WO2006036550A2 - Listeria-based and llo-based vaccines - Google Patents

Listeria-based and llo-based vaccines Download PDF

Info

Publication number
WO2006036550A2
WO2006036550A2 PCT/US2005/032682 US2005032682W WO2006036550A2 WO 2006036550 A2 WO2006036550 A2 WO 2006036550A2 US 2005032682 W US2005032682 W US 2005032682W WO 2006036550 A2 WO2006036550 A2 WO 2006036550A2
Authority
WO
WIPO (PCT)
Prior art keywords
another embodiment
protein
fragment
recombinant
antigen
Prior art date
Application number
PCT/US2005/032682
Other languages
French (fr)
Other versions
WO2006036550A3 (en
Inventor
Yvonne Paterson
Reshma Singh
Original Assignee
Trustess Of The University Of Pennsylvania
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
Priority claimed from US10/949,667 external-priority patent/US7794729B2/en
Application filed by Trustess Of The University Of Pennsylvania filed Critical Trustess Of The University Of Pennsylvania
Priority to DK05811815.9T priority Critical patent/DK1804831T3/en
Priority to EP05811815.9A priority patent/EP1804831B1/en
Priority to CA002581331A priority patent/CA2581331A1/en
Priority to JP2007533537A priority patent/JP2008514199A/en
Priority to EP15182979.3A priority patent/EP2980101B1/en
Priority to AU2005289957A priority patent/AU2005289957A1/en
Priority to ES05811815.9T priority patent/ES2566392T3/en
Publication of WO2006036550A2 publication Critical patent/WO2006036550A2/en
Publication of WO2006036550A3 publication Critical patent/WO2006036550A3/en

Links

Classifications

    • 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
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • 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
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/522Bacterial cells; Fungal cells; Protozoal cells avirulent or attenuated
    • 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
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal 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

Definitions

  • This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune response against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strain expressing the antigen.
  • the present invention also provides recombinant peptides comprising a listeriolysin (LLO) protein fragment fused to a Her- 2 protein or fragment thereof, recombinant Listeria strains expressing a Her-2 protein, vaccines and immunogenic compositions comprising same, and methods of inducing an anti-Her-2 immune response and treating and vaccinating against a Her-2-expressing tumor, comprising same
  • Her-2/neu (refe ⁇ ed to henceforth as "Her-2") is a 185 IcDa glycoprotein that is a member of the epidermal growth factor receptor (EGFR) family of tyrosine kinases, and consists of an extracellular domain, a transmembrane domain, and an intracellular domain which is known to be involved in cellular signaling (Bargmann CI et al, Nature 319: 226, 1986; King CR et al, Science 229: 974, 1985). It is overexpressed in 25 to 40% of all breast cancers and is also overexpressed in many cancers of the ovaries, lung, pancreas, and gastrointestinal tract.
  • EGFR epidermal growth factor receptor
  • Her-2 is associated with uncontrolled cell g ⁇ owth and signaling, both of which contribute to the development of tumors Patients with cancers that overexpress Her-2 exhibit tolerance even with detectable humoral, CD8 + T cell, and CD4 + T cell responses directed against Her-2
  • Listeria monocytogenes is an intracellular pathogen that primarily infects antigen presenting cells and has adapted for life in the cytoplasm of these cells.
  • Host cells such as macrophages, actively phagocytose L monocytogenes and the majority of the bacteria are degraded in the phagolysosome.
  • Some of the bacteria escape into the host cytosol by perforating the phagosomal membrane through the action of a hemolysin, listeriolysin O (LLO)
  • L. monocytogenes can polymerize the host actin and pass directly from cell to cell further evading the host immune system and resulting in a negligible antibody response to L monocytogenes SUMMARY OF THE INVENTION
  • This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune response against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strain expressing the antigen
  • the present invention also provides recombinant peptides comprising a lister iolysin (LLO) protein fragment fused to a Her- 2 protein or fragment thereof, recombinant Listeria stiains expressing a Her-2 protein, vaccines and immunogenic compositions comprising same, and methods of inducing an anti-Her-2 immune response and treating and vaccinating against a Her-2-expressing tumor, comprising same.
  • LLO lister iolysin
  • the present invention provides a recombinant polypeptide comprising an
  • N-terminal fragment of a LLO protein fused to a fragment of a Her-2 protein the fragment of a Her-2 protein having a length of about 150 to about 420 amino acids
  • the present invention provides a recombinant polypeptide comprising an N-lerminal fragment of an LLO protein fused to a Her-2 protein or fused to a fragment thereof.
  • the piesent invention provides a method of inducing an anti-Her-2 immune response in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a Hei-2 protein or fused to a fragment thereof, or administering a recombinant nucleotide encoding the recombinant polypeptide, thereby inducing an anti-Her-2 immune response in a subject-
  • the present invention provides a method of impeding a growth of a Her-2-expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her-2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding the growth of a Her-2-expressing tumor in a subject
  • the present invention provides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her-2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
  • the present invention provides a method of impeding a growth of a Her-2-expressing tumor in a subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
  • the present invention piovides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant form of Lislei ia comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject
  • the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumor, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a fragment of the antigen or a recombinant nucleotide encoding the recombinant polypeptide, wherein the antigen has one or more dominant CD8 + T cell epitopes and wherein the fragment does not contain any of the dominant CD8 + T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen-expressing tumor.
  • the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumor, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding a fragment of the antigen, wherein the antigen has one or more dominant CD8 + T cell epitopes and wherein the fragment does not contain any of the dominant CD8 + T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen- expressing tumor
  • Figure 1 Schematic representation of pGG55, used to construct the Lm- ⁇ -LLO-HER-2 vaccines.
  • FIG. 1 Recombinant Listeria monocytogenes is capable of secreting each of Her-2 fragments as a ⁇ LLO-fusion protein.
  • A Map of rat Her-2 fragments.
  • B Confirmation of secretion of the fusion peptides by Western blot. Marker (lane 1), Lm- ⁇ LLO-E7 (lane 2), Lm- ⁇ LLO-ECl (lane 3), Lm- ⁇ LLO-EC2 (lane 4), Lm- ⁇ LLO-EC3 (lane 5), Lm- ⁇ LLO-ICl (lane 6), and Lm- ⁇ LL0-IC2 (lane 7).
  • Lm- ⁇ LLO-EC2 and Lm- ⁇ LL0-EC3;
  • B Lm- ⁇ LLO-ICl , and Lm- ⁇ LL0-IC2.
  • FIG. 4 CD8 "1 T cells participate in Lm-LLO-Her-2 induced tumor stasis. Tumor measurements are shown only for the surviving mice at a given time point.
  • Lm- ⁇ LLO- ECl , Lm- ⁇ LLO-EC2, and Lm- ⁇ LLO-EC3 both depleted and undepleted for CDS + T cells.
  • Lm- ⁇ LLO-ICl and Lm- ⁇ LLO-IC2 both depleted and undepleted for CD8 + T cells.
  • Lm- ⁇ LLO-EC2 induces a 3-fold increase in tetramer + , CD8 + T cells.
  • FVB/N mice were immunized with Lm- ⁇ LLO-EC2 or PBS. Subsequently, splenocytes were stained with an H-2 q Her-2 tetramer, anti-CD8, and anti-CD62L.
  • FIG. 7 Delivery by LM and fusion to ⁇ LLO increases the anti-tumor immune response of Her-2 vaccines. Average tumor diameter for each mouse is depicted. Tumor measurements are shown only for the surviving mice at a given time point.
  • A Lm- ⁇ LLO-EC 1 vs. pcDNA ⁇ LLO-EC1 + GM-CSF
  • B pcDNA ECl + GM-CSF vs. pcDNA ⁇ LLO-EC1 + GM-CSF
  • C pcDNA neu + GM-CSF vs. pcDNA ⁇ LLO-neu + GM-CSF
  • D pcDNA ⁇ LLO-neu + GM-CSF vs. pcDNA ⁇ LLO-EC1 + GM-CSF
  • E pcDNA neu + GM-CSF vs pcDNA ECl + GM-CSF.
  • Lm- ⁇ LLO-Hei-2 vaccine slow the growth of established rat Her-2 expressing tumors in rat Her-2/neu transgenic mice, in which rat Her-2 is expressed as a self-antigen.
  • FIG. 10 Schematic representation of human Her-2 fragments used to create LLO-human
  • This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune iesponse against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strain expressing the antigen.
  • the present invention also provides recombinant peptides comprising a listeriolysin (LLO) protein fragment fused to a Her-2 protein or fragment thereof, recombinant Listeria strains expressing a Her-2 protein, vaccines and immunogenic compositions comprising same, and methods of inducing an anti-Her-2 immune iesponse and treating and vaccinating against a Her-2-expressing tumor, comprising same.
  • LLO listeriolysin
  • compositions of the present invention has utility for inducing formation of antigen-specific T cells (e.g cytotoxic T cells) that recognize and kill tumor cells, thereby arresting the growth of and shrinking tumor cells and treating the resulting cancer (Examples herein).
  • antigen-specific T cells e.g cytotoxic T cells
  • the present invention provides a recombinant polypeptide comprising an
  • the present invention provides a recombinant polypeptide comprising an N-terminal fragment of an LLO protein fused to a Her-2 protein or fused to a fragment thereof.
  • the present invention provides a recombinant polypeptide comprising a fragment of a Her-2 protein.
  • the fragment consists of about
  • the fragment consists of about AA
  • the fragment consists of about AA 479-655 thereof In another embodiment, the fragment consists of about AA 690-1081 thereof. In another embodiment, the fragment consists of about AA 1020-1255 thereof. In other embodiments, the fragment consists of any of the Her-2 fragments mentioned below Each possibility represents a separate embodiment of the piesent invention.
  • the Her-2 protein of methods and compositions of the present invention is a human Her-2 protein.
  • the Her-2 protein is a mouse Her-2 protein.
  • the Her-2 protein is a rat Her-2 piotein.
  • the Her-2 protein is a primate Hei-2 protein.
  • the Her-2 protein is a Her-2 protein of any other animal species known in the art.
  • the Her-2 protein is a variant of a Her-2 protein,
  • the Her-2 protein is a homologue of a Her-2 protein Each possibility represents a separate embodiment of the present invention.
  • the Her-2 protein is a rat Her-2 protein having the sequence:
  • SEQGPPPSNFEGTPTAENPEYLGLDVPV SEQ ID No: 40 was used to create the fragments in Example 1.
  • the Her-2 protein is encoded for by the nucleic acid sequence set forth in SEQ ID No: 41.
  • the Her-2 protein is a human Her-2 protein having the sequence:
  • the Her-2 protein has a sequence set forth in GenBank Accession No. NM_004448 or NM_001005862. These Her-2 proteins have transmembrane (TM) regions spanning AA 653-675 and 623-645, respectively.
  • TM transmembrane
  • a Her-2 protein is a protein referred to as "HER-2/neu,” “Erbb2,” “v- erb-b2,” “c-erb-b2,” “neu,” or “cNeu.”
  • HER-2/neu a protein referred to as "HER-2/neu,” "Erbb2,” “v- erb-b2,” “c-erb-b2,” “neu,” or “cNeu.”
  • the fragment of a Her-2 protein of methods and compositions of the present invention consists of about amino acid (AA) 20-326 (ECl of Example 1 ; SEQ ID No: 35). In another embodiment, the fragment consists of about AA 303-501 (EC2; SEQ ID NO: 35).
  • the fragment consists of about AA 479-655 (EC3; SEQ ID No: 37) of the Her-2 protein.
  • the fragment of a Her-2 protein consists of about AA 690-1081 (ICl ; SEQ ID No: 38) of the Her-2 protein.
  • the fragment consists of about AA 1020- 1255 (IC2; SEQ ID No: 39) of the Her-2 protein.
  • TM domain spans residues 656-689.
  • Her-2 proteins are determined by aligning the TM domains of the other Her-2 proteins and adjusting the AA ranges.
  • the TM region spans AA 623-645
  • the region of this protein corresponding to EC3 is about AA 446-622, determined by subtracting 33 from the AA numbers to account for the 33 AA difference in the extracellular border of the TM domain
  • the region of this protein corresponding to 1C 1 is 646-] 037, determined by subtracting 44 from the numbers to account for the 44 AA difference in the intracellular border of the TM domain
  • corresponding regions of other Hei-2 proteins are determined by alignment with the ends of the protein. Each possibility represents a separate embodiment of the present invention.
