11.0 8.0 YCUM862 Oropharynx 1.1 0.6 KCCOR891 Oral Floor 0.15 9.0 KCCL871 Larynx 0.5 1.5 YCUM911 Oropharynx 0.2 4.5 KCCTCM901 Metastasis to the chest fluid 1.0 0.7
  • IL-4BAD, cpIL-4BAD and cpS4-BAD fusion proteins were expressed and purified. More specifically, cDNAs of IL-4BAD, cpIL-4BAD and cpS4-BAD were PCR cloned into BamHI/Xhol sites of a pGWO7 E. coli . expression vector ( FIG. 9 ). The obtained vectors were verified by DNA sequencing ( FIGS. 10A-D ).
  • Protein expression was performed in E. coli cells.
  • IL-4BAD, cpIL-4BAD and cpS4-BAD proteins were expressed in 1 L cultures in insoluble form, purified under denaturing conditions using IMAC, followed by “quick dilution” protein refolding. Refolding by “quick dilution” generated intact proteins free of aggregates, as determined by non-reducing SDS-PAGE.
  • BL21(DE3)pLysS-RARE2 cells were transformed with IL-4BAD, cp1L-4BAD and cpS4-BAD protein expression constructs, plated on LB plates supplemented with Amp at 100 ⁇ g/mL, and incubated overnight at 37° C. The next day, colonies from the plate were scraped and re-suspended in liquid LB medium with 100 ⁇ g/mL of Amp. The cultures were then grown at 37° C., with aeration, and protein expression was induced by 1 mM IPTG when the cell culture reached an OD600 of about 0.5. Induction lasted for about 4 hours at 30° C. The cell pellet was then collected and stored at ⁇ 20° C.
  • the cell pellets from the induced cultures were lysed at room temperature and the inclusion bodies fraction was collected and washed with PBS-T.
  • the insoluble material was solubilized in 8M Urea and bound to 3 mL Ni-charged resin.
  • the resin was washed with 15 CV of wash buffer and the bound protein was eluted in 8 CV elutions of step gradient of imidazole in wash buffer. 7.5 ⁇ L from each fraction was analyzed on a SDS-PAGE gel.
  • the fractions with the highest amount of IL-4BAD (about 6 mg) and cp1L-4BAD (about 8 mg) were combined and refolded. The remaining fractions were stored at ⁇ 20° C.
  • IL-4BAD and cp1L-4BAD fractions were combined, reduced by the addition of 1 mM DTT and subjected to a slow step-wise dialysis against storage buffer (150 mM NaCl, 10 mM HEPES, pH 7.4, 0.01% Tween 20) supplemented with a decreasing concentration of urea at each dialysis buffer change.
  • the protein concentration was measured after each dialysis step by UV Spectroscopy: IL-4BAD was about 0.8 mg; and cp1L-4BAD was about 0.45 mg. The samples were then run on an SDS-PAGE gel.
  • each protein ( ⁇ 0.6 mg/mL, 200 mM Imidazole fraction) was diluted in 1 mL of the following buffers: 20 mM HEPES, pH 7.4, 1% glycerol, 10 ⁇ M EDTA, 0.01% Tween (Buffer 1); 10 mM Na-Phosphate, pH 7.0, 1% Glycerol, 10 ⁇ M EDTA, 0.01% Tween (Buffer 2); and PBS, pH 7.2 (Buffer 3). The samples were stored overnight at room temperature. The next day, the samples were spun for 10 minutes at 13,000 rpm and the presence of the pellet in each sample was observed and recorded in Table 2: (+++) indicates an abundant pellet; ( ⁇ ) indicates no visible pellet.
  • the samples were also analyzed on non-reducing SDS-PAGE gel. Refolded samples were concentrated to 100 ⁇ L using Amicon 10 kDa MWCO, spun down, and a 7.5 ⁇ L aliquot of each sample was run on an SDS-PAGE gel.
  • the purification process was repeated using quick dilution folding.
  • 4 ml of 200 mM imidazole fractions containing IL-4BAD, cp1L-4BAD or cpS4-BAD was quickly diluted into 200 mL of refolding buffer (500 mM NaCl, 10 mM Na-Phosphate, pH 7.0 or 6.0 or 7.8, 1% Glycerol, 10[1.M EDTA, 0.01% Tween), incubated overnight at room temperature, spun down for 20 minutes at 4,000 rpm at 4° C., concentrated to 3 mL using an Amicon 10 kDa MWCO, and buffer exchanged into storage buffer (500 mM NaCl, 10 mM Na-Phosphate, pH 7.0 or 6.0 or 7.8, 1% Glycerol, 1 ⁇ M EDTA, 0.01% Tween) using a DG-10 column.
  • refolding buffer 500 mM NaCl, 10 mM
  • Final sample concentrations were as follows: 3.5 mL of IL-4B AD at about 0.24 mg/mL; 3.5 mL of cp1L-4BAD at about 0.23 mg/mL. The final samples were run on an SDS-PAGE gel.
  • the final concentration of cpS4-BAD was about 3.5 mL at about 0.23 mg/mL.
  • the final sample was run on an SDS-PAGE gel.
  • a pKFR4-BAD-H6 fusion protein ( FIGS. 11A-B ) was prepared as follows. cDNA of pKFR4-BAD-H6 was PCR cloned into the Ndel/Xhol sites of a pET-21a(+) vector. The vector was then transformed into HMS174(DE3) cells and induced with 0.1 mM IPTG for 3 hours. Samples were run on an SDS-PAGE gel before and after induction. The cells were pelleted and lysed by ultrasonication in a buffer containing 20 mM Tris-HCl, 300 mM NaCl, 20 mM Imidazole, pH 8.0, and samples were run on an SDS-PAGE gel.
  • Inclusion bodies were dissolved in a dissolving buffer of 20 mM Tris-HCl, 300 mM NaCl, 20 mM Imidazole, 20 mM beta-ME, 7 M GuaHCl, pH 8.0. The supernatant was then purified by Ni2I affinity chromatography. pKFR4-BAD-H6 was eluted by 20 mM Tris-HCl, 300 mM NaCl, 300 mM Imidazole, 8 M Urea, pH 8.0 under reducing and non-reducing conditions. Samples were taken throughout the purification process.
  • pKFR4-BAD-H6 was dialyzed in dialysis buffer: 0.1% TFA, 30% acetonitrile under reducing conditions and non-reducing conditions, yielding a concentration of about 2.67 mg/mL.
  • EH suspension cells that express Type I IL-4R were cultured in RPMI1640 (from Gibco) containing 10% FBS (from Life Technologies), 2 mM L-glutamine (from Gibco) and 10 mM HEPES (from Gibco).
  • cpS4-BAD was added to the cell suspension.
  • the cell suspension was poured into a 96-well Isoplate in an amount of about 1 ⁇ 104 cells/well. The cell suspensions were incubated at 37° C. in a 5% CO2 incubator for 48 hours.
  • the background reading (blank) was subtracted from the readings of all the wells, and the inhibition (% inhibition) was calculated.
  • the IC50 of cpS4-BAD was about 126.3 ng/mL, which was about 3 times more potent than MDNA55 (cpIL4-PE; 362.4 ng/mL) which was used as a reference.

Abstract

The present invention relates to interleukin-4 receptor binding fusion proteins. More specifically, the invention provides, in part, fusion proteins that include an interleukin-4 receptor binding protein moiety joined to a pro-apoptotic Bcl-2 family member protein moiety.

