US20130108661A1 - Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells - Google Patents

Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells Download PDF

Info

Publication number
US20130108661A1
US20130108661A1 US13/577,295 US201113577295A US2013108661A1 US 20130108661 A1 US20130108661 A1 US 20130108661A1 US 201113577295 A US201113577295 A US 201113577295A US 2013108661 A1 US2013108661 A1 US 2013108661A1
Authority
US
United States
Prior art keywords
tumor
ligand
cell
fusion protein
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
US13/577,295
Other languages
English (en)
Inventor
Julie Magarian Blander
Johan Garaude
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.)
Icahn School of Medicine at Mount Sinai
Original Assignee
Mount Sinai School of Medicine
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 Mount Sinai School of Medicine filed Critical Mount Sinai School of Medicine
Priority to US13/577,295 priority Critical patent/US20130108661A1/en
Assigned to MOUNT SINAI SCHOOL OF MEDICINE reassignment MOUNT SINAI SCHOOL OF MEDICINE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GARAUDE, JOHAN, BLANDER, JULIE MAGARIAN
Publication of US20130108661A1 publication Critical patent/US20130108661A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464499Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5152Tumor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6068Other bacterial proteins, e.g. OMP
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule

Definitions

  • the present invention is related to methods for inducing an anti-tumor immune response by immunizing a mammal with a composition comprising a tumor cell which expresses a Nod-like receptor (NLR) and/or a Toll-like receptor (TLR) ligand-tumor associated antigen (TAA) fusion protein or with an activated DC which has internalized a tumor cell which expresses an NLR- and/or TLR ligand-TAA fusion protein.
  • NLR Nod-like receptor
  • TLR Toll-like receptor
  • TAA ligand-tumor associated antigen
  • Multicellular organisms have developed two general systems of immunity to infectious agents.
  • the two systems are innate or natural immunity (usually referred to as “innate immunity”) and adaptive (acquired) or specific immunity.
  • innate immunity innate or natural immunity
  • adaptive (acquired) or specific immunity The major difference between the two systems is the mechanism by which they recognize infectious agents.
  • Recent studies have demonstrated that the innate immune system plays a crucial role in the control of initiation of the adaptive immune response and in the induction of appropriate cell effector responses (Fearon et al. Science 1996;272:50-53 and Medzhitov et al. Cell 1997;91:295-298).
  • the innate immune system uses a set of germline-encoded receptors for the recognition of conserved molecular patterns present in microorganisms. These molecular patterns occur in certain constituents of microorganisms including: lipopolysaccharides, peptidoglycans, lipoteichoic acids, phosphatidyl cholines, bacterial proteins (e.g., flagellin), including lipoproteins, bacterial DNAs, viral single and double-stranded RNAs, unmethylated CpG-DNAs, mannans, and a variety of other bacterial and fungal cell wall components. Such molecular patterns can also occur in other molecules such as plant alkaloids.
  • PAMPs Pathogen Associated Molecular Patterns
  • PRRs Pattern Recognition Receptors
  • TLR3, collectins Some of these receptors recognize PAMPs directly (e.g., TLR3, collectins), while others (e.g., complement receptors) recognize the products generated by PAMP recognition.
  • Cellular PRRs are expressed on effector cells of the innate immune system, including cells that function as professional antigen-presenting cells (APC) in adaptive immunity.
  • effector cells include, but are not limited to, macrophages, dendritic cells, B lymphocytes, and surface epithelia.
  • This expression profile allows PRRs to directly induce innate effector mechanisms, and also to alert the host organism to the presence of infectious agents by inducing the expression of a set of endogenous signals, such as inflammatory cytokines and chemokines. This latter function allows efficient mobilization of effector forces to combat the invaders.
  • PRRs include Nod-like receptors (NLRs) and Toll-like receptors (TLRs).
  • NLRs are cytoplasmic proteins that may have a variety of functions in regulation of inflammatory and apoptotic responses. NLRs are composed of conserved “modules” including a central nucleotide-binding oligomerization domain and a series of tandem leucine-rich repeats. NLRs are encoded by genes from a large gene family present in many different animal species; there are more than 20 NLR genes in humans. Many are thought to serve as PRRs which sense microbial products in the cytoplasm of cells, although some members have different functions. The ligands'are currently known for the NLRs, NLRC1 (NOD1) and NLRC2 (NOD2).
  • NLRC1 recognizes a molecule called Meso-diaminopimelic acid (meso-DAP), which is a peptidoglycan constituent of only Gram negative bacteria.
  • NLRC2 proteins recognize intracellular MDP (muramyl dipeptide), which is a peptidoglycan constituent of both Gram positive and Gram negative bacteria. These proteins transduce signals in the pathway of NF- ⁇ B and MAP kinases.
  • NLRC4 has also been shown to activate caspase-1 in response to bacteria. Further, anthrax toxin activates NLRP1 (previously called NALP1), and Staphylococcus aureus toxins such as alpha-hemolysin (GenBank Accession No.
  • AAA26598 (SEQ ID NO: 1) activate NLRP3.
  • Other NLRs such as NAIP have also been shown to activate caspase-1 in response to Salmonella and Legionella .
  • SEE Inohara et al., NOD-LRR proteins: role in host-microbial interactions and inflammatory disease. Annu Rev Biochem. 2005, Volume 74:355-83; Strober et al., Signalling pathways and molecular interactions of NOD1 and NOD2. Nat Rev Immunol. 2006, Volume 6(1):9-20; Chen G, Shaw M H, Kim Y G, Nu ⁇ ez G. Annu Rev Pathol. 2009, 4:365-98; Martinon F, Mayor A, Tschopp J. Annu Rev Immunol. 2009;27:229-65].
  • TLRs Toll-like receptors
  • TLR4 can induce the secretion of tumor necrosis factor (TNF) and of the interleukins IL-1 and IL-6 as part of an antibacterial response, and can induce the secretion of the interferons INF ⁇ and INF ⁇ as part of an anti viral response.
  • TNF tumor necrosis factor
  • TLR signaling consists of at least two distinct pathways: a MyD88-dependent pathway that leads to the production of inflammatory cytokines, and a MyD88-independent pathway associated with the stimulation of IFN- ⁇ and the maturation of dendritic cells.
  • the MyD88-dependent pathway is common to all TLRs, except TLR3 [Adachi O. et al., 1998. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity. 9(1):143-50.].
  • TLRs Upon activation by microbial antigens, TLRs induce the recruitment of MyD88 via its T1R domain which in turn recruits IRAK1 and IRAK4 and leads to complex downstream signaling cascades leading to the phosphorylation of I ⁇ B and the subsequent nuclear localization of NF- ⁇ B. Activation of NF- ⁇ B triggers the production of pro-inflammatory cytokines such as TNF- ⁇ , IL-1 and IL-12.
  • TLRs have been shown to recognize PAMPs such as the bacterial products LPS (Schwandner et al. J. Biol. Chem. 1999;274:17406-9 and Hoshino et al. J. Immunol 1999;162:3749-3752), lipoteichoic acid (Schwandner et al. J. Biol. Chem. 1999;274:17406-9), peptidoglycan (Yoshimura et al. J. Immunol. 1999;163:1-5), lipoprotein (Aliprantis et al. Science 1999;285:736-9), CpG-DNA (Hemmi et al.
  • TLR2 is essential for the recognition of a variety of PAMPs, including bacterial lipoproteins, peptidoglycan, and lipoteichoic acids.
  • TLR3 is implicated in virus-derived double-stranded RNA.
  • TLR4 is predominantly activated by lipopolysaccharide.
  • TLR9 is required for response to unmethylated CpG DNA. Recently, TLR7 and TLR8 have been shown to recognize single stranded RNA molecules (Hornung V. et al. Handb Exp Pharmacol. 2008;(183):71-86), and small synthetic antiviral molecules (Jurk M. et al. Nat Immunol 2002;3:499). TLR11 detects profilin-like protein (PLP). Furthermore, TLR5 detects bacterial flagellin.
  • Flagellin is a protein expressed by a variety of flagellated bacteria ( Salmonela typhimurium for example) as well as non-flagellated bacteria (such as Escherichia coli ). Sensing of flagellin by cells of the innate immune system (dendritic cells, macrophages, etc) is mediated by the Toll-like receptor 5 (TLR5) as well as by Nod-like receptors (NLRs) Ipaf and Naip5 (Franchi et al (2006) Nat Immunol 7(6):576-582; Miao et al (2006) Nat Immunol 7(6):569-575; and Ren et al (2006) PLoS Pathog 2(3):e18).
  • TLR5 Toll-like receptor 5
  • NLRs Nod-like receptors
  • Bacillus anthracis is the bacterium that causes anthrax.
  • the bacterium secretes a toxin called anthrax lethal toxin, which is the major cause of pathogenesis, and is composed of a protective antigen and a lethal factor (Stephen, J. Anthrax toxin. 1981. Pharmacol. Ther. 12, 501-513).
  • lethal factor component of anthrax toxin enters the cytosol of macrophages and other cell types, and is recognized by the NLR protein Nalp1 or NLRP1 and mediates cell death (Boyden et al. 2006 Nat Genet 38:240-244).