  • the fragment is a fragment of the extracellular domain of the Her-2 protein. In another embodiment, the fragment consists of about one-thhd to one-half of the extracellular domain of the Her-2 protein In another embodiment, the fragment consists of about one-tenth to one-fifth thereof. In another embodiment, the fragment consists of about one-fifth to one-fourth thereof. In another embodiment, the fragment consists of about one- fourth to one-third thereof. In another embodiment, the fragment consists of about one-third to one-half thereof. In another embodiment, the fragment consists of about one-half to three quarters thereof. In another embodiment, the fragment consists of about three quarters to the entire extracellular domain. In another embodiment, the fragment consists of about 5-10% thereof.
  • the fragment consists of about 10-15% thereof. In another embodiment, the fragment consists of about 15-20% thereof. In another embodiment, the fragment consists of about 20-25% thereof In another embodiment, the fragment consists of about 25-30% thereof In another embodiment, the fragment consists of about 30-35% thereof In another embodiment, the fragment consists of about 35-40% thereof In another embodiment, the fragment consists of about 45-50% thereof In another embodiment, the fragment consists of about 50-55% thereof. In another embodiment, the fragment consists of about 55-60% thereof In another embodiment, the fragment consists of about 5-15% thereof In another embodiment, the fragment consists of about 10-20% thereof. In another embodiment, the fragment consists of about 15-25% thereof In another embodiment, the fragment consists of about 20-30% thereof.
  • the fragment consists of about 25-35% thereof In another embodiment, the fragment consists of about 30-40% thereof. In another embodiment, the fragment consists of about 35-45% thereof. In another embodiment, the fragment consists of about 45-55% thereof In another embodiment, the fragment consists of about 50-60% thereof. In another embodiment, the fragment consists of about 55-65% thereof, In another embodiment, the fragment consists of about 60-70% thereof. In another embodiment, the fragment consists of about 65-75% thereof. In another embodiment, the fragment consists of about 70-80% thereof. In another embodiment, the fragment consists of about 5-20% thereof. In another embodiment, the fragment consists of about 10-25% thereof. In another embodiment, the fragment consists of about 15-30% thereof.
  • the fragment consists of about 20-35% thereof. In another embodiment, the fragment consists of about 25-40% thereof. In another embodiment, the fragment consists of about 30-45% thereof. In another embodiment, the fragment consists of about 35-50% thereof. In another embodiment, the fragment consists of about 45-60% thereof. In another embodiment, the fragment consists of about 50-65% thereof, In another embodiment, the fragment consists of about 55-70% thereof. In another embodiment, the fragment consists of about 60-759ffthereof. In another embodiment, the fragment consists of about 65-80% thereof. In another embodiment, the fragment consists of about 70-85% thereof. In another embodiment, the fragment consists of about 75-90% thereof. In another embodiment, the fragment consists of about 80-95% thereof.
  • the fragment consists of about 85-100% thereof. In another embodiment, the fragment consists of about 5-25% thereof. In another embodiment,- the fragment consists of about 10-30% thereof, In another embodiment, the fragment consists of about 15-35% thereof. In another embodiment, the fragment consists of about 20-40% thereof. In another embodiment, the fragment consists of about 30-50% thereof In another embodiment, the fragment consists of about 40-60% thereof. In another embodiment, the fragment consists of about 50-70% thereof. In another embodiment, the fragment consists of about 60-80% thereof In another embodiment, the fragment consists of about 70-90% thereof. In another embodiment, the fragment consists of about 80- 100% thereof. In another embodiment, the fragment consists of about 5-35% thereof.
  • the fragment consists of about 10-40% thereof, In another embodiment, the fragment consists of about 15-45% thereof. In another embodiment, the fragment consists of about 20-50% thereof In another embodiment, the fragment consists of about 30-60% thereof. In another embodiment, the fragment consists of about 40-70% thereof. In another embodiment, the fragment consists of about 50-80% thereof. In another embodiment, the fragment consists of about 60-90% thereof. In another embodiment, the fragment consists of about 70-100% thereof. In another embodiment, the fragment consists of about 5-45% thereof In another embodiment, the fragment consists of about 10-50% thereof In another embodiment, the fragment consists of about 20-60% thereof.
  • the fragment consists of about 30-70% thereof In another embodiment, the fragment consists of about 40-80% thereof- In another embodiment, the fragment consists of about 50-90% thereof In another embodiment, the fragment consists of about 60-100% thereof. In anothei embodiment, the fragment consists of about 5-55% thereof In another embodiment, the fragment consists of about 10-60% thereof. In another embodiment, the fragment consists of about 20-70% thereof. In another embodiment, the fragment consists of about 30-80% thereof. In another embodiment, the fiagment consists of about 40-90% thereof. In another embodiment, the fiagment consists of about 50-100% thereof In another embodiment, the fragment consists of about 5-65% thereof. In another embodiment, the fragment consists of about 10-70% thereof.
  • the fiagment consists of about 20-80% thereof In another embodiment, the fragment consists of about 30-90% thereof. In another embodiment, the fragment consists of about 40-100% thereof In another embodiment, the fragment consists of about 5-75% thereof. In another embodiment, the fragment consists of about 10-80% thereof. In another embodiment, the fragment consists of about 20-90% thereof. In another embodiment, the fragment consists of about 30-100% thereof. In another embodiment, the fragment consists of about 10-90% thereof. In another embodiment, the fragment consists of about 20- 100% thereof. In another embodiment, the fragment consists of about 10-100% thereof.
  • the fragment consists of about 5% of the extracellular domain. In another embodiment, the fragment consists of about 6% thereof. In another embodiment, the fragment consists of about 8% thereof. In another embodiment, the fragment consists of about 10% thereof. In another embodiment, the fragment consists of about 12% thereof In another embodiment, the fragment consists of about 15% thereof. In another embodiment, the fragment consists of about 18% thereof. In another embodiment, the fragment consists of about 20% thereof. In another embodiment, the fragment consists of about 25% thereof. In another embodiment, the fragment consists of about 30% thereof In another embodiment, the fiagment consists of about 35% thereof. In another embodiment, the fragment consists of about 40% thereof.
  • the fragment consists of about 45% thereof In another embodiment, the fragment consists of about 50% thereof. In another embodiment, the fragment consists of about 55% thereof. In another embodiment, the fiagment consists of about 60% thereof. In another embodiment, the fragment consists of about 65% thereof. In another embodiment, the fragment consists of about 70% thereof. In another embodiment, the fragment consists of about 75% thereof. In another embodiment, the fragment consists of about 80% thereof. In another embodiment, the fragment consists of about 85% thereof. In another embodiment, the fragment consists of about 90% thereof. In another embodiment, the fragment consists of about 95% thereof In another embodiment, the fragment consists of about 100% thereof. Each possibility represents a separate embodiment of the present invention.
  • the fragment is a fragment of the intracellular domain of the Her-2 protein. In one embodiment, the fragment is from about one-third to one-half of the intracellular domain. In another embodiment, the fragment of the intracellular domain is any of the amounts, fractions, or ranges listed above for the extracellular domain. Each possibility represents a separate embodiment of the present invention
  • the fiagment of a Her-2 protein of methods and compositions of the present invention does not include a signal sequence thereof.
  • omission of the signal sequence enables the Her-2 fragment to be successfully expressed in Listeria, due the high hydrophobicity of the signal sequence
  • the fragment of a Her-2 protein of methods and compositions of the present invention does not include a TM domain thereof.
  • omission of the TM enables the Her-2 fragment to be successfully expressed in Listeria, due the high hydrophobicity of the TM
  • Each possibility represents a separate embodiment of the present invention.
  • Does not include refers, in one embodiment, to not including more than 1 amino acid (AA) of the signal sequence or TM domain. In another embodiment, the terms refers to not including more than 2 AA. In other embodiments, the term refers to not including more than
  • the LLO utilized in methods and compositions of the present invention is, in one embodiment, is a Listeria LLO
  • the Listeria from which the LLO is derived is Listeria monocytogenes (LM).
  • the Listeria is Listeria ivanovii.
  • the Listeria is Listeria welshimeri
  • the Listeria is Listeria seelige ⁇ .
  • the LLO protein is a non-Listerial
  • the LLO protein has the sequence:
  • the full length active LLO protein is 504 residues long
  • the LLO protein has a sequence set forth in GenBank Accession No DQ054588, DQ054589, AY878649, U25452, or U25452
  • the LLO protein is a variant of an LLO protein.
  • the LLO protein is a homologue of an LLO protein.
  • truncated LLO or “ ⁇ LLO'” refers to a fragment of LLO that comprises the PEST-like domain
  • the terms refer to an LLO fragment that does not contain the activation domain at the amino terminus and does not include cystine 484.
  • the LLO fragment consists of a PEST sequence.
  • the LLO fragment comprises a PEST sequence.
  • the LLO fragment consists of about the first 441 amino acids of the LLO protein.
  • the LLO fragment is a non-hemolytic form of the LLO protein.
  • the PEST-like domain referred to above has the sequence set forth in SEQ ID NO: 42.
  • the PEST-like domain is any other PEST-like domain known in the art. Each possibility represents a separate embodiment of the present invention.
  • the LLO fragment consists of about residues 1-25. In another embodiment, the LLO fragment consists of about residues 1-50. In another embodiment, the LLO fragment consists of about residues 1-75 In another embodiment, the LLO fragment consists of about residues 1-100. In another embodiment, the LLO fragment consists of about residues 1-125. In another embodiment, the LLO fragment consists of about residues 1-150. In another embodiment, the LLO fragment consists of about residues 1 175. In another embodiment, the LLO fragment consists of about residues 1 -200. In another embodiment, the LLO fragment consists of about residues 1 -225 In another embodiment, the LLO fragment consists of about residues 1 -250. In another embodiment, the LLO fragment consists of about residues 1-275.
  • the LLO fragment consists of about residues 1 -300. In another embodiment, the LLO fragment consists of about residues 1-325. In another embodiment, the LLO fragment consists of about residues 1-350, In another embodiment, the LLO fragment consists of about residues 1-375 In another embodiment, the LLO fragment consists of about residues 1 -400. In another embodiment, the LLO fragment consists of about residues 1-425. Each possibility represents a separate embodiment of the present invention.
  • the present invention provides a vaccine comprising a recombinant polypeptide of the present invention.
  • the present invention provides a nucleotide molecule encoding a recombinant polypeptide of the present invention. In another embodiment, the present invention provides a vaccine comprising the nucleotide molecule.
  • the present invention provides a nucleotide molecule encoding a recombinant polypeptide of the present invention. In another embodiment, the present invention provides a vaccine comprising a nucleotide molecule or recombinant polypeptide of the present invention.
  • the present invention provides an immunogenic composition comprising a nucleotide molecule or recombinant polypeptide of the present invention.
  • the present invention provides a vector comprising a nucleotide molecule or recombinant polypeptide of the present invention
  • the present invention provides a recombinant form of Listeria comprising a nucleotide molecule of the present invention.
  • the present invention provides a vaccine comprising a recombinant form of Listeria of the present invention
  • the present invention provides a culture of a recombinant form of Listeria of the present invention
  • the Listeria of methods and compositions of the present invention is LM.
  • the Listeria is Listeria ivanovii.
  • the Listeria is Listeria welshime ⁇ .
  • the Listeria is Listeria seeligeri
  • Each type of Listeria represents a separate embodiment of the present invention.
  • the present invention provides a recombinant form of Listeria comprising a nucleotide molecule encoding a Her-2 protein or a fragment thereof.
  • the present invention provides a method of inducing an anli-Her-2 immune response in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a Her-2 protein or fused to a fragment thereof, thereby inducing an anti-Her-2 immune response in a subject.
  • the fusion protein of methods and compositions of the present invention comprises an LLO signal sequence from LLO.
  • the two molecules of the protein are joined directly.
  • the two molecules are joined by a short spacer peptide, consisting of one or more amino acids.
  • the spacer has no specific biological activity other than to join the proteins or to preserve some minimum distance or other spatial relationship between them.
  • the constituent amino acids of the spacer are selected to influence some property of the molecule such as the folding, net charge, or hydrophobicity Each possibility represents a separate embodiment of the present invention.