Description

    FIELD OF INVENTION
  • The present invention relates to interleukin-4 receptor-binding protein fusions. More specifically, the invention provides, in part, fusion proteins that include an interleukin-4 or interleukin-13 protein moiety joined to a pro-apoptotic Bcl-2 family member protein moiety.
  • REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
  • This disclosure incorporates by reference the Sequence Listing text copy submitted herewith via EFS-Web, which was created on Sep. 8, 2021, entitled 117802-5002-US02_Sequence_Listing.txt which is 67,734 bytes in size.
  • BACKGROUND OF THE INVENTION
  • Interleukin-4 (IL-4) is a pleiotropic cytokine produced by activated T cells, and is the ligand for the IL-4 receptor (IL-4R), which can also bind to interleukin-13 (IL-13). IL-4, like many cytokines, first binds to a high-affinity receptor chain (designated “a”), followed by binding of the IL-4-a chain complex with a second low-affinity receptor chain designated “yc”. Therefore, the primary binding chain for IL-4 is the IL-4 receptor alpha (IL-4Ra), which binds with high affinity (KD=˜10-10 M). The IL-4/1L-4Ra complex can then bind the second component of the IL-4 receptor, yc (the “Type I” receptor) with relatively low affinity. Additionally, the IL-4/IL-4Ra complex can also bind the interleukin-13 (IL-13) receptor al (IL-13R al) (the “Type II receptor).
  • Different cell types express different amounts of the Type 1 and Type 11 receptor chains. For example, while 1L-4Ra is present on most cells, yc is generally expressed on hematopoietic cells and IL-13R al is generally expressed on non-hematopoietic cells. Accordingly, yc, but not IL-13R al, is found on T cells, natural killer (NK) cells, basophils, mast cells, and most mouse B cells (most human B cells express both yc and IL-13R al)
  • Some bone marrow-derived cells, including macrophages and dendritic cells, express both yc and IL-13R al and consequently respond to both IL-4 and IL-13. IL-13R al, but little or no yc, is found on most non-bone marrow-derived cells, including smooth muscle and epithelial cells.
  • Variant IL-4 molecules having differential selectivities for Type I and Type II receptors have been proposed (Junttila et al. Nature Chemical Biology 8:990-998, 2012.)
  • Circularly permuted molecules are those in which the termini of a linear molecule (e.g., ligand) have been joined together, either directly or via a linker, to produce a circular molecule, after which the circular molecule is opened at another location to produce a new linear molecule with termini different from the termini of the original molecule. Circularly permuted variants of IL-4 have been described in, for example, U.S. Pat. No. 6,011,002, issued Jan. 4, 2000, to Pastan el al.
  • Programmed cell death or “apoptosis,” is a common phenomenon in the development of animal cells and is both positively and negatively regulated. In addition to its involvement in neuronal and lymphoid system development and overall cell population homeostasis, apoptosis also plays a significant role in various diseases and injuries resulting from aberrant regulation of apoptotic pathways. For example, aberrant activation of neuronal cell death by apoptosis has been implicated in many neurodegenerative diseases and conditions, such as Alzheimer disease (Barinaga, Science 281:1303-1304), Huntington's disease, spinal-muscular atrophy, neuronal damage caused during stroke (reviewed in Rubin, British Med. Bulle., 53(3):617-631, 1997; and Barinaga, Science 281:1302-1303), transient ischemic neuronal injury (e.g., spinal cord injury), etc. Conversely, aberrant suppression of apoptosis can result in hyperproliferation of cells, leading to cancer and other hyperproliferative disorders.
  • Apoptosis is regulated by a number of proteins, including members of the Bcl-2 family. Bcl-2 was one of the first proteins identified as regulating apoptosis (Cleary et al., Cell 47:19-28, 1986; Tsujimoto and Croce, Proc. Natl. Acad. Sci. USA 83:5214-5218, 1986). Since its discovery, several Bcl-2-related proteins (“Bcl-2 family proteins” or “Bcl-2 family members”) have been identified as regulators of apoptosis (White, Genes Dev. 10:1-15, 1996; Yang et al., Cell 80:285-291, 1995; Lomonosova, E. and G. Chinnadurai, Oncogene 27, S2S19, 2009).
  • Several therapeutic agents for treatment of neurodegenerative diseases, cancer, etc have been explored but exhibit limitations that restrict their use in the clinic. For example, many chemotherapeutic agents act by inducing apoptosis in proliferating neoplastic cells, but their therapeutic value is limited by the extent to which they are toxic to normal cells. Treatment with standard apoptosis inhibitory molecules, for instance peptide-type caspase inhibitors (e.g., DEVD-type), has proven unsatisfactory for clinical work due to low membrane permeability of these inhibitors.
  • Targeted immunotoxins (genetic or biochemical fusions between a toxic molecule, for instance a bacterial toxin, and a targeting domain derived, typically from an antibody molecule) have been proposed in attempts to selectively eliminate cancer cells. For example, diphtheria toxin (DT) variants have been generated and tested for their ability to selectively kill cancer cells (Thorpe et al., Nature 271:752-755, 1978; Laske et al., Nature Medicine 3:1362-1368, 1997). Similarly, Pseudomonas exotoxin (PE) fusion proteins have been investigated as potential cancer therapeutics (Kreitman and Pastan, Blood 90:252-259, 1997; Shimamura et al. Cancer Res. 67:9903-9912; 2007).
  • SUMMARY OF THE INVENTION
  • The present invention relates to interleukin-4 receptor binding fusion proteins. More specifically, the invention provides, in part, fusion proteins that include a interleukin-4 receptor-binding protein moiety joined to a pro-apoptotic Bcl-2 family member protein moiety and uses thereof
  • In one aspect, the invention provides a fusion protein including a interleukin-4 (IL-4) receptor binding protein and a pro-apoptotic Bcl-2 family polypeptide. In some embodiments, the IL-4 receptor binding protein may be circularly permuted (cp). In some embodiments, the Bcl-2 family polypeptide may be a pro-apoptotic Bcl-2 family polypeptide comprising a BH3 domain (such as Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak or Bax). The BH3 domain may further include a mutation that reduces phosphorylation. The pro-apoptotic Bcl-2 family polypeptide including a BH3 domain that further includes a mutation that reduces phosphorylation may be a Bad polypeptide. The fusion protein may be capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis of a target cell expressing an IL-4R.
  • In some embodiments, the IL-4 receptor binding protein may be a mutant IL-4 or IL-13 selective for binding to a Type I or a Type TT IL-4 receptor (IL-4R) The mutant IL-4 selective for binding to a Type II IL-4R may include a KFR variant or a KF variant. The mutant IL-4 selective for binding to a Type I IL-4R may include an RGA variant. The mutant IL-13 may be an All variant or a DN variant.
  • In some embodiments, the fusion protein may further include a linker. The linker may have the sequence GS or may be a ubiquitin or ubiquitin variant molecule. The fusion protein may include the sequence set forth in SEQ ID NOs: 24-27.
  • In some aspects, there is provided a nucleic acid molecule encoding a fusion protein as described herein, or a vector including the nucleic acid molecule, or a host cell including the vector. In some aspects, there is provided a nucleic acid molecule encoding a fusion protein as set forth in SEQ ID NOs: 24-27 or comprising SEQ ID NOs: 35-38.
  • In some aspects, there is provided a pharmaceutical composition including a fusion protein as described herein, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule, or a host cell including the vector.
  • In some aspects, there is provided a method of inducing cell death by administering a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule, or a host cell including the vector, to a subject in need thereof.
  • In some aspects, there is provided a method of inducing cell death by contacting a target cell that expresses an IL-4R with a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule.
  • In some aspects, there is provided a method of treating cancer by administering: a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule, or a host cell including the vector, to a subject in need thereof.
  • In some aspects, there is provided a method of treating cancer by contacting a neoplastic cell that expresses an IL-4R with a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule.
  • In some aspects, there is provided a method of treating cancer by contacting a non-malignant cell that expresses an IL-4R in a tumour microenvironment in a subject in need thereof with a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule. In some embodiments, the non-malignant cell is contacted prior to the subject starting a therapy.
  • In some aspects, there is provided a method of treating a hyperproliferative or differentiative disorder by administering a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule to a subject in need thereof. The hyperproliferative or differentiative disorder may be a fibrosis or hyperplasia, an inflammatory conditions or an autoimmune condition. The fibrosis or hyperplasia may be pulmonary fibrosis or hyperplasia (such as benign prostatic hyperplasia), cardiac fibrosis, or liver fibrosis; the inflammatory condition may be prostatitis, vernal keratoconjunctivitis, artherosclerosis, or idiopathic pulmonary pneumonia; or the autoimmune condition may be Graves disease.
  • In some aspects, there is provided a use of a fusion protein including a pro-apoptotic Bcl-2 family polypeptide, a nucleic acid molecule encoding the fusion protein, or a vector including the nucleic acid molecule for inducing cell death or treating cancer or treating a hyperproliferative or differentiative disorder in a subject in need thereof.
  • In various embodiments of the alternative aspects, the subject may be a human.
  • This summary does not necessarily describe all features of the invention.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • These and other features of the invention will become more apparent from the following description in which reference is made to the appended drawings wherein:
  • FIG. 1 is an illustration of a cp1L-4BAD (cpIL-4g:s-BADaa) in a pET 24a expression vector;
  • FIG. 2 is a graph showing the effect of cp1L-4BAD (cpIL-4BADaa) on IL-4Ra positive tumor cell (U 251) viability;
  • FIG. 3 is a graph showing the effect of cp1L-4BAD (cpIL-4BADaa) (squares) on 1L-4Ra positive tumor cell (U 251) viability in the presence of excess IL-4 (triangles);
  • FIG. 4 is a graph showing the effect of cp1L-4BAD (cpIL-4BADaa) on IL-4Ra positive tumor cell (Daudi cell) viability;
  • FIG. 5 is a graph showing the effect of cp1L-4BAD (cp1L-4BADaa) (triangles) on IL-41ta positive tumor cell Daudi viability in the presence of excess IL-4 (squares);
  • FIG. 6 is a graph showing the effect of cp1L-4BAD (cp1L-4BADaa) on the colony number of IL-4Ra positive tumor cells (U 251);
  • FIG. 7 is a graph showing the effect of intratumoural (squares) and intraperitoneal (triangles) injection of cpIL-4BAD fusion protein in athymic mice after the development of subcutaneous glioma tumors with U 251 tumor cells (circles=controls);
  • FIG. 8 is a graph showing the survival of cpIL-4BAD-treated mice (intratumoural injection group=open diamonds, intraperitoneal injection group=triangles, controls=solid diamonds);
  • FIG. 9 shows an illustration of a pGWO7 E. coli expression vector;
  • FIGS. 10A-F show the nucleic acid (SEQ ID NOs: 29, 30 and 32) and amino sequences (SEQ ID NOs: 18, 19 and 21) of IL-4BAD (FIGS. 10A and 10B), cpIL-4BAD (FIGS. 10C and 10D) and cpS4-BAD (FIGS. 10E and 10F) fusion constructs;
  • FIGS. 11A-B show the nucleic acid (FIG. 11A; SEQ ID NO: 37) and amino sequence (FIG. 11B; SEQ ID NO: 26) of a pKFR4-BAD-H6 fusion construct.
  • DETAILED DESCRIPTION
  • The present disclosure provides, in part, a fusion protein including an IL-4R binding protein joined to a pro-apoptotic Bcl-2 family protein and uses thereof.
  • IL-4R Binding Proteins
  • IL-4R binding proteins include IL-4 and IL-13.
  • IL-4 proteins or IL-4 “protein moieties” include native IL-4 proteins, as well as variant IL-4 proteins. A “native” or “wild type” IL-4 sequence, as used herein, refers to a human IL-4 sequence, whether purified from natural sources or made using recombinant techniques, and including the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • (SEQ ID NO: 1)
    MHKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAASKNTTEKETFCRA
    ATVLRQFYSHHEKDTRCLGATAQQFH RH KQURFLKRLDR 
    NLWGLAGLNSCPVKEANQSTLENFLERLKTEVIREKYSKCSS.
  • Alternative human IL-4 sequences include the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • (SEQ ID NO: 2)
    MHKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAASKDTTEKETFCRA
    ATVLRQFYSHHEKDTRCLGATAQQFH RH KQURFLKRLDRN
    LWGLAGLNSCPVKEANQSTLENFLERLKTEVIREKYSKCSS.
  • In some embodiments, IL-4 proteins that can be used in the fusion proteins of the present disclosure are variant IL-4 proteins that have increased selectivity for yc (Type I receptor) relative to IL-13R al (Type II receptor) or vice versa as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012). In some embodiments, a variant IL-4 protein that has increased selectivity for yc (Type I receptor) is an IL-4 protein that includes the following mutations relative to the sequence of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus: R121Q/Y124W/S125F (the “RGA” or “super-4” or “S4” variant) as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012).
  • In some embodiments, a variant IL-4 protein that has increased selectivity for IL-13R al (Type II receptor) is an IL-4 protein that includes the following mutations relative to the sequence of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus: R121K/Y124F/S125R (the “KFR” or “KFR4” variant) or R121K/Y124F (the “KF” variant).
  • In some embodiments, IL-4 proteins that can be used in the fusion proteins of the present disclosure are circularly permuted (cp), as described in, for example, U.S. Pat. No. 6,011,002, issued Jan. 4, 2000, to Pastan et al. In some embodiments, a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, as follows:
  • (SEQ ID NO: 3)
    M D TTEKETFCRAATVLRQFYSHHEKDTRCLGATAQQFHRHKQURFLKRL
    DRNLWGLAGLNSCPVKEANQSTLENFLERLKTIMREKYSKCSSGGNGGH
    KCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAAS.
  • In alternative embodiments, a cp1L-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, in the context of an “RGA” or “super-4” or “S4” variant, as follows:
  • (SEQ ID NO: 4)
    M D TTEKETFCRAATVLRQFYSHHEKDTRCLGATAQQFHRHKQURFLKRL
    DRNLWGLAGLNSCPVKEANQSTLENFLERL RV IM QS K WF KC GA
    Figure US20220048965A1-20220217-P00001
    HKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAAS.
  • In alternative embodiments, a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, in the context of a “KFR” variant, as follows:
  • (SEQ ID NO: 5)
    M D TTEKETFCRAATVLRQFYSHHEKDTRCLGATAQQFHRHKQURFLKRL
    DRNLWGLAGLNSCPVKEANQSTLENFLERLKTIM K EK FR KCSS
    Figure US20220048965A1-20220217-P00002
    HKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAAS.
  • In alternative embodiments, a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 38-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-37 with a GGNGG linker and an initial methionine residue, in the context of a “KF” variant, as follows:
  • (SEQ ID NO: 6)
    M D TTEKETFCRAATVLRQFYSHHEKDTRCLGATAQQFHRHKQURFLKRL
    DRNLWGLAGLNSCPVKEANQSTLENFLERLKTIM K EK F KCSS
    Figure US20220048965A1-20220217-P00003
    HKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAAS.
  • In alternative embodiments, a cpIL-4 protein that can be used in the fusion proteins of the present disclosure includes an IL-4 protein in which residues 105-129 of native human IL-4 (e.g., SEQ ID NO: 1) or an alternative IL-4 sequence (e.g., SEQ ID NO:2), the numbering excluding the methionine at the N-terminus, are joined to residues 1-104 with a GGNGG linker and an initial methionine residue, as described in, for example, U.S. Pat. No. 6,011,002, issued Jan. 4, 2000, to Pastan et al.
  • Exemplary IL-4 proteins that can be used in the fusion proteins of the present disclosure include those described herein, as well as sequences having at least 80% sequence identity, at least 85%, at least 90%, at least 95%, at least 98% or even at least 99% sequence identity to native IL-4 (“variant IL-4 proteins”), as long as the variant IL-4 protein retains the ability to bind the IL-4 receptor, or retains increased selectivity for the yc (Type I receptor) relative to IL-13R al (Type II receptor) or vice versa as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012), or retains a desired biological activity.
  • It is to be understood that IL-4 proteins according to the present disclosure include fragments that can be smaller than the native 129 amino acid IL-4 protein, as long as the IL-4 protein fragment retains the ability to bind the IL-4 receptor, or retains increased selectivity for the yc (Type I receptor) relative to IL-13R al (Type II receptor) or vice versa as described, for example, in Junttila et al. (Nature Chemical Biology 8:990-998, 2012), or retains a desired biological activity, whether as a fragment of the native sequence, or in a cp form or fragment thereof
  • It is also to be understood that the present disclosure encompasses nucleic acid molecules that encode an IL-4 protein as described herein or known in the art, including but not limited to RNA sequences corresponding to the DNA sequences described herein.
  • Exemplary IL-4 nucleic acid molecules include:
  • (IL4; SEQ ID NO: 30)
    ATGCACAAATGCGACATTACCCTGCAAGAGATCATTAAGACCCTGAACA
    GCCTGACCGAGCAAAAGACCCTGTGTACCGAACTGACCGTCACGGACAT
    CTTCGCTGCGTCCAAGGACACTACGGAAAAGGAAACGTTCTGTCGTGCG
    GCGACGGTGCTGCGCCAGTTCTACAGCCACCATGAGAAAGATACCCGTT
    GCCTCGGTGCGACCGCGCAACAGTTCCACCGTCACAAACAGCTGATTCG
    CTTCCTGAAGCGTCTGGATCGCAACCTGTGGGGTTTGGCGGGTCTGAAC
    TCCTGTCCAGTCAAAGAAGCCAATCAGTCTACGCTGGAAAACTTTTTGG
    AGCGTCTGAAAACTATCATGCGTGAGAAGTACAGCAAATGCAGCAGC;
    (cpIL4; SEQ ID NO: 31)
    ATGGATACCACCGAGAAAGAAACGTTCTGCCGTGCTGCCACTGTCCTGC
    GCCAGTTTTACAGCCATCACGAAAAGGACACCCGTTGCCTGGGTGCGAC
    GGCGCAGCAATTCCACCGCCACAAACAGCTGATTCGTTTCCTGAAGCGT
    CTGGACCGTAACCTGTGGGGTCTGGCGGGTCTGAACAGCTGTCCAGTGA
    AAGAAGCGAATCAGAGCACCTTGGAGAATTTCCTCGAACGCCTGAAAAC
    CATCATGCGTGAGAAATACAGCAAGTGTTCTAGCGGCGGTAACGGTGGC
    CACAAATGCGATATCACCCTGCAAGAGATCATTAAGACGCTGAACTCCT
    TGACGGAACAAAAGACCCTGTGTACTGAGCTGACGGTCACCGACATTTT
    CGCGGCGTCC;
    (the cpKFR; SEQ ID NO: 32)
    ATGGATACTACCGAGAAAGAAACGTTTTGCCGTGCTGCGACCGTCCTGC
    GTCAGTTCTACAGCCACCACGAAAAGGACACCCGCTGTCTGGGTGCGAC
    TGCCCAACAATTCCATCGTCACAAACAGCTGATTCGTTTCCTGAAGCGT
    CTGGACCGCAACCTGTGGGGTCTGGCGGGCTTGAACTCCTGCCCAGTCA
    AAGAAGCGAACCAAAGCACCCTGGAAAACTTCTTGGAGCGTCTGAAAAC
    GATCATGAAAGAGAAGTTCCGCAAGTGTAGCAGCGGTGGTAATGGTGGC
    CACAAGTGCGACATTACGCTGCAGGAAATCATTAAGACCCTGAACTCTC
    TGACCGAGCAGAAAACCCTCTGTACCGAGCTGACGGTGACGGATATCTT
    TGCGGCGAGC;
    and
    (cpS4; SEQ ID NO: 33)
    ATGGATACCACCGAAAAAGAAACTTTTTGTCGTGCCGCGACTGTCCTGC
    GCCAGTTCTACAGCCACCACGAAAAGGACACCCGTTGCCTGGGTGCGAC
    CGCTCAACAATTCCATCGCCACAAACAGCTGATTCGTTTCCTGAAACGT
    CTGGATCGCAACCTGTGGGGTCTGGCGGGTTTGAACAGCTGTCCAGTCA
    AAGAAGCGAACCAGAGCACCCTGGAAAACTTTCTGGAGCGTCTGCGTGT
    TATCATGCAGAGCAAGTGGTTCAAGTGCGGTGCGGGTGGCAATGGTGGC
    CACAAGTGTGACATTACCTTGCAAGAGATTATCAAAACGCTGAACTCTC
    TGACCGAGCAAAAGACGCTGTGCACCGAGCTGACGGTGACGGACATCTT
    CGCGGCGTCC.
  • IL-13 proteins or IL-13 “protein moieties” include native IL-13 proteins, as well as variant IL-13 proteins. A “native” or “wild type” IL-13 sequence, as used herein, refers to a human IL-13 sequence, whether purified from natural sources or made using recombinant techniques, and including the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • (SEQ ID NO: 7)
    MPGPVPPSTALRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL
    ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRDTKIEVAQFVK
    DLLLHLKKLFREGQFN.
  • In some embodiments, IL-13 proteins that can be used in the fusion proteins of the present disclosure are variant IL-13 proteins that have increased selectivity for IL-13Ra1 (type II receptor) relative wild-type IL-13 protein. For example, the IL-13 variant sequence may include the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • (the ″All″ variant; SEQ ID NO: 8)
    MPGPVPPSTAVRELIEELINITQNQKAPLCNGSMVWSIN R TAGMYCAAL
    ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVR SS KIEVAQFVK
    DLL F HL R TLFREG QFN.
  • In some embodiments, a variant IL-13 protein that has increased selectivity for IL-13Ra1 (type II receptor) relative wild-type IL-13 protein is an IL-13 protein that includes the following mutations relative to the sequence of native human IL-13 (SEQ ID NO: 7), the numbering excluding the methionine at the N-terminus:
  • L 10V/E12A/V18I/R65D/D87S/T88S/L 101F/K104R/K105T (the “DN” variant). For example, the IL-13 variant sequence may include the amino acid sequence (with an additional methionine at the N-terminus) as follows:
  • (SEQ ID NO: 9)
    MPGPVPPSTA V R A LIEELINITQNQKAPLCNGSMVWSINLTAGMYCA
    ALESLINVSGCSAIEKTQ D MLSGFCPHKVSAGQF SS LHVRSSKIEVA
    QFVKDLL F HL RT LFREGQFN.
  • In some embodiments, IL-13 proteins that can be used in the fusion proteins of the present disclosure are circularly permuted (cp). In some embodiments, a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure includes an IL-13 protein in which residues 44-114 of native human IL-13 (SEQ ID NO: 7) are joined to residues 1-43 with a linker and an initial methionine residue, as follows:
  • (SEQ ID NO: 10)
    MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRDTK
    IEVAQFVKDLLLHLKKLFREGQFNGGSGPGPVPPSTALRELIEELVN
    ITQNQKAPLCNGSMVWSINLTAG.
  • In some embodiments, a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure is as follows:
  • (SEQ ID NO: 11)
    MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRDTK
    IEVAQFVKDLLLHLKKLFREGQFN
    Figure US20220048965A1-20220217-P00004
    PGPVPPSTALRELIEEL
    VNITQNQKAPLCNGSMVWSINLTAG.
  • In alternative embodiments, a variant cp1L-13 protein that can be used in the fusion proteins of the present disclosure includes an IL-13 protein in which residues 44-114 of native human IL-13 (SEQ ID NO: 7) are joined to residues 1-43 with a linker and an initial methionine residue, in the context of the “Al 1” variant, as follows:
  • (SEQ ID NO: 12)
    MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQF SS LHVRSSK
    IEVAQFVKDLL F HL RT LFREGQFN
    Figure US20220048965A1-20220217-P00005
     PVPPSTA V RELIEEL
    I NITQNQKAPLCNGSMVWSIN R TAG.
  • [In some embodiments, a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure is as follows:
  • (SEQ ID NO: 13)
    MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRSSK
    IEVAQFVKDLL F HL RT LFREGQFN
    Figure US20220048965A1-20220217-P00006