  • Staphylococcus aureus is a Gram positive bacterium responsible for a wide variety of superficial as well as serious life-threatening infections (Lowy et al. 1998. N. Engl. J. Med. 339: 520-525). S. aureus secretes many toxins among which ⁇ -hemolysin has been implicated in the pathogenesis of S. aureus necrotizing pneumonia and various other symptoms in animal models. ⁇ -Hemolysin is secreted as a 33-kDa monomer and oligomerizes, forming heptameric transmembrane pores (Song et al. 1996 Science 274: 1859-1866).
  • TLR ligands have been exploited as adjuvant in numerous therapy regimens [Koski, G. K. et al., Reengineering dendritic cell-based anti-cancer vaccines. Immunol Rev 222, 256 (2008)].
  • BCG live bacilli Calmette-Guerin
  • TLR2 and TLR4 have been proven to be beneficial in the treatment of tumors such as bladder cancer
  • tumors such as bladder cancer
  • Imiquimod an agonist for TLR7
  • TLR9 ligand CpG has also been used in different mono-therapies, combination therapies and Phase I/II trials [Dougan, M. and Dranoff, G., Immune Therapy for Cancer. Annu Rev Immunol (2008)].
  • TLR ligands in conjunction with long peptides containing helper and cytotoxic T lymphocytes (CTL) epitopes, has shown to be efficient at promoting helper CD4+ T cells [Melief, C. J. et al., Effective therapeutic anticancer vaccines based on precision guiding of cytolytic T lymphocytes. Immunol Rev 188, 177 (2002); Jackson, D. C. et al., A totally synthetic vaccine of generic structure that targets Toll-like receptor 2 on dendritic cells and promotes antibody or cytotoxic T cell responses. Proc Natl Acad Sci USA 101 (43), 15440 (2004)].
  • CTL cytotoxic T lymphocytes
  • TLR ligands many adjuvants used in human vaccine also include ligands for NLRs and can activate DCs [Martinon, F., Mayor, A., and Tschopp, J., The inflammasomes: guardians of the body. Annu Rev Immunol 27, 229 (2009)]. Further, recently it has been discovered that TLR ligands enhance presentation of phagocytosed antigens within major histocompatibility class II MHC molecules [Blander, J. M. and Medzhitov, R., Nature (2006), Vol. 440, pp. 808].
  • TLR Toll-like receptor
  • mice immunized with a fusion protein consisting of the polypeptide PAMP BLP linked to Leishmania major antigens mounted a Type 1 immune response characterized by antigen-induced production of ⁇ -interferon and antigen-specific IgG2a (Cote-Sierra et al. Infect Immun 2002;70:240-248).
  • the response was protective, as demonstrated in experiments in which immunized mice developed smaller lesions than control mice did following challenge with live L. major .
  • flagellin fusion to well defined antigens promotes protective immunity in mice [Huleatt, J. W. et al., in Vaccine (2007), Vol. 25, pp. 763; Huleatt, J. W.
  • CD8 ⁇ T cells such as cytotoxic T lymphocytes (CTLs)
  • CTLs cytotoxic T lymphocytes
  • Th CD4 T helper
  • Th cells can also contribute to anti-tumor activity through direct killing of tumors, by supporting both the activation and long-term maintenance of CD8+ T cells, and through the production of cytokines.
  • Th cells can also support the humoral immune response mediated by B cells [Koski et al. (2008) supra].
  • Immunotherapy if successful, would be particularly appealing for use as a cancer treatment, for which new and better treatments are urgent needed.
  • chemotherapeutic agents include countless instances occur either where tumors are not amenable to any existing therapy or they respond initially only to recur in forms resistant to front-line therapies, leaving limited treatment options.
  • immunotherapies would be suitable to prevent relapse.
  • DCs dendritic cells
  • the present invention provides a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • NLR Nod-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand, a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • NLR Nod-like receptor
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • NLR Nod-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand, a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for treating a cancer in a patient comprising administering to said patient in need of such treatment a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA), wherein said composition is administered in an effective amount for eliciting an anti-tumor immune response.
  • DC dendritic cell
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for treating a cancer in a patient comprising administering to said patient in need of such treatment a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand and a tumor-associated antigen (TAA), wherein said composition is administered in an effective amount for eliciting an anti-tumor immune response.
  • DC dendritic cell
  • NLR Nod-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand, a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA).
  • DC dendritic cell
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the present invention provides a method for treating a cancer in a patient comprising administering to said patient in need of such treatment a composition comprising a dendritic cell (DC), wherein said DC has internalized a tumor cell expressing a fusion protein, said fusion protein comprising a Nod-like receptor (NLR) ligand, a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA), wherein said composition is administered in an effective amount for eliciting an anti-tumor immune response.
  • DC dendritic cell
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a tumor cell expressing a fusion protein, wherein said fusion protein comprises a TLR ligand and a tumor-associated antigen (TAA).
  • TAA tumor-associated antigen
  • the invention provides a method for treating a cancer in a patient comprising administering to said patient in need of such treatment a composition comprising a tumor cell expressing a fusion protein, wherein said fusion protein comprises a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA), in an effective amount for eliciting an anti-tumor immune response.
  • a composition comprising a tumor cell expressing a fusion protein, wherein said fusion protein comprises a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA), in an effective amount for eliciting an anti-tumor immune response.
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a tumor cell expressing a fusion protein, wherein said fusion protein comprises a Nod-like receptor (NLR) ligand and a tumor-associated antigen (TAA).
  • NLR Nod-like receptor
  • TAA tumor-associated antigen
  • the invention provides a method for treating a cancer in a patient comprising administering to said patient in need of such treatment a composition comprising a tumor cell expressing a fusion protein in an effective amount for eliciting an anti-tumor immune response, wherein said fusion protein comprises a Nod-like receptor (NLR) ligand and a tumor-associated antigen (TAA).
  • NLR Nod-like receptor
  • TAA tumor-associated antigen
  • the invention provides a method for inducing an anti-tumor immune response in a mammal comprising administering to said mammal in need thereof an immunogenically effective amount of a composition comprising a tumor cell expressing a fusion protein, wherein said fusion protein comprises a fusion protein, wherein said fusion protein comprises a said fusion protein comprising a Nod-like receptor (NLR) ligand, a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA).
  • NLR Nod-like receptor
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the invention provides a method for treating a cancer in a patient comprising administering to said patient in need of such treatment a composition comprising a tumor cell expressing a fusion protein, wherein said fusion protein comprises a said fusion protein comprising a Nod-like receptor (NLR) ligand, a Toll-like receptor (TLR) ligand and a tumor-associated antigen (TAA), in an effective amount for eliciting an anti-tumor immune response.
  • NLR Nod-like receptor
  • TLR Toll-like receptor
  • TAA tumor-associated antigen
  • the TLR ligand is a polypeptide. In certain of the above embodiments, the TLR ligand is a flagellin or profilin-like protein (PLP), or a fragment thereof.
  • PLP profilin-like protein
  • the tumor cell has been transfected with a vector expressing said fusion protein.
  • the DC is an autologous cell.
  • the tumor cell is an autologous cell.
  • the tumor cell is lethally irradiated prior to internalization by said DC.
  • the DC has phagocytosed said tumor cell.
  • the anti-tumor immune response comprises a CD4 or CD8 T cell-mediated immune response.
  • the mammal or patient is a human.
  • the NLR ligand is selected from the group consisting of a flagellin, an anthrax toxin, and a Staphylococcus aureus toxin, or a fragment thereof.
  • the TLR ligand is also an NLR ligand. In other embodiments, the NLR ligand is also a TLR ligand. In some aspects of the invention, the fusion protein comprising a TAA and a TLR ligand further comprises a distinct NLR ligand. In other aspects of the invention, the fusion protein comprising a TAA and an NLR ligand further comprises a distinct TLR ligand.
  • the TLR ligand is profilin-like protein (PLP) and said NLR ligand is anthrax toxin or a fragment thereof.
  • PLP profilin-like protein
  • the tumor cell is lethally irradiated.