  • the present invention provides a method of inducing an anti-Her-2 immune response in a subject, comprising administering to the subject a recombinant nucleotide encoding a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a Her-2 protein or fused to a fragment thereof, thereby inducing an anti-Her-2 immune response in a subject.
  • the step of administering a recombinant polypeptide or recombinant nucleotide of the present invention is performed with a recombinant form of Listeria comprising the recombinant nucleotide or expressing the recombinant polypeptide.
  • the administering is performed with a different bacterial vector.
  • the administering is performed with a viral vector.
  • the administering is performed with a DNA vaccine (e.g. a naked DNA vaccine).
  • administration of a recombinant polypeptide of the present invention is performed by producing the protein recombinantly, then administering the recombinant protein to a subject.
  • the immune response elicited by methods and compositions of the present invention comprises a CD8 T cell-mediated response.
  • the immune response consists primarily of a CDS + T cell-mediated response.
  • the only delectable component of the immune response is a CD8 + T cell- mediated response
  • the immune response elicited by methods and compositions of the present ' invention comprises a CD4 + T cell-mediated response.
  • the immune response consists primarily of a CD4 + T cell-mediated response.
  • the only detectable component of the immune response is a CD4 + T cell- mediated response.
  • the CD4 + T cell-mediated response is accompanied by a measurable antibody response against the antigen.
  • the CD4 + T cell-mediated response is not accompanied by a measurable antibody response against the antigen.
  • the immune response elicited by methods and compositions of the present invention comprises an immune response to a subdominant epitope of the antigen.
  • the immune response does not comprise an immune response to a subdominant epitope.
  • the immune response consists primarily of an immune response to a subdominant epitope.
  • the only measurable component of the immune response is an immune response to a subdominant epitope.
  • Methods of measuring immune responses include, e.g. measuring suppression of tumor growth (Examples 2, 5, 8, and 9 herein), flow cytometry (FACS; Example 3), target cell lysis assays (e.g chromium release assay; Examples 4 and 6), the use of tetramers, and others
  • FACS flow cytometry
  • target cell lysis assays e.g chromium release assay; Examples 4 and 6
  • tetramers e.g. chromium release assay
  • the present invention provides a method of impeding a growth of a
  • Her-2-expressing tumor in a subject comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her -2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
  • the present invention provides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her-2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressi ⁇ g tumor in a subject.
  • the present invention provides a method of breaking immune tolerance of a subject to a Her-2-expressing tumor, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her -2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby breaking immune tolerance of a subject to a Her-2-expressing tumor,
  • the present invention provides a method of impeding a growth of a Her-2-expressing rumor in a subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
  • the present invention provides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-cxpressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
  • the present invention provides a method of breaking immune tolerance of a subject to a Hei-2-expressing tumor, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby breaking immune tolerance of a subject to a Her-2-expressing tumor.
  • the present invention provides a method of improving an antigenicity of a Her-2 protein, comprising the step of fusing a nucleotide encoding an N- terminal fragment of a LLO protein to a nucleotide encoding the Her-2 protein or a fragment thereof to create a recombinant nucleotide, thereby improving an antigenicity of a Her-2 protein.
  • Her-2 protein further comprises engineering a Listeria strain to express the recombinant nucleotide
  • a different bacterial vector is used to express the recombinant nucleotide.
  • a viral vector is used to express the recombinant nucleotide.
  • a DNA vaccine e.g. a naked DNA vaccine
  • administration of the LLO-Her-2 fusion peptide encoded by the nucleotide is performed by producing the protein recombinantly, then administering the recombinant protein to a subject.
  • the anti-Her-2 immune response elicted by methods and compositions of the present invention comprises a first immune response against an epitope of the Her-2 protein that is present in the fragment and a second immune response to an epitope of the Her-2 protein that is not present in the fragment, as further detailed hereinbelow.
  • the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumoi, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a fragment of the antigen or a recombinant nucleotide encoding the recombinant polypeptide, wherein the antigen has one or more dominant CDS + T cell epitopes and wherein the fragment does not contain any of the dominant CDS + T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen-expressing tumor
  • the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumor, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding a fragment of the antigen, wherein the antigen has one or more dominant CDS + T cell epitopes and wherein the fragment does not contain any of the dominant CD8 + T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen- expressing tumor.
  • the present invention provides a method of identifying a CD8 + T cell epitope of an antigen, comprising the steps of (a) fusing a nucleotide molecule encoding the antigen to a nucleotide molecule encoding an N-terminal fragment of a LLO protein, thereby creating a recombinant nucleotide an LLO-antigen fusion protein; (b) administering the LLO-antigen fusion to a subject; (c) isolating a CD8 + T cell from the subject; and (d) determining the epitope recognized by the CD8 + T cell; thereby identifying a CDS + T cell epitope of an antigen.
  • the CDS + T cell epitope is a subdominant epitope.
  • Dominant CD8 + T cell epitope refers to an epitope that is recognized by over 30% of the antigen-specific CD8 + T cells that are elicited by vaccination, infection, or a malignant growth with a protein or a pathogen oi cancer cell containing the protein. In another embodiment, the term refers to an epitope recognized by over 35% of the antigen- specific CD8 + T cells that are elicited thereby. In another embodiment, the term refers to an epitope recognized by over 40% of the antigen-specific CDS + T cells In another embodiment, the term refers to an epitope recognized by over 45% of the antigen-specific CDS + T cells.
  • the term refers to an epitope recognized by over 50% of the antigen-specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by over 55% of the antigen-specific CD8 + T cells In another embodiment, the term iefers to an epitope recognized by over 60% of the antigen-specific CDS + T cells. In another embodiment, the term refers to an epitope recognized by over 65% of the antigen- specific CD8 + T cells. In another embodiment, the te ⁇ n refers to an epitope recognized by over 70% of the antigen-specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by over 75% of the antigen-specific CD8 + T cells. In another embodiment, the te ⁇ n refers to an epitope recognized by over 80% of the antigen-specific
  • the term refers to an epitope recognized by over 85% of the antigen-specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by over 90% of the antigen-specific CDS + T cells. In another embodiment, the term refers to an epitope recognized by over 95% of the antigen-specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by over 96% of the antigen- specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by over 97% of the antigen-specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by over 98% of the antigen-specific CDS + T cells.
  • the term refers to an epitope recognized by fewer than 24% of the antigen-specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by over 22% of the antigen- specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by fewer than 20% of the antigen-specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by ovei 18% of the antigen-specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by fewer than 16% of the antigen- specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by ovei 14% of the antigen-specific CD8 + T cells.
  • the term refers to an epitope recognized by over 12% of the antigen-specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by fewer than 10% of the antigen- specific CD8 + T cells. In another embodiment, the term refers to an epitope recognized by over 8% of the antigen-specific CD8 + T cells. In another embodiment, the te ⁇ n refers to an epitope recognized by fewer than 6% of the antigen-specific CDS + T cells. In another embodiment, the term refers to an epitope recognized by fewer than 5% of the antigen- specific CD8 + T cells In another embodiment, the term refers to an epitope recognized by over 4% of the antigen-specific CD8 + T cells.
  • the term refers to an epitope recognized by fewer than 3% of the antigen-specific CDS + T cells. In another embodiment, the term refers to an epitope recognized by fewer than 2% of the antigen- specific CD8 + T cells. In another embodiment, the te ⁇ n refers to an epitope recognized by fewer than 1 % of the antigen-specific CD8 H T cells. In another embodiment, the term refers to an epitope recognized by fewer than 0.5% of the antigen-specific CDS + T cells
  • the "subdominant epitope” refers to an epitope not revealed by other methods of vaccination.
  • Ercolini et al J Immunol 2003, 170: 4273-4280 vaccinated subjects with both neu-expressing tumor cells transfected with GM-CSF and recombinant vaccinia expressing Her-2, yet found a single dominant epitope, AA 420-429
  • the dominant epitope or subdominant epitope is dominant or subdominant, respectively, in the subject being treated. In another embodiment, the dominant epitope or subdominant epitope is dominant or subdominant in a population being treated.
  • Each type of the dominant epitope and subdominant epitope represents a separate embodiment of the present invention.
  • the present invention provides a method of breaking an immune tolerance of a subject to an antigen-expressing tumor, wherein the antigen is expressed at a detectable level on a non-tumor cell of the subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the antigen or fused to a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking an immune tolerance of a subject to an antigen-expressing tumor.
  • the present invention provides a method of breaking an immune tolerance of a subject to an antigen-expressing tumor, wherein the antigen is expressed at a detectable level on a non-tumor cell of the subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking an immune tolerance of a subject to an antigen-expressing tumor.
  • Detectable level refers, in one embodiment, to a level delectable by a standard assay
  • the assay is an immunological assay.
  • the assay is enzyme-linked immunoassay (HLISA).
  • the assay is Western blot
  • the assay is FACS (Example 3)
  • a detectable level is determined relative to the background level of a particular assay.
  • the antigen in methods and compositions of the present invention is, in one embodiment, expressed at a detectable level on a non-rumor cell of the subject In another embodiment, the antigen is expressed at a detectable level on at least a certain percentage (e.g 0.01%, 0 03%, 0 1 %, 0 3%, 1 %, 2%, 3%, or 5%) of non-tumor cells of the subject.
  • “non-tumor cell” refers to a cell outside the body of the tumor.
  • non-tumor cell refers to a non-malignant cell.
  • non- tumor cell refers to a non-transformed cell.
  • the non-tumor cell is a somatic cell, hi another embodiment, the non-tumor cell is a germ cell
  • Each possibility represents a separate embodiment of the present invention.
  • the present invention provides a method of inducing a CD8 + T cell- mediated immune response in a subject against a subdominant CD8 + T cell epitope of an antigen, comprising the steps of (a) fusing a nucleotide molecule encoding the antigen or a fragment thereof to a nucleotide molecule encoding an N-terminal fragment of a LLO protein, thereby creating a recombinant nucleotide encoding an LLO-antigen fusion protein; and (b) administering the recombinant nucleotide or the LLO-antigen fusion to the subject; thereby inducing a CDS + T cell-mediated immune response against a subdominant CD8 + T cell epitope of an antigen.
  • the present invention provides a method of inducing an immune response to a first epitope of an antigen, in an animal expressing the antigen on a tumor or an infectious agent, by vaccinating an animal with a recombinant Listeria expressing a fragment of the antigen, wherein the fragment used in vaccination does not include the first epitope. Rather, the fragment contains a second epitope of the same antigen, against which the animal mounts an immune response.
  • a continuing immune response against the tumor or infectious agent results in recognition of the first epitope by epitope spreading, as shown herein
  • the present invention provides a method of inducing an immune response to a first epitope of an antigen, in an animal expressing the antigen on a tumor or an infectious agent, by vaccinating an animal with a vaccine comprising LLO fused to a fragment of the antigen, wherein the fragment used in vaccination does not include the first epitope. Rather, the fragment contains a second epitope, against which the animal mounts an immune response A continuing immune response against the tumor or infectious agent results in recognition of the first epitope by epitope spreading, as shown herein.
  • the immune response to the first epitope is initialed at least 2 weeks following the step of administering. In another embodiment, the immune response to the first epitope is initiated at least 2 weeks following conclusion of the step of administering.
  • the time frame is 1 week. In another embodiment, the time frame is 10 days. In another embodiment, the time frame is 17 days. In another embodiment, the time frame is 3 weeks. In another embodiment, the time frame is 4 weeks. Each possibility represents a separate embodiment of the present invention.
  • the results of the present invention further demonstrate that vaccination with recombinant antigen-expressing LM induces epitope spreading.
  • vaccination with LLO-antigen fusions even outside the context of LM, induces epitope spreading as well.
  • Each possibility represents a separate embodiment of the present invention.
  • the present invention provides a method of impeding a growth of an antigen-expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a fragment of the antigen oi a recombinant nucleotide encoding the recombinant polypeptide, wheiein the antigen has one or more dominant CDS + T cell epitopes and wherein the fragment does not contain any of the dominant CD8 + T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby impeding a growth of an antigen- expressing tumor in a subject,
  • the present invention provides a method of impeding a growth of an antigen-expressing tumor in a subject, comprising administering to the subject a iecombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding a fragment of the antigen, wherein the antigen has one or more dominant CDS + T cell epitopes and wherein the fragment docs not contain any of the dominant CDS "1 T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby impeding a growth of an antigen-expressing tumor in a subject
  • the antigen of methods of the present invention is a Her -2 protein
  • the antigen is a HPV-16 E7 protein
  • the antigen is bcr/abl.