     MPGPVPPSTAVRELIEE
    LINITQNQKAPLCNGSMVWSIN R TAG.
  • In alternative embodiments, a variant cp1L-13 protein that can be used in the fusion proteins of the present disclosure includes an IL-13 protein in which residues 44-114 of native human IL-13 (SEQ ID NO: 7) are joined to residues 1-43 with a linker and an initial methionine residue, in the context of the “DN” variant, as follows:
  • (SEQ ID NO: 14)
    MYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQF SS LHVRSSK
    IEVAQFVKDLL F HL RT LFREGQFNGGSGPGPVPPSTA V RALIEELIN
    I TQNQKAPLCNGSMVWSINLTAG.
  • In some embodiments, a variant cpIL-13 protein that can be used in the fusion proteins of the present disclosure is as follows:
  • (SEQ ID NO: 15)
    MYCAALESLINVSGCSAIEKTQ D MLSGFCPHKVSAGQF SS LHVRSSK
    IEVAQFVKDLL F HL RT LFREGQFNGGSGMPGPVPPSTA V R A LIEEL I
    NITQNQKAPLCNGSMVWSINLTAG.
  • Exemplary IL-13 proteins that can be used in the fusion proteins of the present disclosure include those described herein, as well as sequences having at least 80% sequence identity, at least 85%, at least 90%, at least 95%, at least 98% or even at least 99% sequence identity to native IL-13 (“variant IL-13 proteins”), as long as the variant IL-13 protein retains the ability to bind the IL-13 receptor, or retains increased selectivity for the IL-13Ra1 (type II receptor) relative to wild-type IL-13 protein, or retains a desired biological activity.
  • It is to be understood that IL-13 proteins according to the present disclosure include fragments that can be smaller than the native 114 amino acid IL-13 protein, as long as the IL-13 protein fragment retains the ability to bind the IL-13 receptor, or retains increased selectivity for the IL-13Ra1 (type II receptor) relative to wild-type IL-13 protein, or retains a desired biological activity.
  • It is also to be understood that the present disclosure encompasses nucleic acid molecules (including but not limited to RNA sequences or DNA sequences) that encode an IL-13 protein as described herein or known in the art.
  • BCL-2 Family Proteins
  • Bcl-2-related proteins or polypeptides (“Bcl-2 family proteins” or “Bcl-2 family members”) are involved in regulation of apoptosis. Bcl-2 family proteins fall into two distinct categories: those that inhibit cell death (the “anti-apoptotic” Bcl-2 family proteins) and those that enhance cell death (the “pro-apoptotic” Bcl-2 family proteins). Bcl-2 family proteins share one to four conserved Bcl-2 homology (BH) domains, designated BH1, BH2, BH3, and BH4.
  • Pro-apoptotic Bcl-2 family proteins include those having a BH3 domain, such as Bad (e.g., Accession no: NP116784, CAG46757 or Q92934), Bik/Nbk (e.g., Accession no: CAG30276 or Q13323), Bid (e.g., Accession no: CAG28531 or P55957), Bim/Bod (e.g., Accession no: NP619527), Hrk (Accession no: 000198), Bak, or Bax. In some embodiments, pro-apoptotic Bcl-2 family proteins that can be used in the fusion proteins according to the present disclosure are mutated (for example at serine residues e.g., serine to alanine mutations) to prevent phosphorylation.
  • Bad, Bcl-2-associated agonist of cell death, is a regulator of programmed cell death (apoptosis). Bad positively regulates cell apoptosis by forming heterodimers with Bcl-xL and Bcl-2, and reversing their death repressor activity. Pro-apoptotic activity of Bad is regulated through its phosphorylation. Exemplary Bad proteins that can be used in the fusion proteins of the present disclosure include those in GenBank Accession Nos. CAG46757; AAH01901.1; and CAG46733.1, as well as those sequences provided in U.S. Pat. No. 6,737,511 (sequences herein incorporated by reference) and described herein, as well as sequences having at least 80% sequence identity, at least 85%, at least 90%, at least 95%, at least 98% or even at least 99% sequence identity to such sequences, as long as the variant retains or has enhanced biological activity of the native Bad protein. In some embodiments, a Bad protein that can be used in the fusion proteins according to the present disclosure contains serine mutations at positions 112 and/or 136 to reduce phosphorylation. In some embodiments, a Bad protein that can be used in the fusion proteins according to the present disclosure contains serine to alanine mutations at positions 112 and/or 136 to reduce phosphorylation. In some embodiments, a Bad protein that can be used in the fusion proteins according to the present disclosure includes a sequence as follows, or fragment thereof:
  • (SEQ ID NO: 16)
    FQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPGLLWDAS
    HQQEQPTSSSHEIGGAGAVEIRSRHSSYPAGTEDDEGMGEEPSPFRGR
    SRAAPPNLWAAQRYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQS
    SSWTRVFQSWWDRNLGRGSSAPSQ;
    (Accession no: NP116784; SEQ ID NO: 18)
    FQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPGLLWDAS
    HQQEQPTSSSHEIGGAGAVEIRSRHSSYPAGTEDDEGMGEEPSPFRGR
    SRSAPPNLWAAQRYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQS
    SSWTRVFQSWWDRNLGRGSSAPSQ;
    or
    (Accession no: CAG46757; SEQ ID NO: 19)
    FQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPGLLWDAS
    HQQEQPTSSSHEIGGAGAVEIRSRHSSYPAGTEDDEGMGEEPSPFRGR
    SRSAPPNLWAAQRYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQS
    SSVVTRVFQSVVVVDRNLGRGSSAPSQ.
  • In some embodiments, a Bad protein that can be used in the fusion proteins according to the present disclosure includes a variant sequence as follows, or fragment thereof:
  • (SEQ ID NO: 17)
    FQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPGLLWDAS
    HQQEQPTSSSHEIGGAGAVEIRSRHS A YPAGTEDDEGMGEEPSPFRGR
    SR A APPNLWAAQRYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQS
    SSWTRVFQSWWDRNLGRGSSAPSQ.
  • An exemplary Bik/Nbk protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows, or fragment thereof:
  • (Accession no: CAG30276; SEQ ID NO: 20)
    SEVRPLSRDILMETLLYEQLLEPPTMEVLGMTDSEEDLDPMEDFDSLE
    CMEGSDALALRLACIGDEMDVSLRAPRLAQLSEVAMHSLGLAFIYDQT
    EDIRDVLRSFMDGFTTLKENIMRFWRSPNPGSVVVSCEQVLLALLLLL
    ALLLPLLSGGLHLLLK.
  • An exemplary Bid protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows, or fragment thereof:
  • (Accession no: CAG28531; SEQ ID NO: 21)
    DCEVNNGSSLRDECITNLLVFGFLQSCSDNSFRRELDALGHELPVLAP
    QWEGYDELQTDGNRSSHSRLGRIEADSESQEDIIRNIARHLAQVGDSM
    DRSIPPGLVNGLALQLRNTSRSEEDRNRDLATALEQLLQAYPRDMEKE
    KTMLVLALLLAKKVASHTPSLLRDVFHTTVNFINQNLRTYVRSLARNG
    MD.
  • An exemplary Bim/Bod protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows:
  • (Accession no: MP619527; SEQ ID NO: 22)
    AKQPSDVSSECDREGRQLQPAERPPQLRPGAPTSLQTEPQGNPEGNHG
    GEGDSCPHGSPQGPLAPPASPGPFATRSPLFIFMRRSSLLSRSSSGYF
    SFDTDRSPAPMSCDKSTQTPSPPCQAFNHYLSAMASMRQAEPADMRPE
    IWIAQELRRIGDEFNAYYARRVFLNNYQAAEDHPRMVILRLLRYIVRL
    VWRMH.
  • An exemplary Hrk protein molecule that can be used in the fusion proteins according to the present disclosure includes a sequence as follows:
  • (Accession no: 000198; SEQ ID NO: 23)
    CPCPLHRGRGPPAVCACSAGRLGLRSSAAQLTAARLKALGDELHQRTM
    WRRRARSRRAPAPGALPTYWPWLCAAAQVAALAAWLLGRRN.
  • In some embodiments, a pro-apoptotic Bcl-2 family protein includes at least a fragment of a Bcl-2 family member, where the pro-apoptotic Bcl-2 family protein or fragment is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis. By “inhibiting cell survival” is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will survive. By “inhibiting cell proliferation” is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the growth or proliferation of a cell. By “enhancing cell death or apoptosis” is meant increasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will undergo apoptotic, necrotic, or any other form of cell death. Suitable assays for measuring the inhibition of cell survival, inhibition of cell proliferation, or enhancement of cell death or apoptosis are described herein or known in the art.
  • It is also to be understood that the present disclosure encompasses nucleic acid molecules (e.g., RNA sequences or DNA sequences) that encode a pro-apoptotic Bcl-2 family member as described herein, including but not limited to RNA sequences corresponding to the DNA sequences described herein.
  • An exemplary pro-apoptotic Bcl-2 family member nucleic acid molecule includes:
  • (variant BAD; SEQ ID NO: 34)
    GGTAGCTTTCAGATCCCGGAATTTGAGCCGAGCGAGCAAGAGGATTCA
    AGCAGCGCGGAGCGCGGTCTGGGTCCGAGCCCGGCAGGCGACGGTCCG
    AGCGGCAGCGGCAAGCATCACCGCCAGGCGCCAGGCCTGCTGTGGGAT
    GCATCGCATCAACAGGAACAACCGACGAGCAGCAGCCATCATGGTGGC
    GCTGGTGCGGTTGAGATTAGATCGCGCCACTCCGCATATCCTGCCGGC
    ACCGAAGATGACGAAGGCATGGGCGAGGAACCGAGCCCGTTCCGTGGC
    CGTAGCCGTGCTGCACCGCCGAATCTGTGGGCCGCACAGCGTTATGGT
    CGCGAGTTGCGTCGCATGTCCGACGAGTTTGTTGACTCCTTCAAGAAA
    GGTTTACCGCGTCCGAAATCTGCCGGTACCGCGACGCAGATGCGTCAG
    AGCAGCAGCTGGACCCGCGTGTTTCAATCTTGGTGGGATCGTAATCTG
    GGTCGTGGTAGCAGCGCACCGAGCCAA.
  • IL-4 Receptor Binding Protein-Bcl-2 Family Fusion Proteins
  • “Fusion proteins” according to the present disclosure include IL-4R binding proteins, such as IL-4 and IL-13, joined to a pro-apoptotic Bcl-2 family member, with optional additional sequences or moieties (such as linkers), as described herein, as well as nucleic acid molecules encoding such fusion proteins. Also encompassed are recombinant nucleic acid molecules in which a nucleic acid sequence encoding a fusion protein is operably linked to a promoter, vectors containing such a molecule, and transgenic cells comprising such a molecule.
  • IL-4 (including cp1L-4 and IL-4 fragments and variants) can be linked to pro-apoptotic Bcl-2 family polypeptides comprising a BH3 domain as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak, or Bax or combinations thereof, or fragments or variants thereof, as long as pro-apoptotic activity is retained. Any form or derivative of IL-4 can be used. For example, IL-4 or fragments of IL-4 that bind to the IL-4 receptor can be used. Additionally, multiple pro-apoptotic Bcl-2 family proteins or fragments or variants thereof can be joined to 1L-4 or fragments or variants thereof or multiple 1L-4 proteins or fragments or variants thereof can be joined to pro-apoptotic Bcl-2 family proteins or fragments or variants thereof.
  • IL-13 (including IL-13 fragments or variants) can be linked to pro-apoptotic Bcl-2 family polypeptides, for example those comprising a BH3 domain, as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, or Hrk, or combinations thereof, as long as the combination or fragments or variants thereof retains pro-apoptotic activity. Any form or derivative of IL-13 can be used. For example, IL-13 or fragments of IL-13 that bind to the IL-13 receptor can be used. Additionally, multiple pro-apoptotic Bcl-2 family proteins or fragments or variants thereof can be joined to IL-13 or fragments or variants thereof or multiple IL-13 proteins or fragments or variants thereof can be joined to pro-apoptotic Bcl-2 family proteins or fragments or variants thereof.
  • A cp1L-4, can be linked to pro-apoptotic Bcl-2 family polypeptides, such as those comprising a BH3 domain as exemplified by Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak, or Bax or combinations thereof, or fragments or variants thereof, as long as pro-apoptotic activity is retained. Any form or derivative of cpIL-4 can be used. Additionally, multiple cpIL-4 proteins or fragments or variants thereof, can be joined to a pro-apoptotic Bcl-2 family protein or fragments or variants thereof, or multiple pro-apoptotic Bcl-2 family proteins or fragments or variants thereof, can be joined to cp1L-4 proteins or fragments or variants thereof.
  • Exemplary fusion proteins are listed in Table 1.
  • TABLE 1
    IL-4/Bcl-2 Family Fusion Proteins
    Circularly permuted Bcl-2 Family
    Name IL-4 Linker Protein Description
    IL4- MHKCDITLQEIIKTLN GS FQIPEFEPSEQED Human IL-4
    Bad SLTEQKTLCTELTVT SSSAERGLGPSP fused to human
    DIFAASK D TTEKETF AGDGPSGSGKHH Bad variant
    CRAATVLRQFYSHH RQAPGLLWDASH via a GS
    EKDTRCLGATAQQF QQEQPTSSSHHG linker
    HRHKQLIRFLKRLDR GAGAVEIRSRHSA
    NLWGLAGLNSCPVK YPAGTEDDEGMG
    EANQSTLENFLERLK EEPSPFRGRSRA
    TIIVIREKYSKCSS APPNLWAAQRYG
    (SEQ ID NO: 2). RELRRMSDEFVD
    SFKKGLPRPKSAG
    TATQMRQSSSVVT
    RVFQSVWVDRNL
    GRGSSAPSQ
    (SEQ ID NO: 17).
    Fusion Amino Acid Sequence:
    MHKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAASKDTTEKETFCRAATVLRQFYSHHEK
    DTRCLGATAQQFHRHKQURFLKRLDRNLWGLAGLNSCPVKEANQSTLENFLERLKTIM
    REKYSKCSSFQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPGLLWDASH
    QQEQPTSSSHEGGAGAVEIRSRHS A YPAGTEDDEGMGEEPSPFRGRSR A APPNLWAA
    QRYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQSSSWTRVFQSVWVDRNLGRGS
    SAPSQ (SEQ ID NO: 24).
    Fusion DNA Sequence:
    ATGCACAAATGCGACATTACCCTGCAAGAGATCATTAAGACCCTGAACAGCCTGACC
    GAGCAAAAGACCCTGTGTACCGAACTGACCGTCACGGACATCTTCGCTGCGTCCAA
    GGACACTACGGAAAAGGAAACGTTCTGTCGTGCGGCGACGGTGCTGCGCCAGTTCT
    ACAGCCACCATGAGAAAGATACCCGTTGCCTCGGTGCGACCGCGCAACAGTTCCAC
    CGTCACAAACAGCTGATTCGCTTCCTGAAGCGTCTGGATCGCAACCTGTGGGGTTTG
    GCGGGTCTGAACTCCTGTCCAGTCAAAGAAGCCAATCAGTCTACGCTGGAAAACTTT
    TTGGAGCGTCTGAAAACTATCATGCGTGAGAAGTACAGCAAATGCAGCAGCGGTAG
    CTTTCAGATCCCGGAATTTGAGCCGAGCGAGCAAGAGGATTCAAGCAGCGCGGAGC
    GCGGTCTGGGTCCGAGCCCGGCAGGCGACGGTCCGAGCGGCAGCGGCAAGCATC
    ACCGCCAGGCGCCAGGCCTGCTGTGGGATGCATCGCATCAACAGGAACAACCGAC
    GAGCAGCAGCCATCATGGTGGCGCTGGTGCGGTTGAGATTAGATCGCGCCACTCCG
    CATATCCTGCCGGCACCGAAGATGACGAAGGCATGGGCGAGGAACCGAGCCCGTT
    CCGTGGCCGTAGCCGTGCTGCACCGCCGAATCTGTGGGCCGCACAGCGTTATGGT
    CGCGAGTTGCGTCGCATGTCCGACGAGTTTGTTGACTCCTTCAAGAAAGGTTTACCG
    CGTCCGAAATCTGCCGGTACCGCGACGCAGATGCGTCAGAGCAGCAGCTGGACCC
    GCGTGTTTCAATCTTGGTGGGATCGTAATCTGGGTCGTGGTAGCAGCGCACCGAGC
    CAA (SEQ ID NO: 35).
    cpl L4-Bad M D TTEKETFCRAAT GS FQIPEFEPSEQED Circularly
    V SSSAERGLGPSP permuted
    LRQFYSHHEKDTRC AGDGPSGSGKHH human IL-4
    LGATAQQFHRHKQLI RQAPGLLWDASH fused to
    RFLKRLDRNLWGLA QQEQPTSSSHHG human
    GLNSCPVKEANQSTL GAGAVEIRSRHS A Bad variant
    ENFLERLKTIMREK YPAGTEDDEGMG via a GS
    YSKCSS
    Figure US20220048965A1-20220217-P00007
    HKC
    EEPSPFRGRSR A linker
    DITLQEIIKTLNSLTE APPNLWAAQRYG
    QKTLCTELTVTDI RELRRMSDEFVD
    FAAS (SEQ ID NO: 3). SFKKGLPRPKSAG
    TATQMRQSSSWT
    RVFQSVWVDRN
    LGRGSSAPSQ (SEQ
    ID NO: 17).
    Fusion Amino Acid Sequence:
    M D TTEKETFCRAATVLRQFYSHHEKDTRCLGATAQQFHRHKWRFLKRLDRNLWGLAGL
    NSCPVKEANQSTLENFLERLKTIMREKYSKCSS
    Figure US20220048965A1-20220217-P00008
    HKCDITLQEllKTLNSLTEQKTLC
    TELTVTDIFAASGSFQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPGLLWDAS
    HQQEQPTSSSHEGGAGAVEIRSRHS A YPAGTEDDEGMGEEPSPFRGRSR A APPNLWAAQ
    RYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQSSSVVTRVFQSVWVDRNLGRGSSA
    PSQ (SEQ ID NO: 25).
    Fusion DNA Sequence:
    ATGGATACCACCGAGAAAGAAACGTTCTGCCGTGCTGCCACTGTCCTGCGCCAGTTTT
    ACAGCCATCACGAAAAGGACACCCGTTGCCTGGGTGCGACGGCGCAGCAATTCCACC
    GCCACAAACAGCTGATTCGTTTCCTGAAGCGTCTGGACCGTAACCTGTGGGGTCTGGC
    GGGTCTGAACAGCTGTCCAGTGAAAGAAGCGAATCAGAGCACCTTGGAGAATTTCCT
    CGAACGCCTGAAAACCATCATGCGTGAGAAATACAGCAAGTGTTCTAGCGGCGGTAA
    CGGTGGCCACAAATGCGATATCACCCTGCAAGAGATCATTAAGACGCTGAACTAGCT
    GGACCCGCGTGTTTCAATCTTGGTGGGATCGTAATCTGGGTCGTGGTAGCAG
    CGCACCGAGCCAA (SEQ ID NO: 37).
    cpS4-Bad M D TTEKETFCRAAT GS FQIPEFEPSEQE Circularly
    V DSSSAERGLGPS permuted
    LRQFYSHHEKDTRCL PAGDGPSGSGKHH RGA
    GATAQQFHRHKQLIR RQAPGLLWD (Super-4)
    FLKRLDRNLWGLAG ASHQQEQPTSSS variant of
    L HHGGAGAVEIRS IL-4 fused
    NSCPVKEANQSTLEN RHS A YPAGTEDD to pro-
    FLERL RV IMG K WFK EGMGEEPSPFR apoptotic
    CGA
    Figure US20220048965A1-20220217-P00009
    HKCDIT
    GRSR A APPNLW human Bad
    L AAQRYGRELRR with GS
    QEIIKTLNSLTEQKTL MSDEFVDSFKKG linker;
    CTELTVTDIFAAS LPRPKSAGTATQMR Mutations in
    (SEQ ID NO: 4). QSSSVVTRVF S75A and
    QSWVVDRNLGRGS S99A of
    SAPSQ(SEQ ID NO: Bad
    17).
    Fusion Amino Acid Sequence:
    M D TTEKETFCRAATVLRQFYSHHEKDTRCLGATAQQFHRHKQLIRFLKRLDRNLWGLAG
    LNSCPVKEANQSTLENFLERL RV IM QS K WF KC GA
    Figure US20220048965A1-20220217-P00010
    HKCDITLQEIIKTLNSLTEQK
    TLCTELTVTDIFAASGSFQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQAPG
    LLWDASHQQEQPTSSSHHGGAGAVEIRSRHSAYPAGTEDDEGMGEEPSPFRGRSR A A
    PPNLWAAQRYGRELRRMSDEFVDSFKKGLPRPKSAGTATQMRQSSSWTRVFQSVWVD
    RNLGRGSSAPSQ (SEQ ID NO: 27).
    Fusion DNA Sequence:
    ATGGATACCACCGAAAAAGAAACTTTTTGTCGTGCCGCGACTGTCCTGCGCCAGTTCT
    ACAGCCACCACGAAAAGGACACCCGTTGCCTGGGTGCGACCGCTCAACAATTCCATC
    GCCACAAACAGCTGATTCGTTTCCTGAAACGTCTGGATCGCAACCTGTGGGGTCTGGC
    GGGTTTGAACAGCTGTCCAGTCAAAGAAGCGAACCAGAGCACCCTGGAAAACTTTCT
    GGAGCGTCTGCGTGTTATCATGCAGAGCAAGTGGTTCAAGTGCGGTGCGGGTGGCAA
    TGGTGGCCACAAGTGTGACATTACCTTGCAAGAGATTATCAAAACGCTGAACTCTCTG
    ACCGAGCAAAAGACGCTGTGCACCGAGCTGACGGTGACGGACATCTTCGCGGCGTCC
    GGTAGCTTTCAGATCCCGGAATTTGAGCCGAGCGAGCAAGAGGATTCAAGCAGCGCG
    GAGCGCGGTCTGGGTCCGAGCCCGGCAGGCGACGGTCCGAGCGGCAGCGGCAAGCA
    TCACCGCCAGGCGCCAGGCCTGCTGTGGGATGCATCGCATCAACAGGAACAACCGAC
    GAGCAGCAGCCATCATGGTGGCGCTGGTGCGGTTGAGATTAGATCGCGCCACTCCGC
    ATATCCTGCCGGCACCGAAGATGACGAAGGCATGGGCGAGGAACCGAGCCCGTTCCG
    TGGCCGTAGCCGTGCTGCACCGCCGAATCTGTGGGCCGCACAGCGTTATGGTCGCGA
    GTTGCGTCGCATGTCCGACGAGTTTGTTGACTCCTTCAAGAAAGGTTTACCGCGTCCG
    AAATCTGCCGGTACCGCGACGCAGATGCGTCAGAGCAGCAGCTGGACCCGCGTGTTT
    CAATCTTGGTGGGATCGTAATCTGGGTCGTGGTAGCAGCGCACCGAGCCAA
    (SEQ ID NO: 38).
  • The joining or “fusion” of an IL-4R binding protein, such as IL-4 or IL-13, to a pro-apoptotic Bcl-2 family member may be direct, such that one portion of the IL-4R binding protein is directly attached to a portion of the pro-apoptotic Bcl-2 family member. For example, one end of the amino acid sequence of an IL-4R binding protein can be directly attached to an end of the amino acid sequence of the pro-apoptotic Bcl-2 family member. For example, the C-terminus of the IL-4R binding protein can be linked to the N-terminus of the pro-apoptotic Bcl-2 family member, or the C-terminus of the pro-apoptotic Bcl-2 family member can be linked to the N-terminus of the IL-4R binding protein. Methods of generating such fusion proteins are routine in the art, for example using recombinant molecular biology methods.
  • Linkers
  • In some embodiments, an IL-4R binding protein moiety can be linked to the pro-apoptotic Bcl-2 family member moiety indirectly through a linker. The linker can serve, for example, simply as a convenient way to link the two moieties, as a means to spatially separate the two moieties, to provide an additional functionality to the IL-4R binding protein or the pro-apoptotic Bcl-2 family member, or a combination thereof
  • In general, the linker joining the IL-4R binding protein moiety and the pro-apoptotic Bcl-2 family member moiety can be designed to (1) allow the two molecules to fold and act independently of each other, (2) not have a propensity for developing an ordered secondary structure which could interfere with the functional domains of the two moieties, (3) have minimal hydrophobic or charged characteristics which could interact with the functional protein domains and/or (4) provide steric separation of the two regions. For example, in some instances, it may be desirable to spatially separate the IL-4R binding protein and the pro-apoptotic Bcl-2 family member to prevent the IL-4R binding protein from interfering with the activity of the pro-apoptotic Bcl-2 family member and/or the pro-apoptotic Bcl-2 family member interfering with the activity of the IL-4R binding protein. The linker can also be used to provide, for example, lability to the connection between the IL-4R binding protein and the pro-apoptotic Bcl-2 family member, an enzyme cleavage site (for example, a cleavage site for a protease), a stability sequence, a molecular tag, a detectable label, or various combinations thereof. In some embodiments, a linker can be present between two domains of an IL-4R binding protein (such as in a cp molecule) or pro-apoptotic Bcl-2 family member.
  • The linker can be bifunctional or polyfunctional, i.e., contain at least about a first reactive functionality at, or proximal to, a first end of the linker that is capable of bonding to, or being modified to bond to, the IL-4R binding protein and a second reactive functionality at, or proximal to, the opposite end of the linker that is capable of bonding to, or being modified to bond to, the pro-apoptotic Bcl-2 family member being modified. The two or more reactive functionalities can be the same (i.e. the linker is homobifunctional) or they can be different (i.e. the linker is heterobifunctional).
  • The length and composition of a linker can be varied considerably. The length and composition of the linker are generally selected taking into consideration the intended function of the linker, and optionally other factors such as ease of synthesis, stability, resistance to certain chemical and/or temperature parameters, and biocompatibility. For example, the linker should not significantly interfere with the activity of the IL-4R binding protein and/or pro-apoptotic Bcl-2 family member.
  • Linkers suitable for use in a fusion protein according to the present disclosure include peptides. The linker can be attached to the IL-4R binding moiety and/or the pro-apoptotic Bcl-2 family member moiety using recombinant DNA technology. Such methods are well-known in the art and details of this technology can be found, for example, in Sambrook, et al. Molecular Cloning: A Laboratory Manual. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989 or Ausubel et al. Current Protocols in Molecular Biology, John Wiley & Sons, 1994) or updates thereto.
  • The linker peptides can have a chain length of 1 to 500 amino acid residues (such as 1 to 100, 1 to 50, 6 to 30, 1 to 40, 1 to 20, or less than 30 amino acids or 5 to 10 amino acids). In some embodiments, a linker can be 2, 3, 4, 5, 6, 7, or 8 amino acids in length, or can be about 10, 20, 30, 40 or 50 amino acids in length.
  • Typically, surface amino acids in flexible protein regions include Gly, Asn and Ser, and such amino acids can be used in linker sequences. Other neutral amino acids, such as Thr and Ala, can also be used in the linker sequence. Additional amino acids can be included in the linker to provide unique restriction sites in the linker sequence to facilitate construction of the fusions. In some embodiments, a linker may for instance include the amino acid sequence Gly-Ser (GS) or may be the amino acid sequence Gly-Ser (GS) or may include a ubiquitin sequence:
  • GGGSM QI FVRTLTGRTITLEVEPSDTI EN VRAR I QDREGI PPDQQRLI FAGRQLEDGRTLS DYNIQRESTLHLVLRLRGGGS (SEQ ID NO: 28) or variant thereof. Ubiquitin molecules suitable for use as linkers are described in, for example, Bachran, C. el al. “Anthrax toxin-mediated delivery of the Pseudomonas exotoxin A enzymatic domain to the cytosol of tumor cells via cleavable ubiquitin fusions MBio. 2013 Apr. 30; 4(3):e00201-13, or in PCT publication WO/2012/139112.