  • the TLR ligand is profilin-like protein (PLP) and said NLR ligand is the Staphylococcus aureus ⁇ -hemolysin, or a fragment thereof.
  • FIG. 1A is a graph showing a time course (days post injection) of tumor volume (mm 3 ) in wild type (wt) mice injected with 1 ⁇ 10 5 EL4 thymoma cells engineered to express either an ovalbumin (OVA) construct or an OVA- S. typhimurium flagellin fusion protein (STFOVA) using retroviral transduction with pMIG-IRES-GFP.
  • OVA ovalbumin
  • STFOVA OVA- S. typhimurium flagellin fusion protein
  • EL4-OVA thymoma cells were injected mice were injected in conjunction with recombinant flagellin (RecFLA) where tumor cells and flagellin were administered together but as separate entities.
  • RecFLA recombinant flagellin
  • FIG. 1B is a graph showing a time course (days post injection) of tumor volume (mm 3 ) in MyD88 knockout (Myd88 ⁇ / ⁇ ) mice injected with 1 ⁇ 10 5 EL4 thymoma cells engineered to express either an ovalbumin (OVA) construct or an OVA- S. typhimurium flagellin fusion protein (STFOVA) using retroviral transduction with pMIG-IRES-GFP.
  • OVA ovalbumin
  • STFOVA OVA- S. typhimurium flagellin fusion protein
  • FIG. 2 is a graph showing a time course (days post-transplantation) of tumor volume (mm 3 ) in wild-type (wt) or Rag ⁇ / ⁇ mice transplanted with 1 ⁇ 10 5 EL4-OVA or EL4-STFOVA tumor cells.
  • FIG. 3A shows two panels of flow cytometry dot plots showing peritoneal cells immunostained at 16 hours for CD45, F4/80, and CD11b in the indicated groups of mice, immunized with PBS, EL4-STFOVA or EL4-OVA, and treated with PBS or clodronate/liposome.
  • the upper panel shows cells stained for CD45. Tumor cells are CD45 + GFP + cells.
  • the lower panel of dot plots shows CD11b and F4/80 stained cells. Macrophages are CD11b + F4/80 + cells.
  • FIG. 4 is a dot plot showing proliferation of CFSE-labeled OVA-specific OT-II transgenic CD4 + T cells in tumor-draining lymph nodes at day 5 after tumor transplantation. Cells were stained with Thy1.2 and CD4 for analysis. The percentage of proliferating cells, demonstrated as cells with lower CFSE fluorescence intensity, in each of the indicated groups is shown.
  • FIG. 5A is a panel of flow cytometry dot plots showing proliferation of OVA-specific CD8 + OT-I T cells in tumor draining lymph node at day 3 after tumor transplantation. Cells were stained with Thy1.1 for analysis. The percentage of proliferating cells (boxed cells) in each of the indicated groups is shown.
  • mice received a concomitant injection of 1 ng recombinant flagellin (RecFLA).
  • FIG. 5B is a graph showing the percentage (%) of IFN- ⁇ and granzyme-B producing CD8 + OT-I T cells (from FIG. 5A ) measured by intracellular cytokine staining. Statistical significance (p value) is indicated on the graph.
  • FIG. 5C is a panel of flow cytometry histograms showing proliferation of OVA-specific CD8 + OT-I T cells in tumor draining lymph node at day 3 after tumor transplantation in wild-type (wt) mice or CD11c-DTR mice treated (+) or not ( ⁇ ) with 2 injections of 100 ng diphtheria toxin (DT) to deplete DC.
  • CD11c-DTR mice express DTR, an abbreviation for diptheria toxin receptor, driven by the CD11c promoter. Cells were stained with Thy1.1 and CD8 for analysis. 1 ⁇ 10 5 EL4 thymoma expressing OVA or STFOVA were transplanted.
  • FIG. 6A is a graph showing a time course of the percentage (%) of tumor-bearing mice vaccinated in the flank with the indicated compositions (PBS (control), irradiated EL4-OVA tumor cells, irradiated EL4-STFOVA tumor cells, irradiated EL4-OVA tumor cells+2 ng RecFLA, or live EL4-STFOVA cells) 30 days after challenge with 50,000 live EL4-OVA cells in the opposite flank. “live” means non-irradiated tumor cells). Groups are composed of 10 mice.
  • FIG. 6B is a panel of flow cytometry dot plots showing percentages (boxed cells) of endogenous OVA-specific CD4 + T cells in tumor draining lymph nodes of vaccinated wt mice (described in FIG. 6A ). Cells were stained with an I-A b -OVA tetramer and CD4.
  • FIG. 7A shows the mutations introduced within the flagellin sequence.
  • the nucleotide sequence of the C-terminal domain of flagellin is shown and key residues for TLR5 activation (Ile 411) and for Ipaf/Naip5 activation (Leu470, 472, 473) are in bold. These residues were mutated into Alanine (underlined) to impair TLR5 and Ipaf/Naip5 activation, either alone or in combination. Expected effect on TLR5 and NLR activation is shown on the right.
  • FIG. 7B is a graph showing tumor volume (mm 3 ) in individual wild-type mice injected subcutaneously in the flank with EL4 cells expressing STFOVA or mutated forms of flagellin within the STFOVA fusion (STFOVA- ⁇ TLR5, STFOVA- ⁇ Naip5 A, STFOVA- ⁇ Naip5 B, or STFOVA-2 ⁇ ) 20 days after injection.
  • STFOVA- ⁇ TLR5, STFOVA- ⁇ Naip5 A, STFOVA- ⁇ Naip5 B, or STFOVA-2 ⁇ 20 days after injection.
  • Statistical significance p value is indicated on the graph (***p ⁇ 0.001). Each symbol represents one mouse.
  • FIG. 7C shows two panels of flow cytometry plots showing proliferation of OVA-specific CD8 + OT-1 T cells (upper panels) in tumor draining lymph node at day 3 after tumor transplantation of EL4 thymoma expressing OVA, STFOVA or mutated forms of flagellin within the STFOVA fusion protein (STFOVA- ⁇ NLR, which lacks the NLR Naip5 activating residues, or STFOVA-2 ⁇ which lacks both the TLR5 and NLR Naip5 activating residues).
  • Lower panels shows flow cytometry dot plots showing IFN- ⁇ and granzyme B secretion by OT-I cells from each indicated groups. Cells were stained with Thy1.1 for analysis.
  • FIG. 8A shows histograms of CD40 expression on splenic dendritic cells after phagocytosis of EL4 or A20 apoptotic tumor cells expressing STFOVA (Flagellin-OVA fusion protein) or OVA (control).
  • STFOVA Fiberllin-OVA fusion protein
  • OVA control
  • FIG. 8B is a graph quantifying IL-12 secretion (ng/ml) by wild-type (wt) and MyD88 ⁇ / ⁇ splenic dendritic cells in response to apoptotic tumor cells expressing EL4-STFOVA or EL4-OVA and in resting dendritic cells.
  • FIG. 9 shows pictures of lungs (3 mice per group) isolated at day 28 from wt mice injected with 100 000 B16 melanoma cells expressing E ⁇ or E ⁇ fused to flagellin (Stf.E ⁇ ).
  • E ⁇ expressing B16 cells were co-injected with 2 ng recombinant flagellin (RecFLA). B16 metastases are visualized as black foci.
  • the present invention is related to immunogenic compositions comprising a tumor cell which expresses a TLR ligand-tumor associated antigen (TAA) fusion protein.
  • TAA TLR ligand-tumor associated antigen
  • NLR Nod-like receptor
  • a fusion protein of the invention comprises a TAA and ligand which is both a TLR ligand and an NLR ligand.
  • the fusion protein comprises at least two ligands, wherein at least one is a TLR ligand and at least one is an NLR ligand.
  • the invention also provides methods for inducing an anti-tumor immune response by immunizing a mammal with a composition comprising a tumor cell which expresses a TLR ligand-TAA fusion protein and/or an NLR ligand-TAA fusion protein.
  • the tumor cell can also express a fusion protein comprising a TAA, a TLR ligand and an NLR ligand.
  • methods for inducing an anti-tumor immune response by immunizing a mammal with a composition comprising a dendritic cell (DC) that has internalized (e.g., phagocytosed) a tumor cell expressing a TLR ligand-TAA fusion protein and/or a tumor cell expressing an NLR-TAA fusion protein.
  • DC dendritic cell
  • the TLR ligand of a fusion protein of the invention is a flagellin or a profilin-like protein (PLP), or fragment thereof.
  • an NLR ligand of the invention is an anthrax toxin, which activates NLRP1, or an Staphylococcus aureus toxin such as alpha-hemolysin, which can activate NLRP3, or a C-terminal fragment of Flagellin which is involved in Ipaf and Naip5 activation.
  • the present invention contemplates the use of any polypeptide with the capacity to stimulate an NLR.
  • the full length flagellin protein is both a TLR ligand and an NLR ligand.
  • compositions comprising autologous tumor cells modified ex vivo to express an NLR-ligand and/or TLR ligand-TAA fusion protein produce a much improved immune response in vivo as compared to autologous tumor cells expressing TAA co-administered (but not physically linked) with a TLR or NLR ligand.
  • the present Examples demonstrate that wild-type mice transplanted with tumor cells expressing flagellin (which is both a TLR ligand and an NLR ligand)-ovalbumin ((OVA)(which is a model TAA)) fusion protein fail to develop tumors, whereas mice transplanted with tumor cells expressing OVA alone or mice transplanted with tumor cells expressing OVA alone and treated with recombinant flagellin (RecFLA) develop tumors.
  • flagellin which is both a TLR ligand and an NLR ligand
  • OVA ovalbumin
  • compositions of the present invention are particularly effective for eliciting innate immune cell activation (e.g. macrophages, dendritic cells).
  • innate immune cell activation e.g. macrophages, dendritic cells.
  • compositions of the present invention are particularly effective for eliciting both CD4 + and CD8 + T cell driven immune responses.
  • a new growth comprising tumor cells is a tumor, also known as a neoplasm.
  • a tumor is an abnormal tissue growth, generally forming a distinct mass, that grows by cellular proliferation more rapidly than normal tissue growth.