  • the antigen is HPV E6
  • the antigen is MZ2-E.
  • the antigen is MAGE-I .
  • the antigen is MUC-I .
  • the antigen is NY/ESO-1.
  • the antigen is Wilms tumor antigen.
  • the antigen is telomerase
  • the antigen is Proteinase 3
  • the antigen is Tyrosinase related protein 2.
  • the antigen is HIV-I Gag protein.
  • the antigen is SIV-I Gag protein. In another embodiment, the antigen is HIV-I Env protein. In another embodiment, the antigen is any other tumor antigen known in the art. In another embodiment, the antigen is any other infectious disease antigen known in the art Each possibility represents a separate embodiment of the present invention.
  • the antigen is derived from a tumor or an infectious organism, including, but not limited to fungal pathogens, bacteria, parasites, helminths, viruses, and the like.
  • the antigen is selected from tetanus toxoid, hemagglutinin molecules from influenza virus, diphtheria toxoid, HIV gpl20, HIV gag protein, IgA protease, insulin peptide B, Spongospora subterranea antigen, vibriose antigens, Salmonella antigens, pneumococcus antigens, respiratory syncytial virus antigens, Haemophilus influenza outer membrane proteins, Helicobacter pylori urease, Neisseria meningitidis pilins, N.
  • gonorrhoeae pilins the melanoma-associated antigens (TRP-2, MAGE-I , MAGE-3, gp- 100, tyrosinase, MART-I , HSP-70, beta-HCG), human papilloma virus antigens El and E2 from type HPV-16, -18, -31, -33, -35 oi -45 human papilloma viruses, the tumor antigens CEA, the ras protein, mutated or otherwise, the p53 protein, mutated or otherwise, Mucl, or pSA
  • the antigen is an antigen associated with one of the following diseases; cholera, diphtheria, Haemophilus, hepatitis A, hepatitis B, influenza, measles, meningitis, mumps, pertussis, small pox, pneumococcal pneumonia, polio, rabies, rubella, tetanus, tuberculosis, typhoid, Varicella-zoster, whooping cough3 yellow fever, the immunogens and antigens from Addison's disease, allergies, anaphylaxis, Biuton's syndrome, cancer, including solid and blood borne tumors, eczema, Hashimoto's thyroiditis, polymyositis, dermatomyositis, type 1 diabetes mellitus, acquired immune deficiency syndrome, transplant rejection, such as kidney, heart, pancreas, lung, bone, and liver transplants, Graves' disease, polyendocrine
  • Sjogren's syndrome systemic sclerosis, sclerosing cholangitis, Wegener's granulomatosis, dermatitis herpetiformis, psoriasis, vitiligo, multiple sclerosis, encephalomyelitis, Guillain- Barre syndrome, myasthenia gravis, Lambert-Eaton syndrome, sclera, episclera, uveitis, chronic mucocutaneous candidiasis, urticaria, transient hypogammaglobulinemia of infancy, myeloma, X-linked hyper IgM syndrome, Wiskott-Aldrich syndrome, ataxia telangiectasia, autoimmune hemolytic anemia, autoimmune thrombocytopenia, autoimmune neutropenia, Waldenstrom's macroglobulinemia, amyloidosis, chronic lymphocytic leukemia, non-
  • Hodgkin's lymphoma Hodgkin's lymphoma, malarial circumsporozite protein, microbial antigens, viral antigens, autoantigens, and lesteriosis,
  • the antigen is one of the following tumor antigens: a MAGE (Melanoma-Associated Antigen E) protein, e.g. MAGE 1, MAGE 2, MAGE 3, MAGE 4, a tyrosinase; a mutant ras protein; a mutant p53 protein; p97 melanoma antigen, a ras peptide or p53 peptide associated with advanced cancers; the HPV 16/18 antigens associated with cervical cancers, KLH antigen associated with breast carcinoma, CEA (carcinoembryonic antigen) associated with colorectal cancer, gpl OO, a MARTl antigen associated with melanoma, or the PSA antigen associated with prostate cancer.
  • any of the above antigens can be fused to an LLO fragment.
  • Each of the above antigens represents a separate embodiment of the present invention.
  • the present invention provides a method for suppressing formation of tumors in a host, comprising administering to the host a composition of the present invention, thereby suppressing formation of tumors in a host.
  • the present invention provides a method for inducing formation of tumor-infiltrating CDS + T cells in a host having cancer, comprising administering to the host a composition of the present invention, thereby inducing formation of tumor-infiltrating CD8 + T cells in a host having cancer.
  • the present invention provides a method for inducing formation of cytotoxic T cells in a host having cancer, comprising administering to the host a composition of the present invention, thereby inducing formation of cytotoxic T cells in a host having cancer.
  • the present invention provides a method of reducing an incidence of cancer, comprising administering a composition of the present invention.
  • the present invention provides a method of ameliorating cancer, comprising administering a composition of the present invention.
  • the composition is administered to the cells of the subject ex vivo; in another embodiment, the composition is administered to the cells of a donor ex vivo, in another embodiment, the composition is administered to the cells of a donor in vivo, then is transferred to the subject.
  • the composition is administered to the cells of the subject ex vivo; in another embodiment, the composition is administered to the cells of a donor ex vivo, in another embodiment, the composition is administered to the cells of a donor in vivo, then is transferred to the subject.
  • the cancer treated by a method of the present invention is breast cancer.
  • the cancer is a melanoma In another embodiment, the cancer is pancreatic cancer. In another embodiment, the cancer is ovaiian cancer. In another embodiment, the cancer is gastric cancel. In another embodiment, the cancer is a carcinomatous lesion of the pancreas. In another embodiment, the cancer is pulmonary adenocarcinoma. In another embodiment, the cancer is colorectal adenocarcinoma. In another embodiment, the cancer is pulmonary squamous adenocarcinoma In another embodiment, the cancer is gastric adenocarcinoma. In another embodiment, the cancer is an ovarian surface epithelial neoplasm (e.g a benign, proliferative or malignant variety thereof).
  • ovarian surface epithelial neoplasm e.g a benign, proliferative or malignant variety thereof.
  • the cancer is an oral squamous cell carcinoma. In another embodiment, the cancer is non small-cell lung carcinoma. In another embodiment, the cancer is an endometrial carcinoma. In another embodiment, the cancer is a bladder cancer. In another embodiment, the cancer is a head and neck cancer. In another embodiment, the cancer is a prostate carcinoma.
  • the cancer is an oral squamous cell carcinoma. In another embodiment, the cancer is non small-cell lung carcinoma. In another embodiment, the cancer is an endometrial carcinoma. In another embodiment, the cancer is a bladder cancer. In another embodiment, the cancer is a head and neck cancer. In another embodiment, the cancer is a prostate carcinoma. Each possibility represents a separate embodiment of the present invention.
  • the subject mounts an immune response against the antigen-expressing tumor or target antigen, thereby mediating the anti -tumor effects.
  • the present invention provides an immunogenic composition for treating cancer, the composition comprising a fusion of a truncated LLO to a Her-2 protein.
  • the immunogenic composition further comprises a Listeria strain expressing the fusion Each possibility represents a separate embodiment of the present invention.
  • the present invention provides an immunogenic composition for treating cancer, the composition comprising a Listeria strain expressing a Her-2 protein
  • a treatment protocol of the present invention is therapeutic. In another embodiment, the protocol is prophylactic.
  • the vaccines of the present invention are used to protect people at risk for cancer because of familial genetics or other circumstances that predispose them to certain types of cancer, e g., cervical cancer in women whose husbands have papilloma virus.
  • the vaccines are used as a cancer immunotherapy after debulking of tumor growth by surgery, conventional chemotherapy or radiation treatment Following such treatments, the vaccines of the present invention are administered so that the CTL response to the tumor antigen of the vaccine destroys remaining metastases and prolongs remission from the cancer
  • vaccines of the present invention are used to effect the growth of previously established tumors and to kill existing tumor cells.
  • the vaccines and immunogenic compositions utilized in any of the methods described above have any of the characteristics of vaccines and immunogenic compositions of the present invention. Each characteristic represents a separate embodiment of the present invention.
  • the dosage in the range of 0.001 LDso/dose. In another embodiment, the dosage is 0.002 LDso/dose. In another embodiment the dosage is 0.003 LD 50 /dose In another embodiment the dosage is 0 004 LD 5 o/dose. In another embodiment the dosage is 0 006 LDso/dose In another embodiment the dosage is 0.008
  • the dosage is 0 01 LDso/dose. In another embodiment the dosage is 0 02 LDso/dose. In another embodiment the dosage is 0 03 LD 50 /dose. In one embodiment, the dosage is 0 04 LDso/dose In another embodiment, the dosage is 0.06 LDso/dose. In another embodiment, the dosage 0.08 LDso/dose. In another embodiment, the dosage is 0.1 LDso/dose. In another embodiment, the dosage is 0.15 LDso/dose. In another embodiment, the dosage is 0 2 LDso/dose. In another embodiment, the dosage is 0.25 LD5o/dose. In another embodiment, the dosage is 0 3 LDso/dose. In another embodiment, the dosage is 0.4 LDso/dose. In another embodiment, the dosage is 0 5 LDso/dose.
  • the dosage is 10 7 bacteria/dose. In another embodiment, the dosage is 1.5 x 10 7 bacteria/dose In another embodiment, the dosage is 2 x 10 7 bacteria/dose. In another embodiment, the dosage is 3 x 10 7 bacteria/dose, hi another embodiment, the dosage is 4 x 10 7 bacteria/dose. In another embodiment, the dosage is 6 x 10 7 bacteria/dose In another embodiment, the dosage is 8 x 10 7 bacteria/dose. In another embodiment, the dosage is 1 x 10 8 bacteria/dose. In another embodiment, the dosage is 1 5 x 10 8 bacteria/dose. In another embodiment, the dosage is 2 x 10 8 bacteria/dose. In another embodiment, the dosage is 3 x 10 8 bacteria/dose.
  • the dosage is 4 x 10 s bacteria/dose In another embodiment, the dosage is 6 x 10 bacteria/dose. In another embodiment, the dosage is 8 x 10 8 bacteria/dose In another embodiment, the dosage is 1 x 10 9 bacteria/dose. In another embodiment, the dosage is 1 5 x 10 bacteria/dose In another embodiment, the dosage is 2 x 10 9 bacteria/dose. In another embodiment, the dosage is 3 x 10 9 bacteria/dose
  • the dosage is 5 x 10 9 bacteria/dose. In anothei embodiment, the dosage is 6 x 10 9 bacteria/dose. In another embodiment, the dosage is 8 x 10 9 bacteria/dose In another embodiment, the dosage is 1 x I O 10 bacteria/dose. In another embodiment, the dosage is 1.5 x 10 10 bacteria/dose. In another embodiment, the dosage is 2 x 10 10 bacteria/dose. In another embodiment, the dosage is 3 x 10 10 bacteria/dose. In another embodiment, the dosage is 5 x 10 10 bacteria/dose. In another embodiment, the dosage is 6 x 10 10 bacteria/dose. In another embodiment, the dosage is 8 x 10 10 bacteria/dose. In another embodiment, the dosage is 8 x 10 9 bacteria/dose.
  • the dosage is 1 x 10 1 ' bacteria/dose. In another embodiment, the dosage is 1 5 x 10 " bacteria/dose. In another embodiment, the dosage is 2 x 10 1 ' bacteria/dose In another embodiment, the dosage is 3 x l ⁇ " bacteria/dose. In another embodiment, the dosage is 5 x l ⁇ " bacteria/dose. In another embodiment, the dosage is 6 x 10 1 ' bacteria/dose. In another embodiment, the dosage is 8 x l ⁇ " bacteria/dose. Each possibility represents a separate embodiment of the present invention.