  • Peptide linkers that are susceptible to cleavage by enzymes of the complement system, urokinase, tissue plasminogen activator, trypsin, plasmin, or another enzyme having proteolytic activity may be used in one example. According to another example, the IL-4R binding protein can be attached via a linker susceptible to cleavage by enzymes having a proteolytic activity such as a urokinase, a tissue plasminogen activator, plasmin, thrombin or trypsin. In addition, the IL-4R binding protein can be attached to the pro-apoptotic Bcl-2 family member via disulfide bonds (for example, the disulfide bonds on a cysteine molecule). For example, in the context of pro-apoptotic Bcl-2 family proteins, since many tumors naturally release high levels of glutathione (a reducing agent) this can reduce the disulfide bonds with subsequent release of the pro-apoptotic Bcl-2 family member at the site of delivery.
  • In some embodiments, a fusion protein according to the present disclosure may include a pro-apoptotic Bcl-2 family member and an IL-4R binding protein linked by a cleavable linker region. In another embodiment, the cleavable linker region can be a protease-cleavable linker, although other linkers, cleavable for example by small molecules, may be used. Examples of protease cleavage sites include those cleaved by factor Xa, thrombin and collagenase. In one example, the protease cleavage site include those cleaved by a protease that is associated with a disease. In another example, the protease cleavage site is one that is cleaved by a protease that is up-regulated or associated with cancers in general. Examples of such proteases are uPA, the matrix metalloproteinase (MiMP) family, the caspases, elastase, prostate specific antigen (PSA, a serine protease), and the plasminogen activator family, as well as fibroblast activation protein. In still another example, the cleavage site is cleaved by a protease secreted by cancer-associated cells. Examples of these proteases include matrixmetalloproteases, elastase, plasmin, thrombin, and uPA. In another example, the protease cleavage site is one that is up-regulated or associated with a specific cancer. The precise sequences are available in the art and the skilled person will have no difficulty in selecting a suitable cleavage site. By way of example, the protease cleavage region targeted by Factor Xa is IEGR. The protease cleavage region targeted by enterokinase is DDDDK. The protease cleavage region targeted by thrombin is LVPRG. In one example, the cleavable linker region is one which is targeted by endocellular proteases.
  • The linker can be attached to the IL-4R binding protein moiety and/or pro-apoptotic Bcl-2 family member moiety using routine techniques as known in the art.
  • Preparation of IL-4R Binding Protein/Pro-Apoptotic Bcl-2 Family Fusion Proteins
  • Fusion proteins can be prepared using routine methods as known in the art. Fusion proteins, as well as modifications thereto, can be made, for example, by engineering the nucleic acid encoding the fusion protein using recombinant DNA technology or by peptide synthesis. Modifications to the fusion protein may be made, for example, by modifying the fusion protein polypeptide itself, using chemical modifications and/or limited proteolysis. Combinations of these methods may also be used to prepare the fusion proteins.
  • Methods of cloning and expressing proteins are well-known in the art, detailed descriptions of techniques and systems for the expression of recombinant proteins can be found, for example, in Current Protocols in Protein Science (Coligan, J. E., et al., Wiley & Sons, New York). Those skilled in the art will understand that a wide variety of expression systems can be used to provide the recombinant protein. Accordingly, the fusion proteins can be produced in a prokaryotic host (e.g., E. coli, A. salmonicida or B. subtilis) or in a eukaryotic host (e.g., Saccharomyces or Pichia; mammalian cells, e.g., COS, NIH 3T3, CHO, BHK, 293, or HeLa cells; or insect cells (baculovirus)). The fusion proteins can be purified from the host cells using standard techniques known in the art.
  • Sequences for various exemplary fusion proteins are provided in Table 1. Variants and homologs of these sequences can be cloned, if an alternative sequence is desired, using standard techniques (see, for example, Ausubel et al., Current Protocols in Molecular Biology, Wiley & Sons, NY (1997 and updates); Sambrook et al., Sambrook, et al. Molecular Cloning: A Laboratory Manual. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989 or updates thereto). For example, the nucleic acid sequence can be obtained directly from a suitable organism, such as Aeromonas hydrophila, by extracting mRNA and then synthesizing cDNA from the mRNA template (for example by RT-PCR) or by PCR-amplifying the gene from genomic DNA. Alternatively, the nucleic acid sequence encoding either the IL-4R binding moiety or the pro-apoptotic Bcl-2 family moiety can be obtained from an appropriate cDNA library by standard procedures. The isolated cDNA is then inserted into a suitable vector, such as a cloning vector or an expression vector.
  • Mutations (if desired) can be introduced at specific, pre-selected locations by in vitro site-directed mutagenesis techniques well-known in the art. Mutations can be introduced by deletion, insertion, substitution, inversion, or a combination thereof, of one or more of the appropriate nucleotides making up the coding sequence.
  • The expression vector can further include regulatory elements, such as transcriptional elements, required for efficient transcription of the fusion protein-encoding sequences. Examples of regulatory elements that can be incorporated into the vector include, but are not limited to, promoters, enhancers, terminators, and polyadenylation signals. Vectors that include a regulatory element operatively linked to a nucleic acid sequence encoding a genetically engineered fusion protein can be used to produce the fusion protein.
  • The expression vector may additionally contain heterologous nucleic acid sequences that facilitate the purification of the expressed fusion protein, such as affinity tags such (e.g., metal-affinity tags, histidine tags, avidin/streptavidin encoding sequences, glutathione-S-transferase (GST) encoding sequences, maltose binding protein (MBP) encoding sequences or biotin encoding sequences). In one example, such tags are attached to the N- or C-terminus of a fusion protein, or can be located within the fusion protein. The tags can be removed from the expressed fusion protein prior to use according to methods known in the art. Alternatively, the tags can be retained on the fusion protein, providing that they do not interfere with the ability of the desired activity of the fusion protein.
  • The fusion protein can include one or more linkers, as well as other moieties, as desired and/or as discussed herein. These can include a binding region, such as avidin or an epitope, or a tag such as a polyhistidine tag, which can be useful for purification and processing of the fusion protein, as well as other linkers as described herein. In addition, detectable markers can be attached to the fusion protein, so that the traffic of the fusion protein through a body or cell can be monitored conveniently. Such markers include radionuclides, enzymes, fluorophores, chromophores, and the like.
  • One of ordinary skill in the art will appreciate that the DNA can be altered in numerous ways without affecting the biological activity of the encoded protein. For example, PCR can be used to produce variations in the DNA sequence which encodes a fusion protein. Such variations in the DNA sequence encoding a fusion protein can be used to optimize for codon preference in a host cell used to express the protein, or may contain other sequence changes that facilitate expression.
  • A covalent linkage of an IL-4R binding protein directly to a pro-apoptotic Bcl-2 family member or via a linker may take various forms as is known in the art. For example, the covalent linkage may be in the form of a disulfide bond. The DNA encoding one of the components can be engineered to contain a unique cysteine codon. The second component can be derivatized with a sulfhydryl group reactive with the cysteine of the first component. Alternatively, a sulfhydryl group, either by itself or as part of a cysteine residue, can be introduced using solid phase polypeptide techniques. For example, the introduction of sulfhydryl groups into peptides is described by Hiskey (Peptides 3:137, 1981).
  • Assays
  • Fusion proteins can be assayed using standard techniques known in the art or described herein.
  • For example, the ability of the fusion proteins to kill or inhibit growth of cells can be assayed in vitro using suitable cells, typically a cell line expressing the target or a cancer cell. In general, cells of the selected test cell line are grown to an appropriate density and the candidate fusion protein is added. The fusion protein can be added to the culture at around at least 1 ng/mL, at least 1 ug/mL, or at least 1 mg/mL, such as from about 0.01 ug/mL to about 1 mg/mL, from about 0.10 ug/mL to about 0.5 mg/mL, from about 1 ug/mL to about 0.4 mg/mL. In some examples, serial dilutions are tested. After an appropriate incubation time (for example, about 48 to 72 hours), cell survival or growth is assessed. Methods of determining cell survival are well known in the art and include, but are not limited to, the resazurin reduction test (see Fields & Lancaster Am. Biotechnol. Lab., 11:48-50, 1993; O'Brien et al., Eur. J. Biochem., 267:5421-5426, 2000 or U.S. Pat. No. 5,501,959), the sulforhodamine assay (Rubinstein et al., J. Natl. Cancer Inst., 82:113-118, 1999) or the neutral red dye test (Kitano et al., Euro. J. Clin. Investg., 21:53-58, 1991; West et al., J. Investigative Derm., 99:95-100, 1992) or trypan blue assay. Numerous commercially available kits may also be used, for example the CellTiter 96®AQueous One Solution Cell Proliferation Assay (Promega). Cytotoxicity is determined by comparison of cell survival in the treated culture with cell survival in one or more control cultures, for example, untreated cultures and/or cultures pre-treated with a control compound (typically a known therapeutic), or other appropriate control.
  • Additional assays are described in, for example, Crouch et al. (J. Immunol. Meth. 160, 81-8); Kangas et al. (Med. Biol. 62, 338-43, 1984); Lundin et al., (Meth. Enzymol. 133, 27-42, 1986); Petty et al. (Comparison of J. Biolum. Chemilum. 10, 29-34, 1995); and Cree et al. (AntiCancer Drugs 6: 398-404, 1995). Cell viability can be assayed using a variety of methods, including MTT (3-(4,5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide) (Barltrop, Bioorg. & Med. Chem. Lett. 1: 611, 1991; Cory et al., Cancer Comm. 3, 207-12, 1991; Paull J. Heterocyclic Chem. 25, 911, 1988). Assays for cell viability are also available commercially. These assays include but are not limited to CELLTITER-GLO® Luminescent Cell Viability Assay (Promega), which uses luciferase technology to detect ATP and quantify the health or number of cells in culture, and the Cell Titer-Glo® Luminescent Cell Viability Assay, which is a lactate dehyrodgenase (LDH) cytotoxicity assay (Promega).
  • Fusion proteins that confer selectivity for a specific type of cancer may be tested for their ability to target that specific cancer cell type. For example, a fusion protein comprising a specific IL-4 that targets cells displaying IL-4R Type I or Type II can be assessed for its ability to selectively target such cells by comparing the ability of the fusion protein to kill cancer cells to its ability to kill a normal cell, or a different type of cancer cell (e.g., one that does not express IL-4R Type I or Type II). Alternatively, flow cytometric methods, as are known in the art, may be used to determine if a fusion protein comprising a Type I or Type II receptor-specific IL-4 is able to selectively target a specific type of cell. Binding of a labeled antibody to the bound fusion protein will indicate binding of the fusion protein to the target.
  • Similarly, assays for measuring cell apoptosis are known in the art. Apoptotic cells are characterized by characteristic morphological changes, including chromatin condensation, cell shrinkage and membrane blebbing, which can be clearly observed using light microscopy. The biochemical features of apoptosis include DNA fragmentation, protein cleavage at specific locations, increased mitochondria] membrane permeability, and the appearance of phosphatidylserine on the cell membrane surface. Assays for apoptosis are known in the art. Exemplary assays include TUNEL (Terminal deoxynucleotidyl Transferase Biotin-dUTP Nick End Labeling) assays, caspase activity (specifically caspase-3) assays, and assays for fas-ligand and annexin V. Commercially available products for detecting apoptosis include, for example, Apo-ONE® Homogeneous Caspase-3/7 Assay, FragEL TUNEL kit (ONCOGENE RESEARCH PRODUCTS, San Diego, Calif.), the ApoBrdU DNA Fragmentation Assay (BIOVISION, Mountain View, Calif.), and the Quick Apoptotic DNA Ladder Detection Kit (BIOVISION, Mountain View, Calif.).
  • A variety of cell lines suitable for testing the candidate fusion proteins are known in the art and many are commercially available (for example, from the American Type Culture Collection, Manassas, Va.). Similarly, animal models are known in the art and many are commercially available.
  • Therapeutic Indications and Uses
  • The fusion proteins including IL-4R binding protein and a pro-apoptotic Bcl-2 family member, as described herein, can be used for a variety of therapeutic purposes. In general, the fusion proteins described herein can be used in the treatment or prophylaxis of any disease, disorder or condition which involves cells which express an IL-4R, and which would be benefited by inhibiting cell proliferation or enhancing cell death. In some embodiments, the fusion proteins described herein can be used in the treatment or prophylaxis of any disease, disorder or condition which involves cells which express a Type I or Type II IL-4R, and in which selection of one type of receptor over the other is useful, and which would be benefited by inhibiting cell proliferation or enhancing cell death.
  • In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family member can be used to induce apoptosis or cell death or to treat a disorder associated with abnormal apoptosis or cell proliferation, such as cancer. As used herein, the terms “cancer,” “cancerous,” “hyperproliferative,” or “neoplastic” refer to cells having the capacity for autonomous growth (e.g., an abnormal state or condition characterized by rapidly proliferating cell growth). Hyperproliferative and neoplastic disease states may be categorized as pathologic (e.g., as a deviation from normal but not associated with a disease state). Accordingly, by a “cancer” or “neoplasm” is meant any unwanted growth of cells serving no physiological function. In general, a cell of a neoplasm has been released from its normal cell division control, i.e., a cell whose growth is not regulated by the ordinary biochemical and physical influences in the cellular environment. In most cases, a neoplastic cell proliferates to form a clone of cells which are either benign or malignant. Examples of cancers or neoplasms include, without limitation, transformed and immortalized cells, tumours, and carcinomas such as breast cell carcinomas and prostate carcinomas. The term cancer includes cell growths that are technically benign but which carry the risk of becoming malignant. By “malignancy” is meant an abnormal growth of any cell type or tissue. The term malignancy includes cell growths that are technically benign but which carry the risk of becoming malignant. This term also includes any cancer, carcinoma, neoplasm, neoplasia, or tumor. The terms are therefore meant to include all types of cancerous growths or oncogenic processes, metastatic tissue or malignantly transformed cells, tissues or organs, irrespective of hi stopathologic type or stage of invasiveness. In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family member is not used in connection with a cancer affecting a stem cell.
  • Most cancers fall within three broad histological classifications: carcinomas, which are the predominant cancers and are cancers of epithelial cells or cells covering the external or internal surfaces of organs, glands, or other body structures (e.g., skin, uterus, lung, breast, prostate, stomach, bowel), and which tend to metastasize; sarcomas, which are derived from connective or supportive tissue (e.g., bone, cartilage, tendons, ligaments, fat, muscle); and hematologic tumors, which are derived from bone marrow and lymphatic tissue. Examples of cancers include, without limitation, carcinomas, sarcomas, and hematopoietic neoplastic disorders e.g., leukemia.
  • Carcinomas may be adenocarcinomas (which generally develop in organs or glands capable of secretion, such as breast, lung, colon, prostate or bladder) or may be squamous cell carcinomas (which originate in the squamous epithelium and generally develop in most areas of the body).
  • Sarcomas may be osteosarcomas or osteogenic sarcomas (bone), chondrosarcomas (cartilage), leiomyosarcomas (smooth muscle), rhabdomyosarcomas (skeletal muscle), mesothelial sarcomas or mesotheliomas (membranous lining of body cavities), fibrosarcomas (fibrous tissue), angiosarcomas or hemangioendotheliomas (blood vessels), liposarcomas (adipose tissue), gliomas or astrocytomas (neurogenic connective tissue found in the brain), myxosarcomas (primitive embryonic connective tissue), or mesenchymous or mixed mesodermal tumors (mixed connective tissue types).
  • Hematopoietic neoplastic disorders include diseases involving hyperplastic/neoplastic cells of hematopoietic origin e.g., arising from myeloid, lymphoid or erythroid lineages or precursor cells thereof. Preferably, the diseases arise from poorly differentiated acute leukemias (e.g., erythroblastic leukemia and acute megakaryoblastic leukemia). Additional exemplary myeloid disorders include, but are not limited to, acute promyeloid leukemia (APML), acute myeloenous leukemia (AML) and chronic myeloenous leukemia (CML); lymphoid malignancies include but are not limited to acute lymphoblastic leukemia (ALL), which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia, and Waldenstrom's macroglobulinemia.
  • Additional forms of malignant lymphomas include, but are not limited to non-Hodgkin lymphoma and variants thereof, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Stemberg disases.
  • Cancers may also be named based on the organ in which they originate i.e., the “primary site,” for example, cancer of the breast, brain, lung, liver, skin, prostate, testicle, bladder, colon and rectum, cervix, uterus, etc. This naming persists even if the cancer metastasizes to another part of the body, that is different from the primary site. Cancers named based on primary site may be correlated with histological classifications. For example, lung cancers are generally small cell lung cancers or non-small cell lung cancers, which may be squamous cell carcinoma, adenocarcinoma, or large cell carcinoma; skin cancers are generally basal cell cancers, squamous cell cancers, or melanomas. Lymphomas may arise in the lymph nodes associated with the head, neck and chest, as well as in the abdominal lymph nodes or in the axillary or inguinal lymph nodes. Identification and classification of types and stages of cancers may be performed by using for example information provided by the Surveillance, Epidemiology, and End Results (SEER) Program of the National Cancer Institute.
  • In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof, can be used to treat cancers such as gastric carcinoma, invasive pituitary adenomas, biliary tract carcinoma, cervical cancer, lymphoma, melanoma, chronic lymphocytic leukemia, non-hodgkins lymphoma, follicular lymphoma, pancreatic cancer, colorectal cancer, colon cancer, thyroid cancer, liver cancer, ovarian cancer, prostate cancer, bladder cancer, renal cell carcinoma, mesothelioma, rhabdomyosarcoma, breast cancer, non-small cell lung cancer, head and neck cancers, or Kaposi's carcinoma.
  • In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof, can be used to treat CNS cancers such as gliomas, meningeal tumours, diffuse intrinsic pontine glioma, medulloblastoma, neuroblastoma, anaplastic astrocytoma, glioblastoma multiforme, metastatic brain cancer, or CNS lymphoma.
  • The fusion proteins can be used to treat, stabilize or prevent cancer. Fusion proteins can also be used in the treatment of indolent cancers, recurrent cancers including locally recurrent, distantly recurrent and/or refractory cancers (i.e. cancers that have not responded to other anti-cancer treatments), metastatic cancers, locally advanced cancers and aggressive cancers. In these contexts, the fusion proteins may exert either a cytotoxic or cytostatic effect resulting in, for example, a reduction in the number or growth of cancer cells, a reduction in the size of a tumor, the slowing or prevention of an increase in the size of a tumor, an increase in the disease-free survival time between the disappearance or removal of a tumor and its reappearance, prevention of an initial or subsequent occurrence of a tumor (e.g. metastasis), an increase in the time to progression, reduction of one or more adverse symptoms associated with a tumor, or an increase in the overall survival time of a subject having cancer.
  • Other examples of proliferative and/or differentiative disorders that can be treated using a fusion protein including a pro-apoptotic Bcl-2 family member include proliferative non-malignant diseases such as pulmonary fibrosis or hyperplasia (such as benign prostatic hyperplasia), cardiac fibrosis, or liver fibrosis; inflammatory conditions such as prostatitis, vernal keratoconjunctivitis, artherosclerosis, idiopathic pulmonary pneumonia; or autoimmune conditions such as Graves disease.
  • In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof, is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis. In some embodiments, the IL-4R binding protein-pro-apoptotic Bcl-2 family fusion protein is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis, when compared to a suitable control, such as IL-4 alone, IL-4 joined to a non-pro-apoptotic Bcl-2 family protein, etc. A suitable control may also include a previously-established standard. Accordingly, any test or assay for determining the activity or efficacy of an IL-4R binding protein-pro-apoptotic Bcl-2 family fusion protein may be compared to the established standard and it may not be necessary to include a control for comparison each time. By “inhibiting cell survival” is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will survive. By “inhibiting cell proliferation” is meant decreasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the growth or proliferation of a cell. By “enhancing cell death or apoptosis” is meant increasing (e.g., by at least 10%, 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more) the probability that a cell at risk of cell death will undergo apoptotic, necrotic, or any other form of cell death.