  • a tumor may show partial or total lack of structural organization and functional coordination with normal tissue.
  • a tumor is intended to encompass hematopoietic tumors as well as solid tumors.
  • a tumor may be benign (benign tumor) or malignant (malignant tumor or cancer).
  • a tumor or tumor tissue may also comprise non-tumor cells, e.g., vascular cells which form blood vessels to supply the tumor or tumor tissue or stroma cells.
  • non-tumor cells e.g., vascular cells which form blood vessels to supply the tumor or tumor tissue or stroma cells.
  • anti-tumor immune response means an immune response, which can be innate, humoral (e.g., antibody-mediated) or cellular (e.g. CD4 or CD8 T cell mediated), or any combination thereof, directed against a tumor, tumor cell, a cancer cell, and/or antigens expressed by a tumor/cancer cell.
  • TAA tumor associated antigen
  • protein as used herein, is synonymous with the term “peptide” or “polypeptide”, and is understood to mean a chain of amino acids arranged linearly and joined together by peptide bonds between the carboxyl and amino groups of adjacent amino acid residues.
  • polypeptide can refer to a full length amino acid sequence of a protein, or to a fragment thereof.
  • immunogenic means that an agent is capable of eliciting a humoral or cellular immune response, and preferably both.
  • An immunogenic composition is a composition that elicits a humoral or cellular immune response, or both, directed against one or more components of the composition, when administered to an animal having an immune system.
  • autologous cell is synonymous with “syngeneic cell” and means a self cell or cell that is identical or substantially identical to an individual's self cell.
  • non-autologous cell is synonymous with an “allogeneic cell” and means a non-self (non-identical) cell or xenogeneic cell.
  • a fusion protein of the invention includes any of the fusion proteins described herein, such as TLR ligand-TAA fusion protein or an NLR ligand-TAA fusion protein or a fusion protein expressing a TAA, a TLR ligand and an NLR ligand.
  • TLR5 ligand-TAA fusion protein is a flagellin-MUC1 fusion protein or a PLP-MUC1 fusion protein.
  • NLR ligand-TAA fusion proteins include Flagellin-MUC1 fusion protein or anthrax toxin-MUC1 fusion protein, where MUC-1 is a TAA.
  • anthrax toxin Preferably, only the relevant NLR binding residues of anthrax toxin are included in the fusion protein, in order to avoid toxic effects of the full length anthrax toxin.
  • a fusion protein comprising a TAA and a 20 amino acid C-terminal fragment of flagellin (an NLR ligand).
  • the term “distinct TLR ligand” in the context of a fusion protein comprising a TLR ligand and an NLR ligand means that the TLR ligand is a different ligand than the NLR ligand in the fusion protein.
  • the term “distinct NLR ligand” in the context of a fusion protein comprising a TLR ligand and an NLR ligand means that the NLR ligand is a different ligand than the TLR ligand in the fusion protein.
  • composition of the invention includes any of the compositions described herein, such as a composition comprising a tumor cell expressing a fusion protein of the invention or a composition comprising a dendritic cell loaded with a fusion protein of the invention.
  • subject or “individual” as used herein refers to an animal having an immune system, preferably a mammal (e.g., rodent, such as mouse). In particular, the term encompasses humans.
  • the term “about” or “approximately” usually means within an acceptable error range for the type of value and method of measurement. For example, it can mean within 20%, more preferably within 10%, and most preferably still within 5% of a given value or range. Alternatively, especially in biological systems, the term “about” means within about a log (i.e., an order of magnitude) preferably within a factor of two of a given value.
  • sequence identity is the percentage of residues in an amino acid or nucleic acid sequence that are identical after aligning the sequence with a reference sequence and introducing gaps, if necessary, to achieve maximal sequence identity. Methods and computer programs for the alignment, such as BLAST, are well known in the art.
  • a polypeptide is substantially identical with the 170 residues from the N terminus and 90 residues from the C terminus of a naturally occurring bacterial flagellin
  • the polypeptide and the reference sequence (170 residues from the N terminus and 90 residues from the C terminus of the naturally occurring bacterial flagellin) are maximally aligned, at least 30% of the amino acids in the reference sequence are found in the corresponding positions in the polypeptide.
  • substantially identical at the cellular level, means that a cell is sufficiently similar to a cell of a host, such that the host's immune system does not mount an immune response against the substantially similar cell (i.e., the cell is recognized as a self cell by the immune system).
  • Treating” or “treatment” of a state, disorder or condition includes: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a human or other mammal that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition, (2) inhibiting the state, disorder or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof (in case of maintenance treatment) or at least one clinical or subclinical symptom thereof, or (3) relieving the disease, i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms.
  • treating a cancer means alleviating or eliminating the symptoms of a tumor, or slowing down the progress of the tumor.
  • the alleviating or eliminating effect can be determined by any method known in the art, such as measuring the size of the tumor and observing biochemical indicators of the particular tumor.
  • a subject is treated if showing one or more of the following: reduction in the number of cancer cells; reduction in the tumor size; inhibition or elimination of cancer cell infiltration into peripheral organs, including the spread of cancer into soft tissue and bone; inhibition or elimination of tumor metastasis; inhibition of tumor growth; reduction of one or more of the symptoms associated with the specific cancer; and reduced morbidity and mortality.
  • the alleviation is preferably at least about 10%, more preferably at least about 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90%.
  • a “flagellin” may be may be any polypeptide that binds a naturally occurring TLR5 and triggers at least one of the biological functions of the TLR5 in antigen-presenting cells upon such binding.
  • a flagellin may be a polypeptide comprising any of the naturally occurring bacterial flagellin proteins.
  • a flagellin may also be a polypeptide that is substantially identical with any of the naturally occurring bacterial flagellin proteins at the amino acid sequence level, wherein the polypeptide is capable of binding a naturally occurring TLR5.
  • a flagellin may be a polypeptide that is substantially identical with the 170 residues from the N terminus and 90 residues from the C terminus of any of the naturally occurring bacterial flagellin proteins at the amino acid sequence level, wherein the polypeptide is capable of binding a naturally occurring TLR5.
  • the flagellin of this invention may also comprise a modification, such as glycosylation or phosphorylation.
  • the flagellin may also be a mutant or protein variant of flagellin.
  • Flagella are found primarily, although not exclusively, on the surface of rod and spiral shaped bacteria, including members of the genera Escherichia, Salmonella, Proteus, Pseudomonas, Bacillus, Campylobacter, Vibrio, Treponema, Legionella, Clostridia , and Caulobacter . Flagellin sequences are readily obtainable based on knowledge in the art. In fact, the flagellin sequences from numerous bacterial species, as well as structural analyses, have been published.
  • Any analogs, derivatives of flagellin or fragments thereof with flagellin function namely one that binds a naturally occurring TLR5 and/or NLRs (Naip5 and/or Ipaf) and triggers at least one of the biological functions of TLR5 and/or Naip5 and/or Ipaf in antigen-presenting cells upon such binding, can be used in the present invention.
  • These include polypeptides comprising any of the naturally occurring bacterial flagellin proteins, and polypeptides that are substantially identical with any of the naturally occurring bacterial flagellin proteins at the amino acid sequence level, wherein the polypeptides are capable of binding a naturally occurring TLR5 and/or Naip5 and/or Ipaf.
  • Flagellin sequences from numerous bacteria are available in the art, such as but not limited to GenBank accession numbers D13689 (nucleic acid sequence) (SEQ ID NO: 2), YP_275549 (SEQ ID NO: 3), YP — 275550 (SEQ ID NO: 4), AAU18718 (SEQ ID NO: 5), AAU18717 (SEQ ID NO: 6), ZP — 00743095 (SEQ ID NO: 7), EAO52626 (SEQ ID NO: 8), YP_315348 (SEQ ID NO: 9), AAT28337 (SEQ ID NO: 10), AAT28336 (SEQ ID NO: 11), AAT28335 (SEQ ID NO: 12), AAT28334 (SEQ ID NO: 13), AAT28333 (SEQ ID NO: 14), AAZ36356 (SEQ ID NO: 15), AAZ33167 (SEQ ID NO: 16), AAZ94424 (SEQ ID NO: 17), AAZ91670 (SEQ