  • the dosage in another embodiment, in the case of recombinant polypeptides, is 1 mg/dose. In another embodiment, the dosage is 1.5 mg/dose. In another embodiment, the dosage is 2 mg/dose. In another embodiment, the dosage is 3 mg/dose. In another embodiment, the dosage is 4 mg/dose. In another embodiment, the dosage is 6 mg/dose. In another embodiment, the dosage is 8 mg/dose. In another embodiment, the dosage is 10 mg/dose. In another embodiment, the dosage is 15 mg/dose. In another embodiment, the dosage is 20 mg/dose. In another embodiment, the dosage is 30 mg/dose. In another embodiment, the dosage is 40 mg/dose In another embodiment, the dosage is 60 mg/dose In another embodiment, the dosage is 80 mg/dose. In another embodiment, the dosage is 100 mg/dose.
  • the dosage is 150 mg/dose. In another embodiment, the dosage is 200 mg/dose. In another embodiment, the dosage is 300 mg/dose In another embodiment, the dosage is 400 mg/dose In another embodiment, the dosage is 600 mg/dose. In another embodiment, the dosage is 800 mg/dose In another embodiment, the dosage is 1000 mg/dose
  • a vaccine or immunogenic composition of the present invention is administered alone to a subject
  • the vaccine or immunogenic composition is administered together with another cancer therapy
  • the recombinant Listeria of methods and compositions of the present invention is, in one embodiment, stably transformed with a construct encoding an antigen or an LLO-antigen fusion.
  • the construct contains a polylinker to facilitate further subcloning.
  • the construct or heterologous gene is integrated into the Listerial chromosome using homologous recombination Techniques for homologous recombination are well known in the art, and are described, for example, in Baloglu S, Boyle SM, et al, Immune responses of mice to vaccinia vims recombinants expressing either Listeria monocytogenes partial listeriolysin or Brucella abortus ribosomal L7/L12 protein. Vet
  • homologous recombination is performed as described in United States Patent No 6,855,320.
  • a recombinant LM strain that expresses E7 was made by chromosomal integration of the E7 gene under the control of the hly promoter and with the inclusion of the hly signal sequence to ensure secretion of the gene product, yielding the recombinant referred to as Lm-AZ/E7.
  • a temperature sensitive plasmid is used to select the recombinants. Each technique represents a separate embodiment of the present invention.
  • the construct or heterologous gene is integrated into the Listerial chromosome using transposon insertion.
  • Techniques fortransposon insertion are well known in the art, and are described, inter alia, by Sun et al. (Infection and Immunity 1990, 58: 3770-3778) in the construction of DP-L967.
  • Transposon mutagenesis has the advantage, in another embodiment, that a stable genomic insertion mutant can be formed but the disadvantage that the position in the genome where the foreign gene has been inserted is unknown.
  • the construct or heterologous gene is integrated into the Listerial chromosome using phage integration sites (Lauer P, Chow MY et al, Construction, characterization, and use of two Listeria monocytogenes site-specific phage integration vectors. J Bacteriol 2002;184(15): 41 77-86).
  • an integrase gene and attachment site of a bacteriophage e.g. U 153 or PSA lisleriophage
  • endogenous prophages are cured from the attachment site utilized prior to integration of the construct or heterologous gene.
  • this method results in single-copy integrants. Each possibility represents a separate embodiment of the present invention.
  • the construct is carried by the Listeria strain on a plasmid. Cloning of the gene into a prfA-containing vector and using this plasmid to complement a prfA(-) Listeria mutant has been used to construct DP-L2028.
  • DP-L2028 is the influenza NP expressing strain used in the tumor protection experiments.
  • Lm-GG/E7 was made by complementing a prfA-deletion mutant with a plasmid containing a copy of the prfA gene and a copy of the E7 gene fused to a form of the LLO (hly) gene truncated to eliminate the hemolytic activity of the enzyme, as described in United States Patent No. 6,565,852
  • Functional LLO is maintained by the organism via the endogenous chromosomal copy of hly.
  • one of several approaches is taken to express the tumor antigen in Listeria
  • a fusion protein of the selected tumor antigen and a Listerial protein, such as PI-PLC, or a construct encoding same is generated.
  • a signal sequence, of a secreted Listerial protein such as hemolysin or phospholipases, is fused to the antigen-encoding gene.
  • the construct is contained in the Listeria strain in an episomal fashion-
  • the foreign antigen is expressed from a vector harbored by the recombinant Listeria strain.
  • one of various promoters is used to express the antigen or fusion protein containing same.
  • an LM promoter is used, e.g. promoters for the genes hly, acta, pica, plcB and mpl, which encode the Listerial proteins hemolysin, act A, phosphotidylinositol-specific phospholipase, phospholipase C, and metalloprotease, respectively.
  • LM promoter e.g. promoters for the genes hly, acta, pica, plcB and mpl, which encode the Listerial proteins hemolysin, act A, phosphotidylinositol-specific phospholipase, phospholipase C, and metalloprotease, respectively.
  • kits of antigens are, in one embodiment, useful in immunotherapy against cancer to compensate for the fact that antigen-loss variants of the tumors can grow out under immune system pressure (Zhang et al, Clin Cancer Res 1998 4: 2669; Kawashima et al, Hum Immunol 1998 59: 1)
  • methods and compositions of the present invention comprise a cocktail of recombinant fusion proteins, each fusion protein comprising a different tumor associated antigen fused to a truncated LLO, or a cocktail of recombinant LM strains, each expressing a different tumor associated antigen.
  • methods and compositions of the present invention utilize a homologue of a Her-2 or LLO sequence of the present invention (e g SEQ ID No: 33, 34,
  • homology when in reference to any protein or peptide, refer in one embodiment, to a percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology, and not considering any conservative substitutions as part of the sequence identity. Methods and computer programs for the alignment are well known in the art.
  • Homology is, in one embodiment, determined by computer algorithm for sequence alignment, by methods well described in the art.
  • computer algorithm analysis of nucleic acid sequence homology may include the utilization of any number of software packages available, such as, for example, the BLAST, DOMAIN, BEAUTY (BLAST Enhanced Alignment Utility), GENPEPT and TREMBL packages.
  • identity' refers to identity to a sequence selected from SEQ ID No: 33, 34, 40, 41 , 43, and 44 of greater than 70%
  • identity refers to identity to a sequence selected from SEQ ID No: 33, 34, 40, 41 , 43, and 44 of greater than 72%.
  • the identity is greater than 75%.
  • the identity is greater than 78%
  • the identity is greater than 80%
  • the identity is greater than 82%.
  • the identity is greater than 83%.
  • the identity is greater than 85% In another embodiment, the identity is greater than 87%.
  • identity is greater than 88% In another embodiment, the identity is greater than 90%.
  • the identity is greater than 92%. In another embodiment, the identity is greater than 93%. In another embodiment, the identity is greater than 95%. In another embodiment, the identity is greater than 96% In another embodiment, the identity is greater than 97%. In another embodiment, the identity is greater than 98%. In another embodiment, the identity is greater than 99%. In another embodiment, the identity is 100%. Each possibility represents a separate embodiment of the present invention.
  • homology is determined is via determination of candidate sequence hybridization, methods of which are well described in the art (See, for example, “Nucleic Acid Hybridization” Hames, B. D., and Higgins S. T., Eds. (1985); Sambrook et al., 2001, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, N Y.; and Ausubel et al., 1989, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N. Y).
  • methods of hybridization may be carried out under moderate to stringent conditions, to the complement of a DNA encoding a native caspase peptide.
  • Hybridization conditions being, for example, overnight incubation at 42 0 C in a solution comprising: 10-20 % formamide, 5 X SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7. 6), 5 X Denhardt's solution, 10 % dextran sulfate, and 20 ⁇ g/ml denatured, sheared salmon sperm DNA.
  • nucleic acids refers to a string of at least two base-sugar-phosphale combinations.
  • the term includes, in one embodiment, DNA and RNA.
  • Nucleotides refers, in one embodiment, to the monomeric units of nucleic acid polymers.
  • RNA may be, in one embodiment, in the form of a tRNA (transfer RNA), snRNA (small nuclear RNA), rRNA (ribosomal RNA), mRNA (messenger RNA), anti-sense RNA, small inhibitory RNA (siRNA), micro RNA (miRNA) and ribozymes.
  • DNA may be in form of plasmid DNA, viral DNA, linear DNA, or chromosomal DNA or derivatives of these groups.
  • RNA may be single, double, triple, or quadruple stranded
  • the term also includes, in another embodiment, artificial nucleic acids that may contain other types of backbones but the same bases
  • the artificial nucleic acid is a PNA (peptide nucleic acid).
  • PNA peptide nucleic acid
  • PNA contain peptide backbones and nucleotide bases and are able to bind, in one embodiment, to both DNA and RNA molecules.
  • the nucleotide is oxetane modified.
  • the nucleotide is modified by replacement of one or more phosphodiester bonds with a phosphorothioate bond.
  • the artificial nucleic acid contains any other variant of the phosphate backbone of native nucleic acids known in the art.
  • the use of phosphothiorate nucleic acids and PNA are known to those skilled in the art, and are described in, for example, Neilsen PE, Curr Opin Struct Biol
  • nucleic acid derivative represents a separate embodiment of the present invention.
  • Protein and/or peptide homology for any amino acid sequence listed herein is determined, in one embodiment, by methods well described in the art, including immunoblot analysis, or via computer algorithm analysis of amino acid sequences, utilizing any of a number of software packages available, via established methods. Some of these packages may include the FASTA, BLAST, MPsrch or Scanps packages, and may employ the use of the Smith and Waterman algorithms, and/or global/local or BLOCKS alignments for analysis, for example. Each method of determining homology represents a separate embodiment of the present invention.
  • the present invention provides a kit comprising a reagent utilized in performing a method of the present invention. In another embodiment, the present invention provides a kit comprising a composition, tool, or instrument of the present invention.
  • contacting refers to directly contacting the cancer cell oi tumor with a composition of the present invention.
  • the terms refer to indirectly contacting the cancer cell or tumor with a composition of the present invention.
  • methods of the present invention include methods in which the subject is contacted with a composition of the present invention after which the composition is brought in contact with the cancer cell or tumor by diffusion or any othei active transport or passive transport piocess known in the art by which compounds circulate within the body. Each possibility represents a separate embodiment of the present invention.
  • compositions containing vaccines and compositions of the present invention can be, in another embodiment, administered to a subject by any method known to a person skilled in the art, such as parenterally, paracancerally, transmucosally, transdermally, intramuscularly, intravenously, intra-dermally, subcutancously, intra- peritonealy, intra-ventricularly, intra-cranially, intra-vaginally or intra-tumorally.
  • the vaccines or compositions are administered orally, and are thus formulated in a form suitable for oral administration, i.e as a solid or a liquid preparation.
  • suitable solid oral formulations include tablets, capsules, pills, granules, pellets and the like.
  • Suitable liquid oral fo ⁇ nulations include solutions, suspensions, dispersions, emulsions, oils and the like.
  • the active ingredient is formulated in a capsule
  • the compositions of the present invention comprise, in addition to the active compound and the inert carrier or diluent, a hard gelating capsule.
  • the vaccines or compositions are administered by intravenous, intra ⁇ arterial, or intra-muscular injection of a liquid preparation.
  • suitable liquid formulations include solutions, suspensions, dispersions, emulsions, oils and the like.
  • the pharmaceutical compositions are administered intravenously and are thus formulated in a form suitable for intravenous administration.
  • the pharmaceutical compositions are administered intra-arterially and are thus formulated in a form suitable for intra-arterial administration.
  • the pharmaceutical compositions are administered intra-muscularly and are thus formulated in a form suitable for intra-muscular administration.
  • the term “treating” refers to curing a disease. In another embodiment, “treating” refers to preventing a disease. In another embodiment, “treating " refers to reducing the incidence of a disease. In another embodiment, “treating " refers to ameliorating symptoms of a disease. In another embodiment, “treating” refers to inducing remission In another embodiment, “treating” refers to slowing the progression of a disease.
  • the terms “reducing”, “suppressing” and “inhibiting” refer in another embodiment to lessening or decreasing Each possibility represents a separate embodiment of the present invention.
  • pGG-55 the backbone of the Listeria Her-2 constructs used in the Examples, was created from pAM401.
  • pAM401 a shuttle vector able to replicate in both gram + and gram " bacteria, contains a gram + chloramphenicol resistance gene and a giam ' tetracycline resistance gene ( Wirth, R et al, .1 Bacterid, 165: 831, 1986).