  • In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof, is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis by at least 20%, 30%, or by as much as 50%, 75%, 85% or 90% or more, when compared to a cell cultured under similar conditions but not contacted with the fusion protein. Suitable assays for measuring the inhibition of cell survival, inhibition of cell proliferation, or enhancement of cell death or apoptosis are described herein or known in the art.
  • In some embodiments, the IC5o of a fusion protein including a pro-apoptotic Bcl-2 family protein member, or a fragment thereof, in inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis, can be in the range from about 0.1 ng/mL to about 10,000 ng/mL, or any value therebetween, such as about 0.5 ng/mL, 1 ng/mL, 5 ng/mL, 10 ng/mL, 25 ng/mL, 50 ng/mL, 75 ng/mL, 100 ng/mL, 150 ng/mL, 200 ng/mL, 250 ng/mL, 300 ng/mL, 350 ng/mL, 400 ng/mL, 450 ng/mL, 500 ng/mL, 550 ng/mL, 600 ng/mL, 650 ng/mL, 700 ng/mL, 750 ng/mL, 800 ng/mL, 850 ng/mL, 900 ng/mL, 950 ng/mL, or 1000 ng/mL.
  • “Target cells” include, without limitation, neurons, lymphocytes, stem cells, epithelial cells, cancer cells, neoplasm cells, immune cells, non-malignant cells of the tumour microenvironment, hyper-proliferative cells, etc. The target cell chosen will depend on the disease or injury or condition the fusion protein is intended to treat.
  • Pharmaceutical Compositions, Dosages and Administration
  • Pharmaceutical compositions according to the present disclosure can include one or more fusion proteins and one or more non-toxic, pharmaceutically-acceptable carriers, diluents, excipients and/or adjuvants. Such compositions can be suitable for use in treatment of therapeutic indications as described herein.
  • If desired, other active ingredients may be included in the compositions. Accordingly, in some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family member can be administered in therapeutically-effective amounts together with one or more anti-cancer or other therapeutics. The fusion protein(s) can be administered before, during or after treatment with the anti-cancer or other therapeutic. An “anti-cancer therapeutic” is a compound, composition, or treatment (e.g., surgery) that prevents or delays the growth and/or metastasis of cancer cells. Such anti-cancer therapeutics include, but are not limited to, surgery (e.g., removal of all or part of a tumor), chemotherapeutic drug treatment, radiation, gene therapy, hormonal manipulation, immunotherapy (e.g., therapeutic antibodies and cancer vaccines) and antisense or RNAi oligonucleotide therapy. Examples of useful chemotherapeutic drugs include, but are not limited to, hydroxyurea, busulphan, cisplatin, carboplatin, chlorambucil, melphalan, cyclophosphamide, Ifosfamide, danorubicin, doxorubicin, epirubicin, mitoxantrone, vincristine, vinblastine, vinorelbine, etoposide, teniposide, paclitaxel, docetaxel, gemcitabine, cytosine, arabinoside, bleomycin, neocarcinostatin, suramin, taxol, mitomycin C, Avastin, Herceptin®, fluorouracil, temozolamide, etc. The fusion protein(s) are also suitable for use with standard combination therapies employing two or more chemotherapeutic agents. It is to be understood that anti-cancer therapeutics includes novel compounds or treatments developed in the future.
  • The fusion protein can also be administered in combination with a sensitizing agent, such as a radio-sensitizer (see for example Diehn et al., J. Natl. Cancer Inst. 98:1755-7, 2006). Generally a sensitizing agent is any agent that increases the activity of a fusion protein. For example, a sensitizing agent will increase the ability of a fusion protein to inhibit cancer cell growth or kill cancer cells. Exemplary sensitizing agents include antibodies to IL-10, bone morphogenic proteins and HDAC inhibitors (see for example Sakariassen et al., Neoplasia 9(11):882-92, 2007). These sensitizing agents can be administered before or during treatment with the fusion protein. Exemplary dosages of such sensitizing agents include at least 1 ug/mL, such as at least 10 ug/mL, at least 100 ug/mL, for example 5-100 ug/mL or 10-90 ug/mL. The sensitizing agents can be administered daily, three times a week, twice a week, once a week or once every two weeks. Sensitizing agents can also be administered after treatment with the fusion protein is finished.
  • The fusion proteins may be used as part of a neo-adjuvant therapy (to primary therapy), as part of an adjuvant therapy regimen, where the intention is to cure the cancer in a subject. The fusion proteins can also be administered at various stages in tumor development and progression, including in the treatment of advanced and/or aggressive neoplasias (e.g., overt disease in a subject that is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy), metastatic disease, locally advanced disease and/or refractory tumors (e.g., a cancer or tumor that has not responded to treatment). “Primary therapy” refers to a first line of treatment upon the initial diagnosis of cancer in a subject. Exemplary primary therapies may involve surgery, a wide range of chemotherapies and radiotherapy. “Adjuvant therapy” refers to a therapy that follows a primary therapy and that is administered to subjects at risk of relapsing. Adjuvant systemic therapy is begun soon after primary therapy, for example 2, 3, 4, 5, or 6 weeks after the last primary therapy treatment to delay recurrence, prolong survival or cure a subject. As discussed herein, it is contemplated that the fusion proteins can be used alone or in combination with one or more other chemotherapeutic agents as part of an adjuvant therapy. Combinations of the fusion proteins and standard chemotherapeutics may act to improve the efficacy of the chemotherapeutic and, therefore, can be used to improve standard cancer therapies. This application can be particularly important in the treatment of drug-resistant cancers which are not responsive to standard treatment.
  • In cancer, the microenvironment of a tumor contains both malignant and non-malignant cells. The tumor microenvironment can be identified using one or more of the following criteria: (a) a region comprising non-malignant cells which share the same physiological environment, or which are directly adjacent to malignant cells; (b) the extended tumor region; (c) an area of inflammation surrounding or proximal to a tumor; (d) an area in which the number or rate of proliferation of regulatory T cells is elevated; and (e) an area in which tumor-associated macrophages, dendritic cells, myeloid-derived suppressor cells, Th2 cells or fibrocytes are elevated. Within the context of non-solid tumor types, the tumor microenvironment may also be determined by the local cell-cell interactions between malignant cells and between malignant cells and any adjacent or nearby non-malignant cells. Such interactions may include, for example, cell adhesion events and/or paracrine effects of soluble mediators produced by one cell (malignant or non-malignant) on another cell (malignant or non-malignant) in the tumor microenvironment.
  • The non-malignant cells in the tumor microenvironment can be important for tumor initiation and progression (Reynolds et al., Cancer Res., 1996, 56(24):5754-5757). The non-malignant cells, also called stromal cells, occupy or accumulate in the same cellular space as malignant cells, or the cellular space adjacent or proximal to malignant cells, which modulate tumor cell growth or survival. For example, non-malignant cells that normally function to support inflammatory and immune response can be capable of contributing to tumor initiation or progression. Accordingly, in alternative embodiments, a fusion protein including a pro-apoptotic Bcl-2 family member can be used for inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis of a non-malignant cell that expresses a Type I or Type II IL-4R in a tumour microenvironment. Such non-malignant cells can be immunoregulatory or inflammatory cells such as antigen presenting cells (e.g., macrophages, dendritic cells, B cells) or myeloid-derived suppressor cells (e.g., myeloid-derived monocytes and tie-2-expressing monocytes) present within the tumor microenvironment, and inhibition of T cell subsets that function to support tumor progression (e.g., regulatory T cells and Th2 helper cells) and/or suppressing production of one or more inflammatory cytokines in a tumor microenvironment. Among the non-malignant cells of a tumor microenvironment are regulatory T cells, which are observed in higher frequencies in a number of tumors, including Hodgkin's lymphoma, non-Hodgkin's lymphoma (Shi et al., Ai Zheng., 2004, 23(5):597-601 (abstract only)), malignant melanoma (Viguier et al., J. Immunol., 2004, 173(2):1444-53; Javia et al., J. Immunother., 2003, 26(1):85-93), and cancers of the ovary (Woo et al., Cancer Res., 2001, 61(12):4766-72), gastrointestinal tract (Ichihara et al., Clin Cancer Res., 2003, 9(12):4404-4408; Sasada et al., Cancer, 2003, 98(5):1089-1099), breast (Liyanage et at., J Immunol., 2002, 169(5):2756-2761), lung (Woo et al., Cancer Res., 2001, 61(12):4766-72), and pancreas (Liyanage et al., J Immunol., 2002, 169(5):2756-2761). The regulatory T cells are recruited to the tumor site in response to chemokines secreted by the tumor cells. See e.g., Curiel et al., Nat. Med., 2004, 10:942-949. An increase in the number of regulatory T cells may also correlate with poor prognosis (Curiel et al., Nat. Med 2004, 10:942-949; Sasada et al., Cancer, 2003, 98:1089-1099). Conversely, regulatory T cells are observed to decrease following chemotherapy (Beyer et al., Blood, 2005, 106:2018-2025). The tumour micro-environment can also have a higher proportion of Th2 cells, when compared to Th1 cells, which is associated with poor prognosis and survival. In alternative embodiments, a fusion protein including a pro-apoptotic Bcl-2 family member according to the invention, such as cpIL-4-Bad, is useful for restoring the Th1>>Th2 balance by, for example, depleting Th2 cells. In some embodiments, a fusion protein including a pro-apoptotic Bcl-2 family member according to the invention, such as cpIL-4-Bad, may be administered to a subject prior to, or during, chemotherapy, radiation therapy, immunotherapy, etc. to disable or downregulate the tumor microenvironment.
  • Non-malignant cells can also be fibroblasts, myofibroblasts, glial cells, epithelial cells, adipocytes, vascular cells (including blood and lymphatic vascular endothelial cells and pericytes), resident and/or recruited inflammatory and immune (e.g., macrophages, dendritic cells, myeloid suppressor cells, granulocytes, lymphocytes, etc.), resident and/or recruited cells that are capable of giving rise to or differentiating into any of the above-noted non-malignant cells, and any functionally distinct subtypes of the above-noted cells as known in the art.
  • A “subject” can be a mammal in need of treatment, such as a human or veterinary patient (e.g., rodent, such as a mouse or rat, a cat, dog, cow, horse, sheep, goat, or other livestock). In some embodiments, a “subject” may be a clinical patient, a clinical trial volunteer, an experimental animal, etc. The subject may be suspected of having or at risk for having a condition characterized by cell proliferation, be diagnosed with a condition characterized by cell proliferation, or be a control subject that is confirmed to not have a condition characterized by cell proliferation, as described herein. Diagnostic methods for conditions characterized by cell proliferation and the clinical delineation of such diagnoses are known to those of ordinary skill in the art.
  • The composition can be a liquid solution, suspension, emulsion, sustained release formulation, or powder, and can be formulated with a pharmaceutically acceptable carrier. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. The term “pharmaceutically-acceptable carrier” refers to a carrier medium or vehicle which does not interfere with the effectiveness of the biological activity of the active ingredients and which is not toxic to the host or subject.
  • Fusion proteins can be delivered along with a pharmaceutically-acceptable vehicle. In one example, the vehicle may enhance the stability and/or delivery properties. Thus, the disclosure also provides for formulation of the fusion protein with a suitable vehicle, such as an artificial membrane vesicle (including a liposome, noisome, nanosome and the like), microparticle or microcapsule, or as a colloidal formulation that comprises a pharmaceutically acceptable polymer. The use of such vehicles/polymers may be beneficial in achieving sustained release of the fusion proteins. Alternatively, or in addition, the fusion protein formulations can include additives to stabilize the protein in vivo, such as human serum albumin, or other stabilizers for protein therapeutics known in the art. Fusion protein formulations can also include one or more viscosity enhancing agents which act to prevent backflow of the formulation when it is administered, for example by injection or via catheter. Such viscosity enhancing agents include, but are not limited to, biocompatible glycols and sucrose.
  • Pharmaceutical compositions containing one or more fusion proteins can be formulated as a sterile injectable aqueous or oleaginous suspension according to methods known in the art and using suitable one or more dispersing or wetting agents and/or suspending agents, such as those mentioned above. The sterile injectable preparation can be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Acceptable vehicles and solvents that can be employed include, but are not limited to, water, Ringer's solution, lactated Ringer's solution and isotonic sodium chloride solution. Other examples include, sterile, fixed oils, which are conventionally employed as a solvent or suspending medium, and a variety of bland fixed oils including, for example, synthetic mono- or diglycerides. Fatty acids such as oleic acid can also be used in the preparation of injectables.
  • In some embodiments, the fusion protein is conjugated to a water-soluble polymer, e.g., to increase stability or circulating half-life or reduce immunogenicity. Clinically acceptable, water-soluble polymers include, but are not limited to, polyethylene glycol (PEG), polyethylene glycol propionaldehyde, carboxymethylcellulose, dextran, polyvinyl alcohol (PVA), polyvinylpyrrolidone (PVP), polypropylene glycol homopolymers (PPG), polyoxyethylated polyols (POG) (e.g., glycerol) and other polyoxyethylated polyols, polyoxyethylated sorbitol, or polyoxyethylated glucose, and other carbohydrate polymers. Methods for conjugating polypeptides to water-soluble polymers such as PEG are described, e.g., in U.S. patent Pub. No. 20050106148 and references cited therein. In one example the polymer is a pH-sensitive polymers designed to enhance the release of drugs from the acidic endosomal compartment to the cytoplasm (see for example, Henry et al., Biomacromolecules 7(8):2407-14, 2006).
  • Typically vaccines are prepared in an injectable form, either as a liquid solution or as a suspension. Solid forms suitable for injection may also be prepared as emulsions, or with the polypeptides encapsulated in liposomes. The cells are injected in any suitable carrier known in the art. Suitable carriers typically comprise large macromolecules that are slowly metabolized, such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates, and inactive virus particles. Such carriers are well known to those skilled in the art. These carriers may also function as adjuvants.
  • Adjuvants are immunostimulating agents that enhance vaccine effectiveness. Effective adjuvants include, but are not limited to, aluminum salts such as aluminum hydroxide and aluminum phosphate, muramyl peptides, bacterial cell wall components, saponin adjuvants, and other substances that act as immunostimulating agents to enhance the effectiveness of the composition.
  • Vaccines are administered in a manner compatible with the dose formulation. By an effective amount is meant a single dose, or a vaccine administered in a multiple dose schedule, that is effective for the treatment or prevention of a disease or disorder. Preferably, the dose is effective to inhibit the growth of a neoplasm. The dose administered will vary, depending on the subject to be treated, the subject's health and physical condition, the capacity of the subject's immune system to produce antibodies, the degree of protection desired, and other relevant factors. Precise amounts of the active ingredient required will depend on the judgement of the practitioner.
  • The pharmaceutical compositions described herein include one or more fusion proteins in an amount effective to achieve the intended purpose. Typically, compositions including a fusion protein containing a pro-apoptotic Bcl-2 family member are administered to a patient already suffering from a disease, disorder or condition characterized by cell proliferation, or at risk for such a disease, disorder or condition, in an amount sufficient to cure or at least partially arrest a symptom associated with cell proliferation or reduce cell growth.
  • The skilled person will therefore recognize that the dosage to be administered is not subject to defined limits. Prior to administration for therapeutic purposes, the dosage of the fusion protein may need to be modified or adapted for the particular purpose, for example the concentration of fusion protein needed for whole body administration may differ from that used for local administration. Similarly, the toxicity of the therapeutic may change depending upon the mode of administration and overall composition being used (e.g., buffer, diluent, additional chemotherapeutic, etc.).
  • An “effective amount” of a pharmaceutical composition according to the invention includes a therapeutically effective amount or a prophylactically effective amount. A “therapeutically effective amount” refers to an amount of the fusion protein effective, at dosages and for periods of time necessary, that ameliorates the symptoms of the disease, disorder or condition to be treated. A therapeutically effective amount of a compound may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the compound to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the fusion protein are outweighed by the therapeutically beneficial effects. Determination of a therapeutically effective dose of a compound is well within the capability of those skilled in the art. For example, the therapeutically effective dose can be estimated initially either in cell culture assays, or in animal models, such as those described herein. A “prophylactically effective amount” refers to to an amount of the fusion protein effective, at dosages and for periods of time necessary, that achieves the desired prophylactic result, such as delay in onset of symptoms of a neurological disorder or continued remission of a cancer. Animal models can also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in other animals, including humans, using standard methods known in those of ordinary skill in the art.
  • Concentration of the fusion protein in the final formulation can be at least 0.1 mg/mL, such as at least 1 ng/mL or at least 1 ug/mL or at least 1 mg/mL. For example, the concentration in the final formulation can be between about 0.01 ug/mL and about 1,000 ug/mL. In one example, the concentration in the final formulation is between about 0.01 mg/mL and about 100 mg/mL.
  • In some embodiments, a fusion protein including an anti-apoptotic Bcl-2 family protein, or fragment thereof, is administered at concentrations ranging from about 10 ng/mL to about 10,000 ng/mL, or any value therebetween, such as about 25 ng/mL, 50 ng/mL, 75 ng/mL, 100 ng/mL, 150 ng/mL, 200 ng/mL, 250 ng/mL, 300 ng/mL, 350 ng/mL, 400 ng/mL, 450 ng/mL, 500 ng/mL, 550 ng/mL, 600 ng/mL, 650 ng/mL, 700 ng/mL, 750 ng/mL, 800 ng/mL, 850 ng/mL, 900 ng/mL, 950 ng/mL, 1000 ng/mL, 1500 ng/mL, 2000 ng/mL, 2500 ng/mL, 3000 ng/mL, 3500 ng/mL, 4000 ng/mL, 4500 ng/mL, 5000 ng/mL, 5500 ng/mL, 6000 ng/mL, 6500 ng/mL, 7000 ng/mL, 7500 ng/mL, 8000 ng/mL, 8500 ng/mL, 9000 ng/mL, 9500 ng/mL, or 10000 ng/mL.
  • In some embodiments, a fusion protein including an pro-apoptotic Bcl-2 family protein, or fragment thereof, is administered at concentrations ranging from about 0.1 ng/mL to about 10,000 ng/mL
  • However, it will be understood that the actual amount of the compound(s) to be administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms. The above dosage range is given by way of example only and is not intended to limit the scope in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing harmful side effects, for example, by first dividing the larger dose into several smaller doses for administration throughout the day.
  • One of ordinary skill in the art will appreciate that the dosage will depend, among other things, upon the type of fusion protein being used and the type of disorder or condition being treated.
  • In general, the fusion proteins according to the present disclosure contain substantially human sequences and are therefore less antigenic than, for example, immunotoxins or other molecules that contain non-human sequences. In some embodiments, the fusion proteins according to the present disclosure contain at least 80%, for example, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% human sequences. In some embodiments, the fusion proteins according to the present disclosure can be administered at substantially lower doses than for example, immunotoxins, or native IL-4R binding protein, such as IL-4 or IL-13.
  • In some embodiments, the fusion proteins may elicit some level of antibody response when administered to a subject, which in some cases may lead to undesirable side effects. Therefore, if necessary, the antigenicity of the fusion proteins can be assessed as known in the art and/or described herein. For example, in vivo toxic effects of the fusion proteins can be evaluated by measuring their effect on animal body weight during treatment and by performing hematological profiles and liver enzyme analysis after the animal has been killed. The general toxicity of the fusion proteins can be tested according to methods known in the art. For example, the overall systemic toxicity of the fusion proteins can be tested by determining the dose that kills 100% of mice (i.e. LDioo) or kills 50% of mice (i.e. LD50) following a single intravenous injection. Doses that are at least about 2, 5, or 10-fold less than the LDioo or LD50 can be selected for administration into other mammals, such as a human.
  • The kinetics and magnitude of the antibody response to the fusion proteins described herein can be determined, for example, in immunocompetent mice and can be used to facilitate the development of a dosing regimen that can be used in an immunocompetent human.
  • Immunocompetent mice such as the strain C57-BL6 are administered intravenous doses of fusion protein. The mice are killed at varying intervals (e.g. following single dose, following multiple doses) and serum obtained. An ELISA-based assay can be used to detect the presence of anti-fusion protein antibodies.