  • the flagellin proteins from different species exhibit a high degree of protein sequence homology at the amino and carboxy termini (about 170 residues from the N terminus and about 90 residues from the C terminus), and the presence of a polymorphic central region which is responsible for the antigenic diversity among different flagella.
  • the conserved regions are important for TLR5 binding, while the polymorphic central region can be deleted without affecting binding to TLR5.
  • Structural-function analyses of the flagellin proteins have been reported (see, e.g., Smith K D et al., Toll-like receptor 5 recognizes a conserved site on flagellin required for protofilament formation and bacterial motility, Nat Immunol.
  • a fusion protein of the invention comprises a TAA and a fragment of flagellin.
  • the fragment of flagellin is capable of binding to TLR5 but not Naip5 or Ipaf.
  • the Flagellin fragment can be missing the C-terminal portion required for NLR activation and contain a conserved sequence recognized by TLR5 [Sec Smith, K. D. et al. Nat Immunol 2003 vol. 4 (12) pp. 1247-53].
  • the fragment of flagellin is capable of binding to NLRs (e.g., Naip5 and Ipaf) but not to TLR5.
  • NLRs e.g., Naip5 and Ipaf
  • the C-terminal sequence of flagellin from S.
  • VLAQANQVPQNVLSLLR (SEQ ID NO: 31) or the sequence TSVLAQANQVPQNVLSLLR (SEQ ID NO: 32) may be comprised in a fusion protein of the invention. It is to be understood that this sequence can differ from one bacteria to another but the key residues for Ipaf and Naip5 activation are conserved [Karla L Lightfield, et al. Nat Immunol 2008 vol. 9 (10) pp. 1171-1178]. Thus variants of this sequence containing the conserved residues required by binding and activation of NLR are also contemplated for use in the instant invention.
  • a fusion protein of the invention comprises the C-terminal sequence of flagellin from S. typhimurium : VLAQANQVPQNVLSLLR (SEQ ID NO: 31) or the sequence TSVLAQANQVPQNVLSLLR (SEQ ID NO: 32), (both of which can activate Naip5 or Ipaf but not TLR5) and the TLR ligand PLP.
  • this fusion protein contains at least one TLR ligand and at least one NLR ligand.
  • a fusion protein is a fusion protein comprising PLP and a fragment of anthrax toxin (the amino acid sequence required for binding to NLR NLRP1) or a fusion protein comprising PLP and a fragment of the Staphylococcus aureus alpha-hemolysin (the amino acid sequence required for binding to the NLR NLRP3).
  • an example of an amino acid sequence of flagellin which activates both TLR5 and NLR is a full length flagellin.
  • an example of an amino acid sequence of flagellin which activates NLR but not TLR5 is TSVLAQANQVPQNVLSLLR (SEQ ID NO: 32).
  • a sequence which activates TLR5 but not NLR is a flagellin amino acid sequence in which the last 20 residues at the C-terminus are deleted.
  • the present Examples use flagellin-TAA fusion proteins, the invention is not to be limited thereto.
  • fusion proteins comprising other TLR and/or NLR ligands and a TAA.
  • the TLR or NLR ligand is a polypeptide ligand, in order to facilitate its expression as a fusion protein, e.g, in tumor cell.
  • the protein TLR11 ligand, profilin-like protein (PLP), or a fragment thereof, for which amino acid sequences from many different organism are known may also be used as a TLR ligand in a fusion protein of the present invention.
  • Non-limiting examples of amino acid sequences of PLP, which may be used to generate a fusion protein of the invention include GenBank Accession numbers ABB43118 (SEQ ID NO: 21), BAB09877 (SEQ ID NO: 22), ABZ80128 (SEQ ID NO: 23), YP — 717473 (SEQ ID NO: 24), ABD97732 (SEQ ID NO: 25), ABC61055 (SEQ ID NO: 26), ABB16985 (SEQ ID NO: 27), and AAY97753 (SEQ ID NO: 28).
  • Anthrax toxin and Staphylococcus aureus toxins such as alpha-hemolysin (GenBank Accession no. AAA26598) (SEQ ID NO: 1) activate NLRP3 and are non-limiting examples of protein NLR ligands contemplated for use in the fusion proteins of the present invention.
  • a fusion protein of the invention comprises a TAA and a ligand that stimulates both a TLR and an NLR.
  • a fusion protein comprises a TAA and two or more TLR ligands, or two or more NLR ligands, or at least one TLR ligand and at least one NLR ligand.
  • a fusion protein of the invention comprises a TAA (e.g. MUC-1), PLP (a TLR ligand) and anthrax toxin (an NLR ligand).
  • a tumor cell of the invention may be engineered to express two or more fusion proteins, wherein at least one fusion protein comprises a TAA and a TLR ligand (e.g. PLP) and another at least one fusion protein comprises a TAA and an NLR ligand (e.g. anthrax toxin).
  • the TAA in each construct expressed in the tumor may be the same TAA or a different TAA.
  • a mammalian cell preferably a tumor cell, and still more preferably an autologous tumor cell, are engineered to express an NLR- or TLR-ligand-TAA fusion protein.
  • TAAs Tumor-associated antigens are well known and described in the art. Any protein antigen expressed by a tumor cell is contemplated for use in the present invention.
  • Preferred TAAs are those which are known to be highly immunogenic (i.e., that comprise immunodominant epitopes that will stimulate a strong anti-tumor immune response.)
  • Non-limiting examples of TAAs contemplated for use in the fusion proteins of the present invention include ErbB receptors, Melan A [MART 1], gp100, tyrosinase, TRP-1/gp 75, and TRP-2 (in melanoma; for additional examples, see also a list of antigens provided in Storkus and Zarour, Forum (Genova), 2000 July-September, 10(3):256-270); MAGE-1 and MAGE-3 (in bladder, head and neck, and non-small cell carcinoma); HPV E6 and E7 proteins (in cervical cancer); Mucin [MUC-1] (in breast, pancreas, colon
  • Tumor cells may be isolated from a mammalian subject or patient. For example, a tumor may be removed from a patient during a biopsy or surgery, and tumor cells may be obtained and cultured from the biopsy sample [Liangping Li, Establishment of tumor cell lines by transient expression of immortalizing genes Gene Ther Mol Biol Vol 4, 261-274. December 1999]. Tumor cells may be autologous or non-autologous. Tumor cells for use in the instant invention may also be derived from any suitable tumor cell line.
  • Non-limiting examples of tumor cell lines contemplated for use in the present invention include DU145 (Prostate cancer), Lncap (Prostate cancer), MCF-7 (breast cancer), MDA-MB-438 (breast cancer), PC3 (Prostate cancer), T47D (breast cancer), THP-1 (acute myeloid leukemia), BN1 (melanoma), U87 (glioblastoma), SHSY5Y Human neuroblastoma cells, cloned from a myeloma, and Saos-2 cells (bone cancer). Any suitable tumor cell line is contemplated for use in the present invention.
  • One or more fusion protein of the invention are expressed in tumor cells of the invention.
  • Methods of expressing exogenous or recombinant proteins in a cell are well known in the art. Such methods include, but are not limited to, transfection, microinjection, scrape-loading, and receptor-mediated uptake by the cell. Transfection may be transient or stable. Exemplary current methods of transfection include calcium phosphate precipitation, electroporation, lipofection, and peptide-mediated transfection. Ballistic DNA delivery and transduction (i.e., the introduction of foreign DNA by virus or virus vector infection) can also be employed.
  • a flagellin or other TLR or NLR ligand can be delivered to cells by means of an expression vector.
  • Suitable expression vectors comprise a promoter that is active in the cells in which the ligand is to be expressed.
  • Expression vectors useful for practicing the invention may also include selectable markers, cell-type or cell-cycle-specific enhancers or repressors, polylinkers, start codons, ribosome binding sites, internal ribosome entry sites, introns, stop codons, polyadenylation signals, or other features that facilitate cloning and vector stability, mRNA stability and localization in the cell, and translation efficiency, or combinations thereof.
  • Expression vectors include viral expression vectors. Selection of these features is largely based on the cells to be transfected, and the expression characteristics desired. A large number of commercially available vectors are available for expressing polypeptides in cells.
  • modified fusion proteins such as glycosylated or phosyphorylated fusion proteins.
  • Subcellular targeting motifs are also contemplated.
  • vectors may also be expressed in tumor cells using retroviral, adenoviral or lentiviral vectors.
  • Tumor cells for use in the present invention may be apoptotic or live (non-apoptotic) cells.
  • apoptosis is induced in tumor cells just prior to incubation with DCs or prior to administration to a subject. This may be done in order to prevent proliferation of tumor cells in the recipient.
  • apoptotic cells are easily recognized by DC and internalized. Internalization delivers the apoptotic cell and all proteins derived thereof into endo-lysosomal compartments that generate the ligands necessary for the activation of CD4 + and CD8 + T cells.
  • Apoptosis may be induced using chemotherapeutic agents (such as, e.g., oxaliplatin, cisplatin, carboplatin or other platinum-based drugs), alkylating agents (e.g., mitomycin C), toposiomerase II inhibitor (e.g., etoposide) anthracyclins (e.g, mitoxantrone), inducers of endoplasmic reticulum stress (e.g, thapsigargin), or cells may be lethally irradiated.