  • an hly- HPV 16 E7 fusion gene (including the hly promoter and the portion of hly encoding the first 441 amino acids of LLO; referred to below as " ⁇ LLO") and the pluripotent transcription factor, prfA (positive regulatory factor of listeriolysin expression) gene were cloned into pAM401 ( Figure 1 ).
  • L monocytogenes (LM) strains Lm- ⁇ LLO-ECl , Lm- ⁇ LL0-EC2, Lm- ⁇ LLO-EC3, Lm- ⁇ LLO-IC1, and Lm- ⁇ LL0-IC2 each contain a plasmid expressing a fragment of rat Her-2 fused to the Listeria! hl ⁇ gene.
  • Each Her-2 fragment was amplified by PCR from the pNINA plasmid, which contains the full-length rat Her-2 gene, using the following primers. Restriction sites (Xhol in the case of ECl, ICl, and IC2 5' primers; Spel for the 3' primers; and Sail for the EC2 and EC3 5' primers) are underlined, and the FLAG tag sequence in the EC 2 and EC3 the 3' primers are indicated by italics:
  • EC2 5" primer: CCGGGTCGACTGCCCCTACAACTACCTGTCTACG (SEQ IDNo: 3); 3' primer: CCGGACTAGT7T ⁇ CTTGTCA TCGTCGTCCTTGTA GTCCCCAC
  • EC3 5' primer: CCGGGTCGACTGCTTTGTACACACTGTACCTTGG (SEQ ID No: 5); 3' primer: CCGGACTAGT7T ⁇ CTTGTCA TCGTCGTCCTTGTA GTCC
  • ICl 5' primer: CCGGCTCGAGTATACGATGCGTAGGCTGCTGCAGG (SEQ ID No: 7); 3 1 primer: CCGGACTAGTAGCCAGTGGAGATCTGGGGGGCCC (SEQ ID No: 8); 1C2: 5 1 primer: CCGGCTCGAGGGTGACCTGGTAGACGCTGAAG (SEQ ID No: 9) and 3' primer: CCGGACTAGTTACAGGTACATCCAGGCCTAGG (SEQ ID No: 10)
  • BD brain heart infusion medium
  • Champhenicol Chloramphenicol
  • ⁇ LLO-I-ler-2 expressing strains were grown overnight at 37 0 C in Luria-Bertani (LB) medium with 50 microgram per milliliter ( ⁇ g/ml) chloramphenicol. Supernatants were TCA precipitated and resuspended in IX LDS sample buffer (Invitrogen, San Diego, CA). 15 microliter ( ⁇ l) of each sample was loaded on a 4-12% Bis-Tris SDS-PAGE gel (Invitrogen, San Diego, CA).
  • Lm- ⁇ LLO-EC 1 Lm- ⁇ LLO ⁇ C2 Lm- ⁇ LLO-EC3, Lm- ⁇ LLO-ICl, and Lm- ⁇ LLO-lC2 were 83, 70, 68, 92.5, and 74-kDa (lcilodalton), respectively.
  • the strains were attenuated relative to the wild-type 10403S strain, exhibiting virulences comparable to Lm- ⁇ LL0-E7; namely 1x10 8 , 5x10 s , 1x10 9 , 1x10 8 , and 1x10 8 colony forming units (CFU), respectively.
  • mice Six to eight week old female FVB/N mice were purchased from Charles River Laboratories.
  • the FVB/N syngeneic NT-2 tumor cell line derived from a spontaneously occurring mammary tumor in an FVB/N Her-2 tiansgenic mouse (Reilly RT et al, Cane Res 60: 3569, 2000), constitutive! y expresses low levels of rat Her-2 and is tumorigenic in wild type syngeneic mice.
  • NT-2 cells were grown in RPMI 1640 medium with 20% FCS, 10.2 mM HEPES, 2 milliniolar (mM) L-glutamine, 100 micromolar ( ⁇ M) nonessential amino acids, 1 mM sodium pyruvate, 50 U (units)/ml penicillin G, 50 ⁇ g/ml streptomycin, 20 ⁇ g/ml insulin, and 2 ⁇ g/ml gentamycin at 37° C with 5% CO 2 .
  • LM strains expressing ⁇ LLO-Her-2 fusions were compared to PBS and Lm- ⁇ LLO-B7 (negative controls) for theii ability to induce immunity against and reduction of tumors composed of the rat Her-2 expressing tumor line, NT-2.
  • FVB/N mice were injected with NT- 2 tumors, then, on days 7, 14, and 21 following rumor inoculation, were administered 0 1 LD 50 of recombinant LM or PBS.
  • Injection of LM expiessing the ⁇ LLO-Her-2 fusions halted tumor growth aftei the first injection ( Figures 2A and B); the cessation in tumor growth continued through the last timepoint, more than nine weeks after the last Her-2 vaccination.
  • CD8 + T cells were depleted by injection with 0 5 mg of the anti-CD8 antibody 2 43 (Sa ⁇ niento M et al, J Immunol 125(6): 2665-72, 1980) on days 6, 7, 8, 1 1 , 14, 17, 20, and 23 post-tumor injection CDS + T cell populations were reduced by greater than 95% as measured by flow cytometric analysis on day 24.
  • Splenocytes were stained at room temperature (rt) with the tetramer for one hour (hr) at 1 :200 dilution, then at 4° C with anti- CD8 and anti-CD62L antibodies for 30 minutes (min).
  • the CD8 + , CD62L low subset was selected ("gated on"), and percentages of tetramer 4 cells were compared using FlowJo software (Tree Star, Inc, Ashland, OR).
  • FVB/N mice with NT-2 tumors were depleted of CD8 + T cells, beginning 1 day prior to vaccination with Lm- ⁇ LLO-Her-2 vaccines, then vaccinated as described in Example 2.
  • CD8 + -injected mice each of the Lm- ⁇ LLO-Her-2 vaccines lost effectiveness ( Figures 3A and B); while in the non-depleted mice, tumor growth was controlled, as observed in Example 2.
  • NIH 3T3 cells a mouse fibroblast line, were obtained from the American Type Culture Collection (ATCC). The NIH 3T3 and all the derived cells were cultured in DMEM supplemented with 10% FCS, 2 mM L-glutamine, 100 ⁇ M nonessential amino acids, 1 mM sodium pyruvate, 50 U/ml penicillin G, and 50 ⁇ g/ml streptomycin. Culture media for the 3T3-neu cell lines was supplemented with 1 mg/ml G418. Cells were grown at 37° C with
  • wild type 3T3 cells were transduced with overlapping fragments of the rat Her-2 gene, creating nine 3T3 Her-2 fragment lines, and one 3T3 line expressing the full-length rat Her-2.
  • Her-2 fragments were created using the following PCR primers:
  • Fragment 3 (bp 1-508): S'-CCGGGCCGAATTCGCAATGATC (SEQ ID NO: 1 1 ) and 3'- CCCCGAATTCCTACTGAGGGTTCCCACGGATCAA (SEQ ID NO: 12).
  • Fragment 2 (bp 458-886): 5'-
  • GACATGAAGTTGCGGCTCCCTAGTCTCACAGAGATCCTGAAG SEQ ID NO: 13
  • 3'-CCCCGAATTCCTACTCAGGGTTGTGCATGGACTC SEQ ID NO: 14
  • Fragment 4 (bp 1244-1675): 5'- GACATGAAGTTGCGGCTCCCTATCACAGGTTACCTGTACATC (SEQ E ) No: 17) and
  • Fragment 7 (bp 2405-2872), 5'- GACATGAAGTTGCGGCTCCCTTCCACAGTACAGCTGGTGACA(SEQIDNO:23) and3'-CCCCGGTACCCTAGCAGATTGGAGGCTGAGGTAG(SEQIDNO:24)
  • Fragment 8 (bp 2801-3271), 5'- GACATGAAGTTGCGGCTCCCTGATGGAATCCCAGCCCGGGAG(SEQIDNO:25) and3'-CCCCGGTACCCTACCCTTCCGAGGGAGCCAGTGG(SEQIDNO: 26).
  • Fragment 9 (bp 3203-3796), 5'- GACATGAAGTTGCGGCTCCCTGAGCTGACACTGGGCCTGGAG(SEQIDNO:27) and3'-CCCCGGTACCCTATACAGGTACATCCAGGCCTAG(SEQIDNO:28).
  • Fragments 1 -9 span amino acids 1 -165, 148-291 , 274-426, 410-553, 531 -687, 655-820, 797- 952, 929-1085, 1063-1255 of Hei-2, respectively.
  • Each fragment was ligated into the pcDNA3.1 mammalian transfection vector, which contains a cytomegalovirus (CMV) promoter (Invitrogen, Carlsbad, CA), at the multicloning site.
  • CMV cytomegalovirus
  • Constructs were transfected into 3T3 cells using electroporation (20 ⁇ g/1 x 10 7 cells) or Lipofectamine (1 5 ⁇ g/3 x 10 5 cells; Life Technologies).
  • Several clones of each fragment were isolated by limiting dilution. Expression of Her-2 fragments in the clones was determined by RT-PCR
  • FVB/N mice were immunized with 0 1 LD50 of each of the Lm- ⁇ LLO-Her-2 vaccines
  • Splenocytes were harvested 9 days later and cultured for four days with irradiated (20,000 rads) NT-2 tumor cells at a 100:1 ratio of splenocytes to tumor cells with 20 U/ml IL-2 (Roche, Indianapolis, IN).
  • Splenocytes were then used as effector cells in a standard 51 Cr release assay.
  • Target cells were labeled with chiomium-51 ( 51 Cr) and cultured for four hours with splenocytes at effector :target ratios of 200: 1 , 100:1, 50: 1 , and 25: 1 in triplicate.
  • Total counts per minute refers to the total number of counts in the target cell population, measured by lysing the cells after labeling and counting the label. In other words, this is the maximum amount of 51 Cr that could be released
  • a panel of 3T3 cells expressing each of 9 Her-2 fragments were used as target cells in the above-described lysis assay, following vaccination with the co ⁇ esponding ⁇ LLO-Her-2 fusion vaccine.
  • Each vaccine elicited a CTL response to at least one Her-2 fragment.
  • levels of lysis over background reached statistical significance (p ⁇ 0.05).
  • regions of Her-2 containing sub- dominant epitopes were delineated (Table 1 ).
  • vaccination with the Lm- ⁇ LLO-Her-2 fragment vaccines revealed sub-dominant epitopes within Her-2 protein.
  • DNA vaccines were constructed using pcDNA 3.1. Hei-2 and the ECl fragment were amplified by PCR using the following primers:
  • This primer was also used for amplifying unfused ECl fused to ⁇ LLO.
  • Fragments were cloned into the multicloning site of pcDNA3.1, and used to transform Escherichia coli. Bacteria were grown in Luria-Bertani media (BD, Sparks, MD) with 50 micrograms per milliliter ( ⁇ g/ml) ampicillin.
  • Tumor regression experiments were performed as described in Example 2, except that 7 x 10 5 NT-2 cells were utilized, and vaccinations were administered on days 3, 10, and 18.
  • DNA vaccines 50 ⁇ g each of the pcDNA plasmid + and the GM-CSF plasmid or GM-CSF alone) were administered intra-muscularly and Lm administered intraperitoneally.
  • mice were vaccinated with (a) pcDNA 3.1 -full length Her-2 (a DNA vaccine; "pcDNA neu”); (b) pcDNA 3.1 ⁇ LLO-full length Her-2 (pcDNA LLO-neu); (c) pcDNA 3.1-ECl (pcDNA ECl); (d) pcDNA 3 1- ⁇ LLO-EC1 (pcDNA LLO-ECl); or (e) Lm- ⁇ LLO-EC 1 , and a tumor regression expei iment was performed.
  • GM-CSF was included with the DNA vaccines because of its ability to enhance the efficacy of DNA vaccines (McKay PF, Barouch DH et al, Eur J Immunol 2004 Apr;34(4): 101 1 -20.).
  • subdominant CD8 + antigen epitopes can be revealed by either (a) expression by LM; (b) fusion of the antigen to an LLO fragment; or (c) dividing the antigen into smaller fragments.
  • FVB/N are injected with NT-2 tumors, then vaccinated with each of the LM- ⁇ LLO-Hei-2 fragment strains, as described in Example 2, or with DNA ⁇ LLO-Her-2 fragment vaccines, as described in Example 5.