  • Serum samples from mice can be assessed for the presence of anti-fusion protein antibodies as known in the art. As another example, epitope mapping can also be used to determine antigenicity of proteins as described in Stickler, et al., J. Immunotherapy, 23:654-660, 2000. Briefly, immune cells known as dendritic cells and CD4+ T cells are isolated from the blood of community donors who have not been exposed to the protein of interest. Small synthetic peptides spanning the length of the protein are then added to the cells in culture. Proliferation in response to the presence of a particular peptide suggests that a T cell epitope is encompassed in the sequence. This peptide sequence can subsequently be deleted or modified in the fusion protein thereby reducing its antigenicity.
  • Therapeutic efficacy and toxicity can also be determined by standard pharmaceutical procedures such as, for example, by determination of the median effective dose, or ED50 (i.e. the dose therapeutically effective in 50% of the population) and the median lethal dose, or LD50 (i.e. the dose lethal to 50% of the population). The dose ratio between therapeutic and toxic effects is known as the “therapeutic index,” which can be expressed as the ratio, LD50/ED50. The data obtained from cell culture assays and animal studies can be used to formulate a range of dosage for human or animal use. The dosage contained in such compositions is usually within a range of concentrations that include the ED50 and demonstrate little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the subject, and the route of administration and the like.
  • Administration of the fusion proteins can be intralesionally, for instance by direct injection directly into the apoptotic tissue site; into a site that requires cell growth; into a site where a cell, tissue or organ is at risk of cell death; or into a site of hyperproliferati on or into a tumor. Alternatively, the fusion protein can be administered systemically. For methods of combination therapy comprising administration of a fusion protein in combination with a chemotherapeutic agent, the order in which the compositions are administered is interchangeable. Concomitant administration is also envisioned.
  • Typically in the treatment of cancer, fusion proteins are administered systemically to patients, for example, by bolus injection or continuous infusion into a patient's bloodstream. Alternatively, the fusion proteins may be administered locally, at the site of a tumor (intratumorally). When a fusion protein is administered intratumorally, the administration can be via any route, e.g., locally, regionally, focally, systemic, convection enhanced delivery or combinations thereof.
  • When used in conjunction with one or more known chemotherapeutic agents, the compounds can be administered prior to, or after, administration of the chemotherapeutic agents, or they can be administered concomitantly. The one or more chemotherapeutics may be administered systemically, for example, by bolus injection or continuous infusion, or they may be administered orally.
  • For administration to an animal, the pharmaceutical compositions can be formulated for administration by a variety of routes. For example, the compositions can be formulated for topical, rectal or parenteral administration or for administration by inhalation or spray. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrathecal, intrasternal injection or infusion techniques. Direct injection or infusion into a tumor is also contemplated. Convection enhanced delivery can also be used to administer the fusion protein.
  • In one example, the fusion protein can be injected into a subject having cancer, using an administration approach similar to the multiple injection approach of brachytherapy. For example, multiple aliquots of the purified fusion protein in the form of a pharmaceutical composition or formulation and in the appropriate dosage units, may be injected using a needle. Alternative methods of administration of the fusion proteins will be evident to one of ordinary skill in the art. Such methods include, for example, the use of catheters, or implantable pumps to provide continuous infusion of the fusion protein to the subject in need of therapy.
  • As is known in the art, software planning programs can be used in combination with brachytherapy treatment and ultrasound, for example, for placement of catheters for infusing fusion proteins to treat, for example, brain tumors or other localized tumors. For example, the positioning and placement of the needle can generally be achieved under ultrasound guidance. The total volume, and therefore the number of injections and deposits administered to a patient, can be adjusted, for example, according to the volume or area of the organ to be treated. An example of a suitable software planning program is the brachytherapy treatment planning program Variseed 7.1 (Varian Medical Systems, Palo Alto, Calif.) or iPlan (BrainLab, Munich, Germany) for convection enhanced delivery to the brain. Such approaches have been successfully implemented in the treatment of prostate cancer and brain cancer among among others.
  • Fusion proteins can be used in inhibiting cell survival or inhibiting cell proliferation in the central nervous system (CNS). When the site of delivery is the brain, the fusion protein must be capable of being delivered to the brain. The blood-brain barrier limits the uptake of many therapeutic agents into the brain and spinal cord from the general circulation. Molecules which cross the blood-brain barrier use two main mechanisms: free diffusion and facilitated transport. Because of the presence of the blood-brain barrier, attaining beneficial concentrations of a given fusion protein in the CNS may require the use of specific drug delivery strategies. Delivery of fusion proteins to the CNS can be achieved by several methods.
  • One method relies on neurosurgical techniques. For instance, fusion proteins can be delivered by direct physical introduction into the CNS, such as intraventricular, intralesional, or intrathecal injection. Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Methods of introduction are also provided by rechargeable or biodegradable devices. Another approach is the disruption of the blood-brain barrier by substances which increase the permeability of the blood-brain barrier. Examples include intra-arterial infusion of poorly diffusible agents such as mannitol, pharmaceuticals which increase cerebrovascular permeability such as etoposide, or vasoactive agents, such as leukotrienes or by convention enhanced delivery by catheter (CED). Further, it may be desirable to administer the compositions locally to the area in need of treatment; this can be achieved, for example, by local infusion during surgery, by injection, by means of a catheter, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including membranes, such as silastic membranes, or fibers. A suitable membrane is Gliadel® (Eisai Inc.).
  • Non-viral approaches can also be employed for the introduction of a therapeutic to a cell requiring modulation of cell death (e.g., a cell of a patient). For example, a nucleic acid molecule can be introduced into a cell by administering the nucleic acid molecule in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:7413, 1987; Ono et al., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989; Staubinger et al., Methods in Enzymology 101:512, 1983), asialoorosomucoid-polylysine conjugation (Wu et al., Journal of Biological Chemistry 263:14621, 1988; Wu et al., Journal of Biological Chemistry 264:16985, 1989), or by micro-injection under surgical conditions (Wolff et al., Science 247:1465, 1990). Preferably the nucleic acids are administered in combination with a liposome and protamine.
  • Gene transfer can also be achieved using non-viral means involving transfection in vitro. Such methods include the use of calcium phosphate, DEAE dextran, electroporation, and protoplast fusion. Liposomes can also be potentially beneficial for delivery of DNA into a cell. Transplantation of a fusion protein into the affected tissues of a patient can also be accomplished by transferring a normal nucleic acid into a cultivatable cell type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue.
  • cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters), and regulated by any appropriate mammalian regulatory element. For example, if desired, enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid. The enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers. Alternatively, if a genomic clone is used as a therapeutic construct, regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
  • The present invention will be further illustrated in the following examples.
  • EXAMPLES Example 1
  • This example describes making circularly permuted IL-4 proteins.
  • The coding sequence of IL-4 is designed to be reorganized creating a new amino terminus and a new carboxy terminus. The site of reorganization is selected and coding regions are developed synthetically or using the native sequence as a template. PCR can be used to amplify the separate coding regions and the 5′ and 3′ ends of the separate fragments are designed to overlap, thus allowing for the formation of a new coding sequence in which the newly generated peptide can for example encode a first amino acid that in the native protein may have been the 40th amino acid. Specific examples of making circularly permuted ligands are provided in U.S. Pat. No. 6,011,002.
  • The following sequence is used as a reference.
  • Name IL-4 Linker Toxin Description
    cpIL4- M D TTEKETFCRAAT ASGGPE PE38KDEL Circularly
    PE VLRQFYSHHEKDT (SEQ ID permuted
    RCLGATAQQFHRH NO: 29) human IL-4
    KQURFLKRLDRNL fused to a
    WGLAGLNSCPVKE Pseudo-
    ANQSTLENFLERLK monas
    TIMREKYSKCSS
    Figure US20220048965A1-20220217-P00011
    exotoxin
    Figure US20220048965A1-20220217-P00012
    HKCDITLQEIIK
    (PE38KDEL)
    TLNSLTEQKTLCTE via a
    LTVTDIFAASK (SEQ ID ASGGPE
    NO: 2). linker
  • Example 2
  • cpIL-4-BAD was prepared using standard techniques and tested on IL-4Ra-expressing cancer cells in vitro and on IL-4Ra positive xenograft tumors in an athymic nude mouse model, using cpIL4-PE as a reference. The results indicated that cpIL-4-BAD was effective at killing the cancer cells and at regressing the tumors.
  • More specifically, cpIL-4-BAD fusion protein was prepared by transducing BL21(DE3)pLysS bacteria with chimeric plasmid cpIL-4BAD in a pET 24a expression vector (FIG. 1). The freshly transformed bacteria were plated on LB with kanamycin. Colonies were picked and transferred into 1 litre of superbroth enriched with glucose, MgCl2 and kanamycin and the cells were induced with 1 mM IPTG. The growth of the cells was monitored for 6 hours. IL-4-BAD fusion proteins were refolded and purified using a HiTrap Column followed by two rounds of a MonoS column, after which cp1L-4-BAD fractions were evaluated by SDS-PAGE electrophoresis. This strategy yielded about 0.980 mg/L cpIL-4BAD at pH 6.5, and about 2.930 mg/L cp1L-4BAD at pH 6.8.
  • IL-4Ra positive tumor cells (U 251 cells and Daudi cells) were plated in quadruplicate in 6-well petri-dishes overnight in complete medium. 0.1-1000 ng/mL of cpIL-4BAD was added and the plates were incubated for five days in a CO2 incubator at 37° C. On day 5, the plates were washed with 1×PBS and the cells were detached with trypsin treatment. After trypsin inactivation, viable cells were counted using the trypan blue exclusion technique.
  • The results indicated that the cp1L-4BAD fusion protein killed U 251 cells in a concentration dependent manner in vitro, with an IC50 value of about 0.6 ng/mL (FIG. 2). 100× excess of IL-4 blocked the ability of cp1L-4BAD to kill the U 251 cells, demonstrating that the cpIL-4BAD activity was specific (FIG. 3). Also, the IC50 value rose from about 0.3 ng/mL to >1000 ng/mL in 5-day cell viability assays.
  • The cpIL-4BAD fusion protein killed Daudi cells in a concentration-dependant manner in vitro, with an IC50 value of about 0.1 ng/mL (FIG. 4). 100× excess of IL-4 also blocked the ability of cpIL-4BAD to kill the Daudi cells, demonstrating that the cpIL-4BAD activity was specific (FIG. 5). The IC50 value rose from about 0.3 ng/mL to >1000 ng/mL in 5-day cell viability assays.
  • The cp1L-4BAD fusion protein was also found to decrease the colony numbers of IL-4Ra positive tumor cells in a colony formation assay. More specifcally, 5000 U 251 cells were plated in 10 cm culture plates (6 plates in total), cp1L-4BAD was added and the cells were incubated for 10 days. Colonies were stained with Crystal Blue and counted. The results indicated that cp1L-4BAD decreased U 251 colony numbers in a concentration dependent manner in vitro, with an IC50 value of about 5 ng/mL (FIG. 6).
  • The effectiveness of the cp1L-4BAD fusion protein was assessed in athymic mice after the development of subcutaneous glioma tumors with U 251 tumor cells. More specifically, cpIL-4BAD was injected intratumourally (IT) at 100 μg/kg (6 injections) and intraperitoneally (IP) 100 μg/kg (6 injections). 0.2% HAS/PBS was used as a vehicle control. The assessed end points were tumor size and survival. The results indicated that IT administration of cp1L-4BAD regressed the tumor growth significantly compared to the placebo control-treated animals after 6 injections. IP administration regressed the tumors dramatically compared to IT treated mice. Tumors in both groups of mice recurred beyond day 40, when the mice in control groups were euthanized for ethical reasons, as the tumors were of 2000 mm3 on day 36 (FIG. 7). cpIL-4BAD-treated mice also survived longer than placebo treated mice (FIG. 8).
  • Example 3
  • Six additional IL-4Ra positive cell lines were tested for their sensitivity to the cpIL-4BAD fusion protein and cpIL-4PE under identical experimental conditions. ICso values were determined by counting the cells using the trypan blue exclusion technique (Table 3).
  • IC50 (ng/mL)
    Cell Line Origin cpIL-4BAD cpIL-4PE
    YCUT891 Tongue 11.0 8.0
    YCUM862 Oropharynx 1.1 0.6
    KCCOR891 Oral Floor 0.15 9.0
    KCCL871 Larynx 0.5 1.5
    YCUM911 Oropharynx 0.2 4.5
    KCCTCM901 Metastasis to the chest fluid 1.0 0.7
  • Example 4
  • IL-4BAD, cpIL-4BAD and cpS4-BAD fusion proteins were expressed and purified. More specifically, cDNAs of IL-4BAD, cpIL-4BAD and cpS4-BAD were PCR cloned into BamHI/Xhol sites of a pGWO7 E. coli. expression vector (FIG. 9). The obtained vectors were verified by DNA sequencing (FIGS. 10A-D).
  • Protein expression was performed in E. coli cells. IL-4BAD, cpIL-4BAD and cpS4-BAD proteins were expressed in 1 L cultures in insoluble form, purified under denaturing conditions using IMAC, followed by “quick dilution” protein refolding. Refolding by “quick dilution” generated intact proteins free of aggregates, as determined by non-reducing SDS-PAGE. Final sample size and concentrations were as follows: IL-4BAD was about 3.5 mL at 0.24 mg/mL, determined by UV280 nm (UV280 nm Abs at 1 mg/ml=1.14); cpIL-4BAD was about 3.5 mL at 0.23 mg/mL, determined by UV280 nm (UV280 nm Abs at 1 mg/mL=1.13); and cpS4-BAD was about 3.5 mL at 0.23 mg/mL, determined by UV280 nm (UV280 nm Abs at 1 mg/ml=1.25). Protein was stored in a storage buffer composition: 500 nM NaCL, 10 mM Na-Phosphate, pH 7.0, 1% glycerol, 1 μM EDTA, 0.01% Tween 20.
  • BL21(DE3)pLysS-RARE2 cells were transformed with IL-4BAD, cp1L-4BAD and cpS4-BAD protein expression constructs, plated on LB plates supplemented with Amp at 100 μg/mL, and incubated overnight at 37° C. The next day, colonies from the plate were scraped and re-suspended in liquid LB medium with 100 μg/mL of Amp. The cultures were then grown at 37° C., with aeration, and protein expression was induced by 1 mM IPTG when the cell culture reached an OD600 of about 0.5. Induction lasted for about 4 hours at 30° C. The cell pellet was then collected and stored at −20° C. 10μ.l_, samples of uninduced and induced culture were lysed by boiling at 95° C. for 10 minutes in 50 uL of reducing protein loading buffer and run on an SDS-PAGE gel. Cells from a 1 mL sample collected at 4 hours post-induction were lysed in hypotonic buffer, sonicated and centrifuged for 10 minutes at 13,000 rpm. Aliquots from the soluble and insoluble fraction were boiled in reducing protein loading buffer and analyzed on an SDS-PAGE gel. Estimated expression levels observed for IL-4BAD, cp1L-4BAD proteins was more than 50 mg/L of crude material. Estimated expression levels observed for cpS4-BAD was more than 50 mg/L. IL-4BAD was found to be almost completely insoluble, with some possible soluble form (less than 5%). cpS4-BAD were mainly in the insoluble fraction.
  • The cell pellets from the induced cultures were lysed at room temperature and the inclusion bodies fraction was collected and washed with PBS-T. The insoluble material was solubilized in 8M Urea and bound to 3 mL Ni-charged resin. The resin was washed with 15 CV of wash buffer and the bound protein was eluted in 8 CV elutions of step gradient of imidazole in wash buffer. 7.5 μL from each fraction was analyzed on a SDS-PAGE gel. The fractions with the highest amount of IL-4BAD (about 6 mg) and cp1L-4BAD (about 8 mg) were combined and refolded. The remaining fractions were stored at −20° C.
  • The IL-4BAD and cp1L-4BAD fractions were combined, reduced by the addition of 1 mM DTT and subjected to a slow step-wise dialysis against storage buffer (150 mM NaCl, 10 mM HEPES, pH 7.4, 0.01% Tween 20) supplemented with a decreasing concentration of urea at each dialysis buffer change. The protein concentration was measured after each dialysis step by UV Spectroscopy: IL-4BAD was about 0.8 mg; and cp1L-4BAD was about 0.45 mg. The samples were then run on an SDS-PAGE gel.
  • 25 μL of each protein (−0.6 mg/mL, 200 mM Imidazole fraction) was diluted in 1 mL of the following buffers: 20 mM HEPES, pH 7.4, 1% glycerol, 10 μM EDTA, 0.01% Tween (Buffer 1); 10 mM Na-Phosphate, pH 7.0, 1% Glycerol, 10 μM EDTA, 0.01% Tween (Buffer 2); and PBS, pH 7.2 (Buffer 3). The samples were stored overnight at room temperature. The next day, the samples were spun for 10 minutes at 13,000 rpm and the presence of the pellet in each sample was observed and recorded in Table 2: (+++) indicates an abundant pellet; (−) indicates no visible pellet.
  • TABLE 2
    IL-4BAD and cplL-4BAD pellets in Buffers 1, 2 and 3.
    Pellet after Pellet after
    114-BAD refolding cpIL4-BAD, refolding
    10 mM Buffer 1 NaCI, 100 mM Sample 1 (+++) Sample 13 (+++)
    DTT NaCI, 500 mM Sample 2 (+++) Sample 14 (++)
    NaCI, 20 mM Sample 3 (++) Sample 15 (+++)
    Buffer 2 NaCI, 100 mM Sample 4 (−) Sample 16 (+++)
    NaCI, 500 mM Sample 5 (+) Sample 17 (++)
    NaCI, 20 mM Sample 6 (+++) Sample 18 (+++)
    Buffer 3 Sample 25 (++) Sample 27 (+++)
    w/o Buffer 1 NaCI, 100 mM Sample 7 (+) Sample 19 (++)
    reducing NaCI, 500 mM Sample 8 (+) Sample 20 (++)
    agents NaCI, 20 mM Sample 9 (−) Sample 21 (++)
    Buffer 1 NaCI, 100 mM Sample 10 (−) Sample 22 (++)
    NaCI, 500 mM Sample 11 (+) Sample 23 (+)
    NaCI, 20 mM Sample 12 (+) Sample 24 (+++)
    Buffer 3 Sample 26 (+) Sample 28 (−)
  • The samples were also analyzed on non-reducing SDS-PAGE gel. Refolded samples were concentrated to 100 μL using Amicon 10 kDa MWCO, spun down, and a 7.5 μL aliquot of each sample was run on an SDS-PAGE gel.
  • The purification process was repeated using quick dilution folding. 4 ml of 200 mM imidazole fractions containing IL-4BAD, cp1L-4BAD or cpS4-BAD was quickly diluted into 200 mL of refolding buffer (500 mM NaCl, 10 mM Na-Phosphate, pH 7.0 or 6.0 or 7.8, 1% Glycerol, 10[1.M EDTA, 0.01% Tween), incubated overnight at room temperature, spun down for 20 minutes at 4,000 rpm at 4° C., concentrated to 3 mL using an Amicon 10 kDa MWCO, and buffer exchanged into storage buffer (500 mM NaCl, 10 mM Na-Phosphate, pH 7.0 or 6.0 or 7.8, 1% Glycerol, 1 μM EDTA, 0.01% Tween) using a DG-10 column. Final sample concentrations were as follows: 3.5 mL of IL-4B AD at about 0.24 mg/mL; 3.5 mL of cp1L-4BAD at about 0.23 mg/mL. The final samples were run on an SDS-PAGE gel.
  • The final concentration of cpS4-BAD was about 3.5 mL at about 0.23 mg/mL. The final sample was run on an SDS-PAGE gel.
  • Example 5
  • A pKFR4-BAD-H6 fusion protein (FIGS. 11A-B) was prepared as follows. cDNA of pKFR4-BAD-H6 was PCR cloned into the Ndel/Xhol sites of a pET-21a(+) vector. The vector was then transformed into HMS174(DE3) cells and induced with 0.1 mM IPTG for 3 hours. Samples were run on an SDS-PAGE gel before and after induction. The cells were pelleted and lysed by ultrasonication in a buffer containing 20 mM Tris-HCl, 300 mM NaCl, 20 mM Imidazole, pH 8.0, and samples were run on an SDS-PAGE gel.
  • Inclusion bodies were dissolved in a dissolving buffer of 20 mM Tris-HCl, 300 mM NaCl, 20 mM Imidazole, 20 mM beta-ME, 7 M GuaHCl, pH 8.0. The supernatant was then purified by Ni2I affinity chromatography. pKFR4-BAD-H6 was eluted by 20 mM Tris-HCl, 300 mM NaCl, 300 mM Imidazole, 8 M Urea, pH 8.0 under reducing and non-reducing conditions. Samples were taken throughout the purification process.
  • After purification, pKFR4-BAD-H6 was dialyzed in dialysis buffer: 0.1% TFA, 30% acetonitrile under reducing conditions and non-reducing conditions, yielding a concentration of about 2.67 mg/mL.
  • Example 6
  • To evaluate the potency of fusion proteins on IL-4R-expressing cell lines, EH suspension cells that express Type I IL-4R, were cultured in RPMI1640 (from Gibco) containing 10% FBS (from Life Technologies), 2 mM L-glutamine (from Gibco) and 10 mM HEPES (from Gibco). cpS4-BAD was added to the cell suspension. The cell suspension was poured into a 96-well Isoplate in an amount of about 1×104 cells/well. The cell suspensions were incubated at 37° C. in a 5% CO2 incubator for 48 hours. Before the end of the 48-hour incubation, 100 μCi/mL of [3H]-thymidine in complete medium was prepared and 10 μL was added into each well of the culture. After 6 hours of incubation with [3I-1]-thymidine, 50 μL of 50% trichloroacetic acid was slowly added into each well and incubated at 4° C. for 2 hours. The plates were then washed five times with dH2O, and air-dried. 100 μL of scintillation liquid was added to each well and the plates were left at room temperature overnight. The next day, the radioactivity was read in a MicroBeta Trilux. Data was collected and standardized using the reading from the control well (cell only). The background reading (blank) was subtracted from the readings of all the wells, and the inhibition (% inhibition) was calculated. The IC50 of cpS4-BAD was about 126.3 ng/mL, which was about 3 times more potent than MDNA55 (cpIL4-PE; 362.4 ng/mL) which was used as a reference.
  • All citations are hereby incorporated by reference.
  • The present invention has been described with regard to one or more embodiments. However, it will be apparent to persons skilled in the art that a number of variations and modifications can be made without departing from the scope of the invention as defined in the claims.