  • chemotherapeutic agents such as, e.g., oxaliplatin, cisplatin, carboplatin or other platinum-based drugs
  • alkylating agents e.g., mitomycin C
  • toposiomerase II inhibitor e.g., etoposide
  • anthracyclins e.g, mitoxantrone
  • inducers of endoplasmic reticulum stress e
  • Art-recognized methods can be used to determine the dose of radiation and whether the irradiated cells can replicate. For example, cells growing at a density of 5 ⁇ 10 5 cells/ml can be irradiated with 10,000 Rads, then viable cell numbers can be determined over time by, e.g., Trypan blue exclusion.
  • the lethally irradiated cell should preferably be able to continue to express proteins for a period of time (see, e.g., Borrello I et al., A universal granulocyte-macrophage colony-stimulating factor-producing bystander cell line for use in the formulation of autologous tumor cell-based vaccines, Hum Gene Ther. 1999 Aug.
  • Protein expression by the lethally irradiated cells can be assayed by methods known in the art, such as gel electrophoresis and protein staining for protein synthesis in general, or Western analysis for specific protein(s).
  • methods for inducing an anti-tumor immune response in a mammal or for treating a cancer comprise administering to a mammal or patient an immunogenically effective amount of a composition comprising a DC, wherein the DC has internalized a tumor cell expressing a fusion protein if the invention.
  • DCs are known to be potent stimulators of the adaptive immune response (e.g. T and/or B cell mediated immune responses).
  • the present invention is based in part on the results of immunizing mice with dendritic cells loaded with tumor cells expressing TLR-or NLR-ligand-TAA fusion protein compared to immunizing mice with DCs loaded with tumor cells and TLR ligand and TAA separately.
  • the superiority of the methods and compositions of the instant invention is achieved because the physical linkage in a fusion protein of the TAA to the TLR or NLR ligand (PAMP) facilitates delivery of both the activation signal (PAMP) and the antigen (TAA) to the same endo/lysosomal compartment within the DC, thereby increasing DC activation and the ability to induce a potent, antigen-specific immune response to the TAA.
  • PAMP activation signal
  • TAA antigen-specific immune response to the TAA.
  • the tumor cell provides additional antigens that also contribute to the development of a potent anti-tumor immune response of superior quality to presently available methods.
  • tumor cells for direct immunization of patients may be dead tumor cells expressing a fusion protein of the invention.
  • Tumor cells can be rendered dead by various means such as irradiation or as indicated above. It may be confirmed that tumor cells are dead by propidium iodide incorporation, TUNEL assay, Annexin-V and 7-Aminoactinomycin D (7AAD) or any other suitable method known in the art. Tumor cells may also be necrotic.
  • human DC precursors i.e. circulating monocytes
  • the tumor-bearing patient to be treated i.e. autologous cells
  • DC can be subsequently pulsed for, e.g., 6 hours with tumor cell extracts ( ⁇ -irradiated tumor cell for example) that had been previously engineered to express the TLR5 and/or Ipaf and/or Naip5 ligand flagellin fused to a tumor associated antigen of choice (different strategies will be evaluated: retroviral-based gene transfer, adenoviral gene transfer or transfection).
  • tumor cell extracts ⁇ -irradiated tumor cell for example
  • IFN- ⁇ or other inflammatory cytokine such as TNF- ⁇ , or antibodies to the co-stimulatory molecule CD40 (anti-CD40) are added to the culture to increase the maturation (e.g., upregulation of costimulatory molecules) of DC prior to the injection.
  • Flagellin-TAA fusion protein expressing-tumor “loaded” DCs can be injected into the patient via different routes, e.g., intravenously, subcutaneously or directly into the tumor-draining lymph node. Any suitable route of injection is contemplated by the present invention. Direct injection into a tumor-draining lymph node can ensure the proximity of the DC to T cells in order to induce an antigen-specific T cell driven immune response. The number of loaded DCs to be injected as well as the frequency of injection can be determined experimentally. Clinical criteria for evaluating efficacy of immunotherapies are well defined, in particular for solid tumor (J.
  • tumor cells for preparing a composition of the invention may be obtained from the same patient who is to be administered the composition (i.e., the tumor cells may be autologous to the cells of the patient).
  • Autologous tumor cells are preferred as they express the same antigens expressed by the patient's tumor cells, and will thus help drive an effective anti-tumor immune response directed against the patient's tumor, by allowing generation of immune response against additional antigens (in addition to the TAA).
  • Non-autologous tumor cells may also be used however, since they are engineered to express a TAA-containing fusion protein, wherein the TAA is preferably expressed by the patient's tumor cells.
  • DCs may be autologous cells. This is preferred, since self DCs will not stimulate an alloimmune response, and will therefore avoid being eliminated by the host immune system before inducing the anti-tumor immune response.
  • a composition comprises a tumor cell expressing a fusion protein, wherein said fusion protein comprises a TLR ligand and/or an NLR ligand and a TAA.
  • a composition of the invention comprises a DC, wherein the DC has internalized a tumor cell expressing a fusion protein comprising a TLR ligand and/or NLR ligand and a tumor-associated antigen (TAA).
  • TAA tumor-associated antigen
  • the DC is “loaded” with a fusion-protein expressing tumor cell.
  • the loaded DC has been treated with IFN- ⁇ before administration to the patient.
  • the tumor cell with which the DC is loaded may be lethally irradiated and/or apoptotic or necrotic.
  • the DC is loaded with extracts from a tumor cell expressing a fusion protein of the invention.
  • compositions and methods of the present invention are useful for inducing an anti-tumor immune response, and for cancers.
  • cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • cancers include: squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer (including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung), cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer (including gastrointestinal cancer, pancreatic cancer), glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and
  • a cancer includes primary malignant cells (e.g., those that have not migrated to sites in the subject's body other than the site of the original malignancy) and secondary malignant cells (e.g., those arising from metastasis, the migration of malignant cells to secondary sites that are different from the site of the original tumor).
  • primary malignant cells e.g., those that have not migrated to sites in the subject's body other than the site of the original malignancy
  • secondary malignant cells e.g., those arising from metastasis, the migration of malignant cells to secondary sites that are different from the site of the original tumor.
  • the present invention provides a pharmaceutical composition or formulation comprising at least one composition of the invention, or a pharmaceutically acceptable derivative thereof, in association with a pharmaceutically acceptable excipient, diluent, and/or carrier.
  • the excipient, diluent and/or carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • compositions of the invention can be formulated for administration in any convenient way for use in human or veterinary medicine.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions.
  • the carrier can be a solid dosage form carrier, including but not limited to one or more of a binder (for compressed pills), a glidant, an encapsulating agent, a flavorant, and a colorant. Suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin (1990, Mack Publishing Co., Easton, Pa. 18042).
  • vacun refers to a composition that can be used to elicit protective immunity in a recipient. It should be noted that to be effective, a vaccine of the invention can elicit immunity in a portion of the immunized population, as some individuals may fail to mount a robust or protective immune response, or, in some cases, any immune response. This inability may stem from the individual's genetic background or because of an immunodeficiency condition (either acquired or congenital) or immunosuppression (e.g., due to treatment with chemotherapy or use of immunosuppressive drugs). Vaccine efficacy can be established in animal models.
  • compositions and formulations of the present invention may comprise pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers.
  • Such compositions include diluents of various buffer content (e.g., Tris-HCl, acetate, phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc.
  • buffer content e.g., Tris-HCl, acetate, phosphate
  • pH and ionic strength e.g., Tris-HCl
  • Hylauronic acid may also be used. See, e.g., Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, Pa. 18042) pages 1435 1712 which are herein incorporated by reference.
  • Preparations according to this invention for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions.