  • Lymphocytes are isolated from the draining lymph nodes of the tumor site at various time points following vaccination. Epitopes recognized by the lymphocytes are determined by a lysis assay, using 3T3 cells expressing each of 9 Her -2 fragments, as described in Example 4
  • mice are injected with NT-2 tumors, then vaccinated with each of the LM- ⁇ LLO-Her-2 fragment strains oi DNA ⁇ LLO-Her-2 fragment vaccines.
  • Lymphocytes are isolated tumor draining lymph nodes at various time points, and the epitopes recognized by the lymphocytes are determined. Emergence is observed of reactivity to epitopes not present in the vaccine fragment. This broadening of the T cell response approximately correlates with temporally tumor regression.
  • Rat Her-2/neu transgenic mice were provided by Dr. William Muller. Young, virgin HER-
  • 2/neu transgenic mice that had not spontaneously developed tumors were injected with 5 x 10 4 NT-2 cells. Because the transgenic mouse is profoundly tolerant to HER-2/neu, the minimum dose required for tumoi growth in 100% of animals is much lower than wild-type mice (Reilly RT, Gott Kunststoff MB el al, Cancer Res. 2000 JuI l ;60(13): 3569-76). NT-2 cells were injected into the subcutaneous space of the flank. Mice received 0.] LD 5O of the
  • the rat Her-2/neu gene differs from the rat neu by 5-6% of amino acid residues, and thus is immunogenic in the mouse (Nagata Y, Furugen R et al, J Immunol. 159: 1336-43).
  • a transgenic mouse that ovei expresses rat Her-2/neu under the transcriptional control of the Mouse Mammary Tumor Virus (MMTV) promoter and enhancer is immunologically tolerant to rat Her-2/neu.
  • MMTV Mouse Mammary Tumor Virus
  • this mouse is considered to be stringent model for human breast cancer and in general for tumors expressing antigens, such as Her-2/neu, that are expressed at low levels in normal tissue (Muller W I. (] 991 ) Expression of activated oncogenes in the murine mammary gland: transgenic models for human breast cancer. Cane Metastasis Rev 10: 217- 27)
  • mice 6-8 week-old HER-2/neu transgenic mice were injected with NT-2 cells, then immunized with each of the LM- ⁇ LLO-Her-2 vaccines, or with PBS or ⁇ LLO-E7 (negative controls). While most control mice had to be sacrificed by day 42 because of their tumor burden, tumor growth was controlled in all of the vaccinated mice (Figure 8).
  • ⁇ LM-LLO-Her-2 vaccines were administered in the following amounts: Lm-LLO-ECl : 1 x
  • Lm- Lm-LLO-EC2 5 x 10 ⁇ 7 cfu
  • LLO-EC3 1 x 10 ⁇ 8 cfu
  • Lm-LLO-lC2 1 x 10 ⁇ 7 cfu
  • Lm-LLO-ICl 1 x 10 ⁇ 7.
  • the ⁇ LM-LLO-Her-2 vaccines were also evaluated for ability to prevent spontaneous tumor growth in the Her-2/neu transgenic mice.
  • HER-2 human Her-2/neu as was used for rat Her-2 (Example 1).
  • the full-length HER-2 gene was split into five fragments, constituting overlapping fragments of the extracellular domain, (EC-I, EC-2 and EC -3) and the cytoplasmic domain (IC-I and IC-2) ( Figure 10). Hydrophobic regions were not included in the constructs. These sequences differed from the rat sequences slightly due to the small disimilarities between the two sequences.
  • the human fragments corresponding to the rat fragments were 22-326, 303- 501, 479-652, 677-1081 , and 1020-1255.
  • the human HER-2 sequences are isolated from a human breast cell cancer line e g. SK-BR3 (ATCC) by reverse transcription PCR (RT-PCR) using standard molecular biology methods
  • RT-PCR reverse transcription PCR
  • total cellular RNA is isolated using the RNeasy Minipreparation® kit (Qiagen) and a cDNA pool is generated (Titan-One-Tube PCR system®, Roche) using an oligo-dT primer
  • the HER-2 sequences of interest are specifically amplified by the second step PCR using the following primers:
  • Restriction sites Xhol (5') and Spel (3') are added to allow for subsequent cloning into the Listeria vaccine vector, and an ochre stop codon is included in the reverse primers to terminate translation of the fusion protein. There are no Xhol or Spel sequences located in these fragments of the human HER-2 gene.
  • PCR products are purified (Qiaquick® PCR Purification Kit, Qiagen) and cloned into intermediate E coli vector pCR2.1 TOPO® (Invitrogen) After transformation of TOP 10 cells (Invitrogen), plasmid containing colonies are identified by PCR using primers Ml 3forward and M 13reverse (Invitrogen), One positive clone for each construct is giown up, plasmid DNA will be prepared (Qiafilter Midipreparation®, Qiagen) and the sequence of the HER-2/neu insert is verified by sequencing.
  • pLLO-E7 is digested completely with Xhol and partially with Spel, thereby removing the E7 gene.
  • the HER-2/neu sequences are digested with the same enzymes, all digests are separated by agarose gel electrophoresis, and the pLLO-E7 vector and the HER-2/neu insert sequences are purified (Qiaquick).
  • the HER-2/neu antigen sequences are ligated into pLLO-E7, and the ligation mix is transformed into d-alanine racemase-deficient E. coli strain MB2159 by electroporalion.
  • Colonies are tested by PCR for the presence of the HER-2/neu sequence and expanded in Luria Broth (LB) media, and plasmid DNA is prepared, then the sequence verified by restriction digestion with EcoRI, Smal, Ncol or with HindIII which yields a specific band pattern for each construct Plasmids are transformed into Listeria strain Lm(DA-) by electroporation, and individual clones are grown up in LB media containing 50 ⁇ g/ml streptomycin. The presence and sequence of the plasmid is again verified by restriction analysis. Expression and secretion of LLO-HER-2/neu fusion proteins is verified by Western blot of TCA-precipitated culture media, using a polyclonal PEST sequence- specific antibody.
  • the LLO-human Her-2 strains are tested for immunogenicity in mice and in human volunteers. Next, the vaccine strains are tested for their ability to protect mice against a challenge with tumor cells expressing human Her-2, as described in the above Examples.
  • Successful strains are administered to humans having Her-2-expressing tumors, and tested for their ability to induce tumor regression. In addition, the strains are tested for their ability to protect human subjects at risk for developing Her-2-expressing cancer, due to genetic or environmental factors.
  • the vaccine strains are found to be immunogenic and to exhibit substantial anti-tumor activity.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune response against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strains expressing the antigen. The present invention also provides recombinant peptides comprising a listeriolysin (LLO) protein fragment used to a Her-2 protein or fragment thereof, recombinant Listeria strains expressing a Her-2 protein, vaccine4s and immunogenic compositions comprising same, and methods of inducing an Her-2 immune response and treating and vaccinating against a Her-2-expressing tumor, comprising same.

Description

LISTERIA-BASED AND LLO-BASED VACCINES
FIELD OF INVENTION
This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune response against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strain expressing the antigen. The present invention also provides recombinant peptides comprising a listeriolysin (LLO) protein fragment fused to a Her- 2 protein or fragment thereof, recombinant Listeria strains expressing a Her-2 protein, vaccines and immunogenic compositions comprising same, and methods of inducing an anti-Her-2 immune response and treating and vaccinating against a Her-2-expressing tumor, comprising same
BACKGROUND OF THE INVENTION
Her-2/neu (refeπed to henceforth as "Her-2") is a 185 IcDa glycoprotein that is a member of the epidermal growth factor receptor (EGFR) family of tyrosine kinases, and consists of an extracellular domain, a transmembrane domain, and an intracellular domain which is known to be involved in cellular signaling (Bargmann CI et al, Nature 319: 226, 1986; King CR et al, Science 229: 974, 1985). It is overexpressed in 25 to 40% of all breast cancers and is also overexpressed in many cancers of the ovaries, lung, pancreas, and gastrointestinal tract. The overexpression of Her-2 is associated with uncontrolled cell gτowth and signaling, both of which contribute to the development of tumors Patients with cancers that overexpress Her-2 exhibit tolerance even with detectable humoral, CD8+ T cell, and CD4+ T cell responses directed against Her-2
Listeria monocytogenes is an intracellular pathogen that primarily infects antigen presenting cells and has adapted for life in the cytoplasm of these cells. Host cells, such as macrophages, actively phagocytose L monocytogenes and the majority of the bacteria are degraded in the phagolysosome. Some of the bacteria escape into the host cytosol by perforating the phagosomal membrane through the action of a hemolysin, listeriolysin O (LLO) Once in the cytosol, L. monocytogenes can polymerize the host actin and pass directly from cell to cell further evading the host immune system and resulting in a negligible antibody response to L monocytogenes SUMMARY OF THE INVENTION
This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune response against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strain expressing the antigen The present invention also provides recombinant peptides comprising a lister iolysin (LLO) protein fragment fused to a Her- 2 protein or fragment thereof, recombinant Listeria stiains expressing a Her-2 protein, vaccines and immunogenic compositions comprising same, and methods of inducing an anti-Her-2 immune response and treating and vaccinating against a Her-2-expressing tumor, comprising same.
In one embodiment, the present invention provides a recombinant polypeptide comprising an
N-terminal fragment of a LLO protein fused to a fragment of a Her-2 protein, the fragment of a Her-2 protein having a length of about 150 to about 420 amino acids,
In another embodiment, the present invention provides a recombinant polypeptide comprising an N-lerminal fragment of an LLO protein fused to a Her-2 protein or fused to a fragment thereof.
In another embodiment, the piesent invention provides a method of inducing an anti-Her-2 immune response in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a Hei-2 protein or fused to a fragment thereof, or administering a recombinant nucleotide encoding the recombinant polypeptide, thereby inducing an anti-Her-2 immune response in a subject-
In another embodiment, the present invention provides a method of impeding a growth of a Her-2-expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her-2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding the growth of a Her-2-expressing tumor in a subject
In another embodiment, the present invention provides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her-2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
In another embodiment, the present invention provides a method of impeding a growth of a Her-2-expressing tumor in a subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
In another embodiment, the present invention piovides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant form of Lislei ia comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject
In another embodiment, the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumor, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a fragment of the antigen or a recombinant nucleotide encoding the recombinant polypeptide, wherein the antigen has one or more dominant CD8+ T cell epitopes and wherein the fragment does not contain any of the dominant CD8+ T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen-expressing tumor.
In another embodiment, the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumor, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding a fragment of the antigen, wherein the antigen has one or more dominant CD8+ T cell epitopes and wherein the fragment does not contain any of the dominant CD8+ T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen- expressing tumor
BRIEF DESCRIPTION OF THE FIGURES
Figure 1. Schematic representation of pGG55, used to construct the Lm-Δ-LLO-HER-2 vaccines.
Figure 2. Recombinant Listeria monocytogenes is capable of secreting each of Her-2 fragments as a ΔLLO-fusion protein. (A) Map of rat Her-2 fragments. (B) Confirmation of secretion of the fusion peptides by Western blot. Marker (lane 1), Lm-ΔLLO-E7 (lane 2), Lm-ΔLLO-ECl (lane 3), Lm-ΔLLO-EC2 (lane 4), Lm-ΔLLO-EC3 (lane 5), Lm-ΔLLO-ICl (lane 6), and Lm-ΔLL0-IC2 (lane 7).
Figure 3. Lm-ΔLLO-Her-2 vaccines each induce a halt in tumor growth of established NT-2 tumors Each data point represents the aveiage of shortest and longest surface tumor diameter of one mouse at a given time point. Mice were sacrificed when the average tumor diameter reached 2.0 cm; tumor measurements are only shown for the surviving mice at a given time point. Representative figures of two experiments are shown. (A) Lm-ΔLLO-EC 1 ,
Lm-ΔLLO-EC2, and Lm-ΔLL0-EC3; (B) Lm-ΔLLO-ICl , and Lm-ΔLL0-IC2.
Figure 4. CD8"1 T cells participate in Lm-LLO-Her-2 induced tumor stasis. Tumor measurements are shown only for the surviving mice at a given time point. (A) Lm-ΔLLO- ECl , Lm-ΔLLO-EC2, and Lm-ΔLLO-EC3 both depleted and undepleted for CDS+ T cells. (B) Lm-ΔLLO-ICl and Lm-ΔLLO-IC2 both depleted and undepleted for CD8+ T cells.