Claims (29)

What is claimed is:
1. A fusion protein comprising an interleukin-4 (1L-4) receptor binding protein and a pro-apoptotic Bcl-2 family polypeptide
2. The fusion protein of claim 1 wherein the IL-4 receptor binding protein is circularly permuted (cp).
3. The fusion protein of claim 1 or 2 wherein the pro-apoptotic Bcl-2 family polypeptide comprises a BH3 domain.
4. The fusion protein of claim 3 wherein the pro-apoptotic Bcl-2 family polypeptide comprising a BH3 domain is Bad, Bik/Nbk, Bid, Bim/Bod, Hrk, Bak or Bax.
5. The fusion protein of claim 3 or 4 wherein the pro-apoptotic Bcl-2 family polypeptide comprising a BH3 domain further comprises a mutation that reduces phosphorylation.
6. The fusion protein of claim 5 wherein the pro-apoptotic Bel-2 family polypeptide comprising a BH3 domain that further comprises a mutation that reduces phosphorylation is a Bad polypeptide.
7. The fusion protein of any one of claims 1 to 6, wherein the fusion protein is capable of inhibiting cell survival, inhibiting cell proliferation, or enhancing cell death or apoptosis of a target cell expressing an IL-4 receptor (IL-4R).
8. The fusion protein of any one of claims 1 to 7 wherein the IL-4 receptor binding protein is a mutant IL-4 or IL-13 selective for binding to a Type I or a Type II IL-4R.
9. The fusion protein of claim 8 wherein the mutant IL-4 selective for binding to a Type II IL-4R comprises a KFR variant or a KF variant or the mutant IL-4 selective for binding to a Type I IL-4R comprises an RGA variant.
10. The fusion protein of claim 8 wherein the mutant IL-13 comprises an All variant or a DN variant.
11. The fusion protein of any one of claims 1 to 10 further comprising a linker.
12. The fusion protein of claim 11 wherein the linker has the sequence GS or is a ubiquitin or ubiquitin variant molecule.
13. The fusion protein of claim 1 or 2, comprising the amino acid sequence of any one of SEQ ID NOs: 24-27.
14. A nucleic acid molecule encoding the fusion protein of any one of claims 1 to 13.
15. A nucleic acid molecule comprising the nucleic acid sequence of any one of SEQ ID NOs: 35-38.
16. A vector comprising the nucleic acid molecule of claim 14 or 15.
17. A host cell comprising the vector of claim 16.
18. A pharmaceutical composition comprising the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, the vector of claim 16, or the host cell of claim 17.
19. A method of inducing cell death comprising administering: the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, the vector of claim 16, or the host cell of claim 17, to a subject in need thereof.
20. A method of inducing cell death comprising contacting a target cell that expresses an IL-4R with the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, or the vector of claim 16.
21. A method of treating cancer comprising administering: the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, the vector of claim 16, or the host cell of claim 17, to a subject in need thereof
22. A method of treating cancer comprising contacting a neoplastic cell that expresses an IL-4R with the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, or the vector of claim 16.
23. A method of treating cancer comprising contacting a non-malignant cell that expresses an IL-4R in a tumour microenvironment in a subject in need thereof with the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, the vector of claim 16, or the host cell of claim 17.
24. The method of claim 23 wherein the non-malignant cell is contacted prior to said subject starting a therapy.
25. A method of treating a hyperproliferative or differentiative disorder comprising administering: the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, the vector of claim 16, or the host cell of claim 17, to a subject in need thereof.
26. The method of claim 25 wherein the hyperproliferative or differentiative disorder is a fibrosis or hyperplasia, an inflammatory conditions or an autoimmune condition.
27. The method of claim 26 wherein the fibrosis or hyperplasia is pulmonary fibrosis or hyperplasia (such as benign prostatic hyperplasia), cardiac fibrosis, or liver fibrosis; the inflammatory condition is prostatitis, vernal keratoconjunctivitis, artherosclerosis, or idiopathic pulmonary pneumonia; and the autoimmune condition is Graves disease.
28. Use of the fusion protein of any one of claims 1 to 13, the nucleic acid molecule of claim 14 or 15, the vector of claim 16, or the host cell of claim 17, for inducing cell death or treating cancer or treating a hyperproliferative or differentiative disorder in a subject in need thereof.
29. The method of claim 19, 21, 23-27 or the use of claim 28, wherein the subject is a human.
US17/397,287 2013-09-24 2021-08-09 Interleukin-4 receptor-binding fusion proteins and uses thereof Abandoned US20220048965A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/397,287 US20220048965A1 (en) 2013-09-24 2021-08-09 Interleukin-4 receptor-binding fusion proteins and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361881930P 2013-09-24 2013-09-24
PCT/CA2014/050915 WO2015042705A1 (en) 2013-09-24 2014-09-24 Interleukin-4 receptor-binding fusion proteins and uses thereof
US201615024785A 2016-03-24 2016-03-24
US16/125,075 US11084856B2 (en) 2013-09-24 2018-09-07 Interleukin-4 receptor-binding fusion proteins and uses thereof
US17/397,287 US20220048965A1 (en) 2013-09-24 2021-08-09 Interleukin-4 receptor-binding fusion proteins and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/125,075 Division US11084856B2 (en) 2013-09-24 2018-09-07 Interleukin-4 receptor-binding fusion proteins and uses thereof