  • non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate.
  • Such dosage forms may also contain adjuvants, preserving, wetting, emulsifying, and dispersing agents.
  • the pharmaceutical compositions may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use.
  • compositions e.g., pharmaceutical or vaccine compositions
  • formulations of the present invention can be administered parenterally, by inhalation, or by other suitable methods known in the art.
  • parenteral includes injection (for example, intravenous, intraperitoneal, epidural, intrathecal, intramuscular, intraluminal, intratracheal or subcutaneous). Injections directly into the primary site of tumor are also contemplated.
  • the preferred routes of administration are subcutaneous and intravenous and direct injection into a tumor-draining lymph node.
  • compositions and formulations of the present invention may be administered to an animal, preferably a mammal, and most preferably a human.
  • compositions or formulations of the present invention will vary widely, depending upon the nature of the disease, the patient's medical history, age, body weight, sex, sensitivity, the frequency of administration, the manner and route of administration, the clearance of the agent from the host, dosage period, drugs used in combination, and the like.
  • the initial dose may be larger, followed by smaller maintenance doses.
  • the therapeutically effective dose can be estimated initially from animal models. Dose-response curves derived from animal systems are then used to determine testing doses for the initial clinical studies in humans. In safety determinations for each composition, the dose and frequency of administration should meet or exceed those anticipated for use in the clinical studies.
  • the data obtained from the animal studies can be used in formulating a range of doses for use in humans.
  • the therapeutically effective doses of in humans lay preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. Ideally, a single dose of each drug should be used daily.
  • compositions of the invention will typically contain an effective amount of the compositions for achieving the desired effect.
  • therapeutically effective amount/dose is used interchangeably with the terms “immunogenically effective amount/dose” and “effective amount/dose” and refers to an amount of the substance that is sufficient to achieve the intended effect.
  • An immunogenically effective amount of a flagellin-TAA-expressing tumor cell of the invention is an amount of the cell that is sufficient to induce an anti-tumor immune response.
  • An effective amount of a composition e.g.
  • vaccine for inducing an anti-tumor immune response is an amount of the composition sufficient to alleviate or eliminate the symptoms of the tumor, for slowing down the progress of the tumor, for eliminating or reducing the size of the tumor, or for preventing the development of a tumor and metastases.
  • the effective amount will vary with factors such as the nature of the substance, the route of administration, the formulation comprising the substance, and the size, species, and health condition of the recipient of the substance. Methods to determine the effective amount are known in the art.
  • Administration of the compositions or formulations of the invention may be once a day, twice a day, or more often, but frequency may be decreased during a maintenance phase of the disease or disorder, e.g., once every second or third day instead of every day or twice a day.
  • the dose and the administration frequency will depend on the clinical signs, which confirm maintenance of the remission phase, with the reduction or absence of at least one or more preferably more than one clinical signs of the acute phase known to the person skilled in the art. More generally, dose and frequency will depend in part on recession of pathological signs and clinical and subclinical symptoms of a disease, condition or disorder contemplated for treatment with the present compounds.
  • the appropriate dose and dosage times under certain conditions can be determined by the test based on the above-described indices but may be refined and ultimately decided according to the judgment of the practitioner and each patient's circumstances (age, general condition, severity of symptoms, sex, etc.) according to standard clinical techniques.
  • typical dosages of DCs in a composition of the invention range from about 1 ⁇ 10 6 to about 10 ⁇ 10 7 cells. However more or fewer DCs may be used.
  • typical dosages of tumor cells in a composition of the invention range from about 1 ⁇ 10 6 to about 10 ⁇ 10 7 cells. However more or fewer tumor cells may also be used with similar results.
  • the present invention contemplates combination therapies.
  • a subject or patient having a tumor may also be simultaneously, immediately before, or immediately after, subjected to another therapeutic measure such as but not limited to radiotherapy, chemotherapy or surgery.
  • Another possible combination is to administer a composition of the present invention in combination with a humanized anti-CD25 antibody (Daclizumab) that depletes regulatory T cells.
  • Regulatory T cells have been shown to be increased in number in the peripheral blood as well as tumors of patients with cancer, and can suppress the functions of anti-tumor effector CD4 and CD8 T cells.
  • TLR ligands such as various CpG derivatives or TLR7/TLR8 agonists such as Imiquimod, may be administered simultaneously within the formulation to enhance adjuvanticity of the preparations.
  • John Wiley and Sons, Inc. Hoboken, N.J.; Bonifacino et al., eds. (2005) Current Protocols in Cell Biology. John Wiley and Sons, Inc.: Hoboken, N.J.; Coligan et al., eds. (2005) Current Protocols in Immunology, John Wiley and Sons, Inc.: Hoboken, N.J.; Coico et al., eds. (2005) Current Protocols in Microbiology, John Wiley and Sons, Inc.: Hoboken, N.J.; Coligan et al., eds.
  • EL4 thymoma cells engineered to express either an ovalbumin (OVA) construct or an OVA- S. typhimurium flagellin fusion protein (STFOVA) using retroviral transduction with pMIG-IRES-GFP were subcutaneously injected in the flank into age and sex-matched syngeneic C57BL/6 ( FIG. 1A ) or MyD88 ⁇ / ⁇ ( FIG. 1B ) mice in presence or not of recombinant flagellin (RecFLA, InvivoGen, San Diego, Calif.) (for wt only). Tumor progression was monitored by bi-weekly measure of tumor volume.
  • EL4 cells expressing OVA constructs were subcutaneously injected into the flank of Rag ⁇ / ⁇ or wt control mice. Tumor progression was monitored by bi-weekly measure of tumor volume.
  • TLR5/Ipaf/Naip5 Ligand are Efficiently Targeted by the Innate Immune System
  • TLR5 ligand flagellin within OVA-expressing tumors could induce OVA-specific OT-II CD4 + T cells in vivo was investigated. It was found that expression of flagellin within tumor cells strongly enhanced TAA-specific CD4 + T cell proliferation in vivo ( FIG. 4 ). This finding is consistent with the previous observations demonstrating that TLR ligands enhance presentation of phagocytosed antigens within major histocompatibility class II MHC class II molecules [Blander, J. M. and Medzhitov, R., Nature (2006), Vol. 440, pp. 808]. These data also demonstrate that unlike EL4-OVA tumor cells, EL4-STFOVA tumor cells are capable of eliciting a helper anti-tumor CD4 T cell response.
  • NLR Activating Domain of Flagellin is Required for its Anti-Tumor Potential in Vivo
  • DC dendritic cells
  • human DC precursors i.e. circulating monocytes
  • IL-4 and GM-CSF a well-defined protocol that involves culture in the cytokines IL-4 and GM-CSF.
  • Flagellin-TAA fusion protein expressing-tumor loaded DC is then injected into the patient intravenously or directly into the lymph nodes draining the tumor to ensure the proximity of the DC to T cells in the tumor draining lymph nodes.
  • the number of loaded DC to be injected as well as the frequency of injection is evaluated by several clinical trials. However, suitable numbers of DCs can range, e.g. from about 1 ⁇ 10 6 to 10 ⁇ 10 7 cells.
  • Clinical criteria for evaluating of efficacy of immunotherapies are well defined, in particular for solid tumor (J. D Wolchok, A Hoos, S O'day, J. S Weber, O Hamid, C Lebbe, M Maio, M Binder, O Bohnsack, G Nichol, R Humphrey, F. S Hodi. Clinical Cancer Research, 2009 vol. 15 (23) pp. 7412-7420). Evaluation of the efficacy of this DC-based therapy may follow these criteria.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US13/577,295 2010-02-05 2011-02-07 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells Abandoned US20130108661A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/577,295 US20130108661A1 (en) 2010-02-05 2011-02-07 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US30205210P 2010-02-05 2010-02-05
US30661810P 2010-02-22 2010-02-22
US13/577,295 US20130108661A1 (en) 2010-02-05 2011-02-07 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells
PCT/US2011/023919 WO2011097573A2 (en) 2010-02-05 2011-02-07 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/023919 A-371-Of-International WO2011097573A2 (en) 2010-02-05 2011-02-07 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/484,374 Continuation US9314484B2 (en) 2010-02-05 2014-09-12 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells

Publications (1)

Publication Number Publication Date
US20130108661A1 true US20130108661A1 (en) 2013-05-02

Family

ID=44356104

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/577,295 Abandoned US20130108661A1 (en) 2010-02-05 2011-02-07 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells
US14/484,374 Active US9314484B2 (en) 2010-02-05 2014-09-12 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/484,374 Active US9314484B2 (en) 2010-02-05 2014-09-12 Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells

Country Status (4)

Country Link
US (2) US20130108661A1 (es)
EP (1) EP2531207B1 (es)
ES (1) ES2769326T3 (es)
WO (1) WO2011097573A2 (es)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016069502A1 (en) * 2014-10-31 2016-05-06 Anahit Ghochikyan Method for the cancer treatment and prevention of metastatic disease
WO2016112065A1 (en) * 2015-01-06 2016-07-14 Integrated Biotherapeutics, Inc. Immunogenic composition comprising engineered alpha-hemolysin oligopeptides
US10717767B2 (en) 2017-07-20 2020-07-21 Spogen Biotech, Inc. Bioactive polypeptides for improvements in plant protection, growth and productivity
US11672866B2 (en) 2016-01-08 2023-06-13 Paul N. DURFEE Osteotropic nanoparticles for prevention or treatment of bone metastases

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014113625A1 (en) * 2013-01-18 2014-07-24 University Of Georgia Research Foundation, Inc. Therapeutic compositions and methods for treating infections
IT201900016736A1 (it) * 2019-09-19 2021-03-19 Takis S R L Antigeni fusi alla proteina Profilin-like di Toxoplasma Gondii (PFTG) e loro uso nella vaccinazione preventiva e terapeutica.
JP2023503427A (ja) * 2019-11-18 2023-01-30 ザ・チルドレンズ・メデイカル・センター・コーポレーシヨン 超活性樹状細胞は養子細胞移植に基づく持続性抗腫瘍免疫を可能にする
US20240066118A1 (en) * 2021-01-12 2024-02-29 Cornell University Needle and rod proteins as inflammasome agonists for augmenting immune responses

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8324369B2 (en) * 2007-11-30 2012-12-04 Baylor College Of Medicine Dendritic cell vaccine compositions and uses of same

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6130082A (en) 1988-05-05 2000-10-10 American Cyanamid Company Recombinant flagellin vaccines
US20030232055A1 (en) * 2000-07-31 2003-12-18 Ruslan Medzhitov Innate immune system-directed vaccines
US20020061312A1 (en) * 2000-07-31 2002-05-23 Ruslan Medzhitov Innate immune system-directed vaccines
US20030175287A1 (en) * 2001-01-03 2003-09-18 Yale University Innate immune system-directed vaccines
WO2002085933A1 (en) 2001-04-20 2002-10-31 The Institute For Systems Biology Toll-like receptor 5 ligands and methods of use
TW200303759A (en) * 2001-11-27 2003-09-16 Schering Corp Methods for treating cancer
US7404963B2 (en) * 2004-10-01 2008-07-29 The University Of South Florida Flagellin-based adjuvants and vaccines
ES2291071B1 (es) * 2005-06-13 2009-03-16 Proyecto De Biomedicina Cima, S.L. Agentes y metodos basados en el uso del dominio eda de la fibronectina.
EP1806395A1 (en) * 2006-01-06 2007-07-11 Stichting Sanquin Bloedvoorziening Maturation of dendritic cells
US8748405B2 (en) * 2007-01-26 2014-06-10 City Of Hope Methods and compositions for the treatment of cancer or other diseases
US20080199485A1 (en) * 2007-02-15 2008-08-21 Mannkind Corporation Method for enhancing T cell response
US20090124557A1 (en) * 2007-04-16 2009-05-14 Yeda Research And Development Co. Ltd. Compositions and methods for treating cancer
WO2009128949A2 (en) * 2008-04-18 2009-10-22 Vaxinnate Corporation Compositions of dengue viral proteins and methods of use

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8324369B2 (en) * 2007-11-30 2012-12-04 Baylor College Of Medicine Dendritic cell vaccine compositions and uses of same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Kirk et al., Gene-modified dendritic cells for use in tumor vaccines, Human Gene Ther. 11, 797-806, 2000. *
Xu et al. ,Tumor-specific dendritic cells generated by genetic redirection of Toll-like receptor signaling against the tumor-associated antigen, erbB2. Cancer Gene Ther. 14, 773-780, 2007. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016069502A1 (en) * 2014-10-31 2016-05-06 Anahit Ghochikyan Method for the cancer treatment and prevention of metastatic disease
WO2016112065A1 (en) * 2015-01-06 2016-07-14 Integrated Biotherapeutics, Inc. Immunogenic composition comprising engineered alpha-hemolysin oligopeptides
US11672866B2 (en) 2016-01-08 2023-06-13 Paul N. DURFEE Osteotropic nanoparticles for prevention or treatment of bone metastases
US10717767B2 (en) 2017-07-20 2020-07-21 Spogen Biotech, Inc. Bioactive polypeptides for improvements in plant protection, growth and productivity
US11046735B2 (en) 2017-07-20 2021-06-29 Spogen Biotech Inc. Bioactive polypeptides for improvements in plant protection, growth and productivity

Also Published As

Publication number Publication date
WO2011097573A3 (en) 2011-12-29
US9314484B2 (en) 2016-04-19
EP2531207A2 (en) 2012-12-12
US20150064219A1 (en) 2015-03-05
ES2769326T3 (es) 2020-06-25
EP2531207A4 (en) 2013-09-04
WO2011097573A2 (en) 2011-08-11
EP2531207B1 (en) 2019-10-30

Similar Documents

Publication Publication Date Title
US9314484B2 (en) Methods and compositions for cancer immunotherapy using flagellin-tumor associated antigen fusion protein expressing tumor cells
JP6134763B2 (ja) GM−CSF及びインターフェロンαを用いて生成され、且つ熱処理され死滅したがん細胞を取り込んだ樹状細胞
Bocchia et al. Antitumor vaccination: where we stand
CA2664567C (en) Tumor immunity
Vermeij et al. Immunological and clinical effects of vaccines targeting p53‐overexpressing malignancies
US20150291665A1 (en) Therapy of cancer based on targeting adaptive, innate and/or regulatory component of the immune response
Niethammer et al. An oral DNA vaccine against human carcinoembryonic antigen (CEA) prevents growth and dissemination of Lewis lung carcinoma in CEA transgenic mice
JP4723722B2 (ja) ワクチン用アジュバントとしてのmhcクラスiiリガンドの使用および癌治療におけるlag−3の使用
Baxevanis et al. Toll-like receptor agonists: current status and future perspective on their utility as adjuvants in improving antic ancer vaccination strategies
JP2006523688A (ja) ヌクレオチド・ワクチン組成、ヌクレオチド及び細胞ワクチン組成の産出方法、ワクチン組成、ワクチン組成使用、免疫反応産出方法、疾患の治療または予防方法、抗原提示細胞から成るキット
Ishizaki et al. Heterologous prime/boost immunization with p53-based vaccines combined with toll-like receptor stimulation enhances tumor regression
Singh et al. Cancer immunotherapy using recombinant Listeria monocytogenes: transition from bench to clinic
US20210236545A1 (en) Tumor immunotherapy composition based on antigen-presenting cells activated by attenuated listeria monocytogenes, preparation method therefor and application thereof
Reisfeld et al. DNA vaccines suppress tumor growth and metastases by the induction of anti‐angiogenesis
AU2015233542B2 (en) A medicament for use in a method of inducing or extending a cellular cytotoxic immune response
Lapteva et al. Attraction and activation of dendritic cells at the site of tumor elicits potent antitumor immunity
JP2024509935A (ja) 免疫細胞療法における両親媒性物質の使用及びそのための組成物
CN104136040B (zh) 自体癌细胞疫苗
Mocellin et al. Colorectal cancer vaccines: principles, results, and perspectives
Jia et al. Prophylactic and therapeutic efficacy of an attenuated Listeria monocytogenes-based vaccine delivering HPV16 E7 in a mouse model
Reisfeld et al. DNA vaccines designed to inhibit tumor growth by suppression of angiogenesis
KR20190066028A (ko) 암 치료를 위한 면역원성 화합물
US20220152167A1 (en) Immunogenic formulations for treating cancer
US20070110717A1 (en) Dna vaccines against tumor growth and methods of use thereof
Miller et al. From breast cancer immunobiology to her-2 DNA vaccine and autoimmune sequelae

Legal Events

Date Code Title Description
AS Assignment

Owner name: MOUNT SINAI SCHOOL OF MEDICINE, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BLANDER, JULIE MAGARIAN;GARAUDE, JOHAN;SIGNING DATES FROM 20120828 TO 20120829;REEL/FRAME:028979/0605

STCB Information on status: application discontinuation

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