Publications (1)

Publication Number Publication Date
US20220048965A1 true US20220048965A1 (en) 2022-02-17

Family

ID=52741678

Family Applications (6)

Application Number Title Priority Date Filing Date
US15/024,785 Active 2035-01-12 US10093708B2 (en) 2013-09-24 2014-09-24 Interleukin-4 receptor-binding fusion proteins and uses thereof
US15/024,787 Active 2035-01-01 US10106592B2 (en) 2013-09-24 2014-09-24 Interleukin-4 receptor-binding fusion proteins and uses thereof
US16/125,566 Active US11352402B2 (en) 2013-09-24 2018-09-07 Interleukin-4 receptor-binding fusion proteins and uses thereof
US16/125,075 Active US11084856B2 (en) 2013-09-24 2018-09-07 Interleukin-4 receptor-binding fusion proteins and uses thereof
US17/397,287 Abandoned US20220048965A1 (en) 2013-09-24 2021-08-09 Interleukin-4 receptor-binding fusion proteins and uses thereof
US17/738,620 Pending US20220372096A1 (en) 2013-09-24 2022-05-06 Interleukin-4 Receptor-Binding Fusion Proteins and Uses Thereof

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US15/024,785 Active 2035-01-12 US10093708B2 (en) 2013-09-24 2014-09-24 Interleukin-4 receptor-binding fusion proteins and uses thereof
US15/024,787 Active 2035-01-01 US10106592B2 (en) 2013-09-24 2014-09-24 Interleukin-4 receptor-binding fusion proteins and uses thereof
US16/125,566 Active US11352402B2 (en) 2013-09-24 2018-09-07 Interleukin-4 receptor-binding fusion proteins and uses thereof
US16/125,075 Active US11084856B2 (en) 2013-09-24 2018-09-07 Interleukin-4 receptor-binding fusion proteins and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/738,620 Pending US20220372096A1 (en) 2013-09-24 2022-05-06 Interleukin-4 Receptor-Binding Fusion Proteins and Uses Thereof

Country Status (6)

Country Link
US (6) US10093708B2 (en)
EP (3) EP3049525A4 (en)
JP (4) JP6936934B2 (en)
CN (4) CN113563478A (en)
CA (3) CA3180148A1 (en)
WO (2) WO2015042706A1 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2925421C (en) * 2013-09-24 2023-08-29 Medicenna Therapeutics, Inc. Interleukin-2 fusion proteins and uses thereof
WO2015042706A1 (en) * 2013-09-24 2015-04-02 Medicenna Therapeutics Pte Ltd Interleukin-4 receptor-binding fusion proteins and uses thereof
KR102648293B1 (en) 2014-05-21 2024-03-15 우니페르시테트 바젤 Bacteria-based protein delivery
KR101741594B1 (en) * 2015-06-30 2017-05-30 경북대학교 산학협력단 Pharmaceutical composition comprising fusion peptide targeting cancer cells and tumor associated macrophages for treating cancer and inhibiting metastasis
WO2017001568A1 (en) 2015-07-01 2017-01-05 Universitat De Lleida Treatment and prevention of amyotrophic lateral sclerosis
EP3325505B1 (en) * 2015-07-23 2020-08-19 Modernatx, Inc. Messenger ribonucleic acids for the production of intracellular binding polypeptides and methods of use thereof
PT3377094T (en) 2015-11-19 2021-07-08 Univ Basel Virulence attenuated bacteria for treatment of malignant solid tumors
AU2016355975B2 (en) * 2015-11-19 2022-12-08 Universität Basel Bacteria-based protein delivery
HUE055832T2 (en) 2016-12-20 2021-12-28 Univ Basel Virulence attenuated bacteria based protein delivery
WO2019052562A1 (en) * 2017-09-15 2019-03-21 科济生物医药(上海)有限公司 Fusion protein of il-4r and use thereof
CN114457024A (en) * 2020-10-30 2022-05-10 未来智人再生医学研究院(广州)有限公司 Pluripotent stem cell expressing IL-4R alpha blocker or derivative thereof and application
WO2023170475A2 (en) * 2022-03-08 2023-09-14 Medicenna Therapeutics, Inc. Uses and methods for il-2, il-13, and il-4 cytokine bifunctional molecules

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008101671A2 (en) * 2007-02-19 2008-08-28 Apogenix Gmbh Il-4 fc fusion proteins

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5501959A (en) 1989-01-17 1996-03-26 Alamar Biosciences Laboratory, Inc. Antibiotic and cytotoxic drug susceptibility assays using resazurin and poising agents
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
AU5010793A (en) 1992-08-21 1994-03-15 Schering Corporation Human interleukin-13
US5635599A (en) 1994-04-08 1997-06-03 The United States Of America As Represented By The Department Of Health And Human Services Fusion proteins comprising circularly permuted ligands
DE4423131A1 (en) 1994-07-01 1996-01-04 Bayer Ag New hIL-4 mutant proteins as antagonists or partial agonists of human interleukin 4
FR2742156A1 (en) 1995-12-06 1997-06-13 Sanofi Sa IL-13 RECEPTOR POLYPEPTIDE
US6335426B1 (en) 1996-06-14 2002-01-01 Bayer Corporation T-cell selective interleukin-4 agonists
US6028176A (en) 1996-07-19 2000-02-22 Bayer Corporation High-affinity interleukin-4 muteins
US6410046B1 (en) 1996-11-19 2002-06-25 Intrabrain International Nv Administering pharmaceuticals to the mammalian central nervous system
AU2001288405B8 (en) 1998-04-03 2007-06-14 The Penn State Research Foundation Amino acid substitution mutants of interleukin 13
US6737511B1 (en) 1999-08-16 2004-05-18 The United States Of America As Represented By The Department Of Health And Human Services Receptor-mediated uptake of an extracellular BCL-xL fusion protein inhibits apoptosis
MXPA02002684A (en) 1999-09-09 2002-07-30 Schering Corp Mammalian interleukin 12 p40 and interleukin b30, combinations thereof, antibodies, uses in pharmaceutical compositions.
WO2001025282A1 (en) 1999-10-06 2001-04-12 The Penn State Research Foundation Il13 mutants
WO2001034645A2 (en) * 1999-11-11 2001-05-17 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Mutated il-13 molecules and their uses
GB0004016D0 (en) 2000-02-22 2000-04-12 Royal Brompton Hospital Biological material and uses thereof
US7087224B2 (en) 2000-10-31 2006-08-08 Amgen Inc. Method of treating anemia by administering IL-1ra
AU2002240312A1 (en) 2001-02-08 2002-08-19 Pankaj Paranjp Enhanced oral and transcompartmental delivery of therapeutic or diagnostic agents using polymer conjugates
US20030013851A1 (en) 2001-06-07 2003-01-16 Robert Powers Solution structure of IL-13 and uses thereof
US20040248260A1 (en) 2001-10-26 2004-12-09 Heavner George A. IL-13 mutein proteins, antibodies, compositions, methods and uses
US20050027110A1 (en) 2003-07-24 2005-02-03 Michael Russell Drug delivery in the nervous system
JP2008526889A (en) * 2005-01-10 2008-07-24 リサーチ ディベロップメント ファウンデーション Targeted chimeric molecules for cancer therapy
AU2006347606B2 (en) 2005-09-09 2012-10-11 Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods and compositions for inhibiting cell death or enhancing cell proliferation
US20070160658A1 (en) 2005-10-20 2007-07-12 The Penn State Research Foundation Delivery system for diagnostic and therapeutic agents
JP2010536386A (en) * 2007-08-24 2010-12-02 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Receptor targeting reagent
CN104059155A (en) * 2008-05-16 2014-09-24 拜耳医药保健有限公司 Targeted Coagulation Factors And Method Of Using The Same
US20110171219A1 (en) 2008-09-19 2011-07-14 Fahar Merchant Treating cancer stem cells using targeted cargo proteins
US20100183545A1 (en) * 2008-10-14 2010-07-22 The United States Of America, As Represented By The Secretary, Targeted cargo protein combination therapy
EP2358754A1 (en) 2008-11-26 2011-08-24 Glaxo Group Limited Ligands that bind il-13
US20110023680A1 (en) 2009-07-30 2011-02-03 Kuo-Chung Wang Pcb cutter module with detachable cutters
WO2011106779A1 (en) 2010-02-26 2011-09-01 Aerovance Inc. Use of modified il-4 mutien receptor antagonists to treat dermatitis
WO2012054929A2 (en) * 2010-10-22 2012-04-26 Protox Therapeutics Corp. Use of human serum albumin to decrease antigenicity of therapeutic proteins
WO2012139112A1 (en) 2011-04-08 2012-10-11 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Ubiquitin fusions for improving the efficacy of cytosolic acting targeted toxins
WO2013112871A1 (en) * 2012-01-27 2013-08-01 The Board Of Trustees Of The Leland Stanford Junior University Therapeutic il-13 polypeptides
WO2015042706A1 (en) * 2013-09-24 2015-04-02 Medicenna Therapeutics Pte Ltd Interleukin-4 receptor-binding fusion proteins and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008101671A2 (en) * 2007-02-19 2008-08-28 Apogenix Gmbh Il-4 fc fusion proteins

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
Bhattacharya et al. Impact of genetic variation on three dimensional structure and function of proteins. PLoS ONE 12(3): e0171355, 2017 *
Bork, P. Go hunting in sequence databases but watch out for the traps. Trends in Genetics 12(10): 425-427, 1996 *
Bork, P. Powers and pitfalls in sequence analysis: the 70% hurdle. Genome Res 10: 398-400, 2000 *
Brenner. S.E. Errors in genome annotation. Trends in Genetics 15:132-133, 1999 *
Chen et al. Fusion protein linkers: property, design, and functionality. Adv Drug Rev 65: 1357-1369, 2013 (online 29 September 2012) *
Doerks et al. Protein annotation: detective work for function prediction. Trends in Genetics 14:248-250, 1998 *
Fenton et al. Rheostat positions: a new classification of protein positions relevant to pharmacogenomics. Medicinal Chem Res 29: 1133-1146, 2020 *
Gardai et al. Phosphorylation of Bax Ser184 by Akt regulates its activity and apoptosis in neutrophils. J Biol Chem 279(20): 21085-21095, 2004 *
Guo et al. Protein tolerance to random amino acid change. Proc Natl Acad Sci USA 101(25): 9205-9210, 2004 *
Ngo et al. "Computational complexity, protein structure prediction, and the Levinthal paradox" in The Protein Folding Problem and Tertiary Structure Prediction, pp. 492-495, 1994 *
Skolnick et al. From genes to protein structure and function: novel applications of computational approaches in the genomic era. Trends Biotechnol 18(l):34-39 2000 *
Smith et al. The challenges of genome sequence annotation or "the devil is in the details". Nature Biotechnol 15: 1222-1223, 1997 *
Tokuriki et al. Stability effects of mutations and protein evolvability. Curr Opin Structural Biol 19: 596-604, 2009 *
Wang et al. Mono- or double-ste phosphorylation distinctly regulates the proapoptotic function of Bax. PLoS One 5(10): e13393, 2010 (8 total pages) *
Wells, J.A. Additivity of mutational effects in proteins. Biochemistry 29(37): 8509-8517, 1990 *

Also Published As

Publication number Publication date
EP3049525A4 (en) 2017-05-03
EP3049525A1 (en) 2016-08-03
US20220372096A1 (en) 2022-11-24
JP6936934B2 (en) 2021-09-22
US20160215035A1 (en) 2016-07-28
US10093708B2 (en) 2018-10-09
CA3180148A1 (en) 2015-04-02
JP2023052641A (en) 2023-04-11
JP2020010701A (en) 2020-01-23
US20190100568A1 (en) 2019-04-04
JP2016533166A (en) 2016-10-27
CA2930074A1 (en) 2015-04-02
US11352402B2 (en) 2022-06-07
CN105764926A (en) 2016-07-13
EP3049526A4 (en) 2017-10-11
EP3049526A1 (en) 2016-08-03
US20160237135A1 (en) 2016-08-18
US20190100567A1 (en) 2019-04-04
CN116041551A (en) 2023-05-02
US11084856B2 (en) 2021-08-10
EP3910061A1 (en) 2021-11-17
EP3049526B1 (en) 2021-02-24
CN113563478A (en) 2021-10-29
JP6661530B2 (en) 2020-03-11
CA2925417A1 (en) 2015-04-02
WO2015042706A1 (en) 2015-04-02
US10106592B2 (en) 2018-10-23
CN105722982A (en) 2016-06-29
CA2925417C (en) 2023-10-24
JP2016533343A (en) 2016-10-27
WO2015042705A1 (en) 2015-04-02

Similar Documents

Publication Publication Date Title
US20220048965A1 (en) Interleukin-4 receptor-binding fusion proteins and uses thereof
US11680090B2 (en) Interleukin-2 fusion proteins and uses thereof
JP2021506849A (en) Tumor homing and cell-penetrating peptide immune anti-cancer drug complexes, and how to use them
KR102102789B1 (en) Disintegrin variants and pharmaceutical uses thereof
Karageorgis et al. Systemic delivery of tumor-targeted Bax-derived membrane-active peptides for the treatment of melanoma tumors in a humanized SCID mouse model
Jeon et al. Targeted cancer therapy using fusion protein of TNFα and tumor-associated fibronectin-specific aptide
JP2022104932A (en) Chimeric protein for targeting dsRNA
CA3096978A1 (en) Compositions for use in lysis of selective cancer cells
KR20200107842A (en) Active TRAIL trimer and tumor targeting peptide multi-displayed on ferritin nanocage and use in anti-cancer agent thereof
US20180353627A1 (en) Radiolabeled disintegrins as brachytherapy agents

Legal Events

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PURI, RAJ K.;JOSHI, BHARATKUMAR H.;SIGNING DATES FROM 20210125 TO 20210228;REEL/FRAME:062094/0024

Owner name: MEDICENNA THERAPEUTICS, INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MERCHANT, FAHAR;REEL/FRAME:062093/0983

Effective date: 20160321

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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