Figure 5. Lm-ΔLLO-EC2 induces a 3-fold increase in tetramer+, CD8+ T cells. FVB/N mice were immunized with Lm-ΔLLO-EC2 or PBS. Subsequently, splenocytes were stained with an H-2q Her-2 tetramer, anti-CD8, and anti-CD62L.
Figure 6 Each of the Lm-ΔLLO-Her-2 vaccine constructs induces similar levels of anti-Her- 2 CTL activity. A. wild type 3T3 (negative control). B. 3T3-neu (full length Her-2). Results are shown as the mean of triplicate cultures, and are representative of 5 experiments
Figure 7 Delivery by LM and fusion to ΔLLO increases the anti-tumor immune response of Her-2 vaccines. Average tumor diameter for each mouse is depicted. Tumor measurements are shown only for the surviving mice at a given time point. (A) Lm-ΔLLO-EC 1 vs. pcDNA ΔLLO-EC1 + GM-CSF, (B) pcDNA ECl + GM-CSF vs. pcDNA ΔLLO-EC1 + GM-CSF, (C) pcDNA neu + GM-CSF vs. pcDNA ΔLLO-neu + GM-CSF, (D) pcDNA ΔLLO-neu + GM-CSF vs. pcDNA ΔLLO-EC1 + GM-CSF, and (E) pcDNA neu + GM-CSF vs pcDNA ECl + GM-CSF.
Figure 8. Lm-ΔLLO-Hei-2 vaccine slow the growth of established rat Her-2 expressing tumors in rat Her-2/neu transgenic mice, in which rat Her-2 is expressed as a self-antigen.
Figure 9. LLO-Her-2 vaccines control spontaneous tumor growth in Her-2/neu transgenic mice.
Figure 10 Schematic representation of human Her-2 fragments used to create LLO-human
Her-2 vaccines.
DETAILED DESCRIPTION OF THE INVENTION
This invention provides methods of treating and vaccinating against an antigen-expressing tumor and inducing an immune iesponse against a sub-dominant epitope of antigen, comprising a fusion of an LLO fragment to the antigen or a recombinant Listeria strain expressing the antigen. The present invention also provides recombinant peptides comprising a listeriolysin (LLO) protein fragment fused to a Her-2 protein or fragment thereof, recombinant Listeria strains expressing a Her-2 protein, vaccines and immunogenic compositions comprising same, and methods of inducing an anti-Her-2 immune iesponse and treating and vaccinating against a Her-2-expressing tumor, comprising same.
As provided herein, the results of the present invention demonstrate that administration of compositions of the present invention has utility for inducing formation of antigen-specific T cells (e.g cytotoxic T cells) that recognize and kill tumor cells, thereby arresting the growth of and shrinking tumor cells and treating the resulting cancer (Examples herein).
In one embodiment, the present invention provides a recombinant polypeptide comprising an
N-terminal fragment of a LLO protein fused to a fragment of a Her-2 protein, the fragment of a Her-2 protein having a length of about 150 to about 420 amino acids. In another embodiment, the present invention provides a recombinant polypeptide comprising an N-terminal fragment of an LLO protein fused to a Her-2 protein or fused to a fragment thereof.
In another embodiment, the present invention provides a recombinant polypeptide comprising a fragment of a Her-2 protein. In one embodiment, the fragment consists of about
AA 20-326 of the Her-2 protein. In another embodiment, the fragment consists of about AA
303-501 thereof In another embodiment, the fragment consists of about AA 479-655 thereof In another embodiment, the fragment consists of about AA 690-1081 thereof. In another embodiment, the fragment consists of about AA 1020-1255 thereof. In other embodiments, the fragment consists of any of the Her-2 fragments mentioned below Each possibility represents a separate embodiment of the piesent invention.
In one embodiment, the Her-2 protein of methods and compositions of the present invention is a human Her-2 protein. In another embodiment, the Her-2 protein is a mouse Her-2 protein. In another embodiment, the Her-2 protein is a rat Her-2 piotein. In another embodiment, the Her-2 protein is a primate Hei-2 protein. In another embodiment, the Her-2 protein is a Her-2 protein of any other animal species known in the art. In another embodiment, the Her-2 protein is a variant of a Her-2 protein, In another embodiment, the Her-2 protein is a homologue of a Her-2 protein Each possibility represents a separate embodiment of the present invention.
In another embodiment, the Her-2 protein is a rat Her-2 protein having the sequence:
MIIMELAA WCRWGFLLALLPPGIAGTQVCTGTDMKLRLPASPETHLDMLRHLYQGCQV VQGNLELTYVP ANASLSFLQDIQEVQGYMLIAHNQVICRVPLQRLRIVRGTQLFEDKYA LAVLDNRDPQDNVAASTPGRTPEGLRELQLRSLTEILKGGVLIRGNPQLCYQDMVLWK DVFRICNNQLAPVDIDTNRSRACPPCAPACKDNHCWGESPEDCQILTGTICTSGCARCK GRLPTDCCHEQCAAGCTGPICHSDCLACLHFNHSGICELHCPALVTYNTDTFESMHNPE GRYTFGASCVTTCPYNYLSTEVGSCTLVCPPNNQEVTAEDGTQRCEKCSKPCARVCYG LGMEHLRGARAITSDNVQEFDGCICKIFGSLAFLPESFDGDPSSGIAPLRPEQLQVFETLE EITGYLYISAWPDSLRDLSVFQNLRIIRGRILHDGAYSLTLQGLGIHSLGLRSLRELGSGL ALIHRNAHLCFVHTVPWDQLFRNPHQALLHSGNRPEEDCGLEGLVCNSLCAHGHCWG PGPTQCVNCSHFLRGQECVEECRVWKGLPREYVSDKRCLPCHPECQPQNSSETCFGSE ADQCAACAHYICDSSSCVARCPSGVKPDLSYMPIWKYPDEEGICQPCPINCTHSCVDLD ERGCPAEQRASPVTFIIATVEGVLLFLILVVVVGILΠCRRRQKJRKYTMRRLLQETELVEP
LTPSGAMPNQAQMRJLKETELRKVKVLGSGAFGTVYKGIWIPDGENVKJPVAIKVLREN TSPKANKEILDEA YVMAGVGSPYVSRLLGICLTSTVQLVTQLMPYGCLLDHVREHRGR LGSQDLLNWCVQIAKGMSYLEDVRLVHRDLAARNVLVKSPNHVKITDFGLARLLDIDE TEYHADGGKVPIKWMALESILRRJ^FTHQSDVWSYGVTVWELMTFGAKPYDGIPAREIP DLLEKGERLPQPPICTIDVYMIMVKCWMIDSECRPRFRELVSEFSRMARDPQRFVVIQN EDLGPSSPMDSTFYRSLLEDDDMGDLVDAEEYLVPQQGFFSPDPTPGTGSTAHRRHRSS STRSGGGELTLGLEPSEEGPPRSPLAPSEGAGSDVFDGDLAMGVTKGLQSLSPHDLSPL QRYSEDPTLPLPPETDGYVAPLACSPQPEYVNQSEVQPQPPLTPEGPLPPVRPAGATLER PKTLSPGKNGVVKJDVFAFGGAVENPEYLVPREGTASPPHPSPAFSPAFDNLYYWDQNS
SEQGPPPSNFEGTPTAENPEYLGLDVPV (SEQ ID NO: 40). SEQ ID No: 40 was used to create the fragments in Example 1. In another embodiment, the Her-2 protein is encoded for by the nucleic acid sequence set forth in SEQ ID No: 41.
In another embodiment, the Her-2 protein is a human Her-2 protein having the sequence:
MELAALCRWGLLLALLPPGAASTQVCTGTDMKLRLPASPETHLDMLRJ-ILYQGCQVVQ GNLELTYLPTNASLSFLQDIQEVQGYVLIAHNQVRQVPLQRLRIVRGTQLFEDNYALAV LDNGDPLNNTTPVTGASPGGLRELQLRSLTEILKGGVLIQRNPQLCYQDTIL WKDIFHKN NQLALTLIDTNRSRACHPCSPMCKGSRCWGESSEDCQSLTRTVCAGGCARCKGPLPTD CCHEQCAAGCTGPKHSDCLACLHFNHSGICELHCPALVTYNTDTFESMPNPEGRYTFG ASCVTACPYNYLSTDVGSCTLVCPLHNQEVTAEDGTQRCEKCSKPCARVCYGLGMEH LREVRAVTSANIQEFAGCKKIFGSLAFLPESFDGDPASNTAPLQPEQLQVFETLEEITGYL YISAWPDSLPDLSVFQNLQVIRGRJLI-INGAYSLTLQGLGISWLGLRSLRELGSGLALIHH NTHLCFVHTVPWDQLFRNPHQALLHTANRPEDECVGEGLACHQLCARGHCWGPGPTQ CVNCSQFLRGQECVEECRVLQGLPREYVNARHCLPCHPECQPQNGSVTCFGPEADQCV
ACAHYKDPPFCVARCPSGVICPDLSYMPIWKFPDEEGACQPCPINCTHSCVDLDDKGCP AEQRASPLTSIISAVVGILLVVVLGVVFGILIKRRQQKIRKYTMRRLLQETELVEPLTPSG AMPNQAQMRILKETELRICVKVLGSGAFGTV YKGIWIPDGENVKIPVAIKVLRENTSPKA NKEILDEA YVMAGVGSPYVSRLLGICLTSTVQLVTQLMPYGCLLDHVRENRGRLGSQD LLN WCMQIAKGMS YLEDVRLVHRDLAARNVLVKSPNI-TVKJTDFGLARLLDIDFTEYH
ADGGKVPIK WMALESILRRRFTHQSDVWSYGVTVWELMTFGAICP YDGIP AREIPDLLE KGERLPQPPICTIDVYMIMVKCWMIDSECRPRFRELVSEFSRM ARDPQRFVVIQNEDLG PASPLDSTFYRSLLEDDDMGDLVDAEEYLVPQQGFFCPDPAPGAGGMVHHRHRSSSTR SGGGDLTLGLEPSEEEAPRSPLAPSEGAGSDVFDGDLGMGAAKGLQSLPTHDPSPLQRY SEDPTVPLPSETDG YV APLTCSPQPEYVNQPDVRPQPPSPREGPLP AARPAGATLERPKT LSPGKNGVVKDVFAFGGAVENPEYLTPQGGAAPQPHPPPAFSPAFDNLYYWDQDPPER GAPPSTFKGTPTAENPEYLGLDVPV (SEQ ID NO: 4.3). SEQ ID No: 43 is used to create the fragments in Example 10. In another embodiment, the Her-2 protein is encoded for by the nucleic acid sequence set forth in SEQ ID No: 44
In other embodiments, the Her-2 protein has a sequence set forth in GenBank Accession No. NM_004448 or NM_001005862. These Her-2 proteins have transmembrane (TM) regions spanning AA 653-675 and 623-645, respectively. The human Her-2 protein set forth in SEQ
ID No: 43 has a TM region spanning 653-676. Thus, in another embodiment, the generation of Her-2 fragments corresponding to those of the present invention from variations of SEQ ID No: 40 such as these requires adjustment of the residue numbers defining the fragments, as described below
In other embodiments, a Her-2 protein is a protein referred to as "HER-2/neu," "Erbb2," "v- erb-b2," "c-erb-b2," "neu," or "cNeu." Each possibility represents a separate embodiment of the present invention
In another embodiment, the fragment of a Her-2 protein of methods and compositions of the present invention consists of about amino acid (AA) 20-326 (ECl of Example 1 ; SEQ ID No: 35). In another embodiment, the fragment consists of about AA 303-501 (EC2; SEQ ID
No: 36) of the Her-2 protein In another embodiment, the fragment consists of about AA 479-655 (EC3; SEQ ID No: 37) of the Her-2 protein. In another embodiment, the fragment of a Her-2 protein consists of about AA 690-1081 (ICl ; SEQ ID No: 38) of the Her-2 protein. In another embodiment, the fragment consists of about AA 1020- 1255 (IC2; SEQ ID No: 39) of the Her-2 protein. Each possibility represents a separate embodiment of the present invention
The AA numbers and ranges listed above are based on the rat Her-2 sequence, for which the
TM domain spans residues 656-689. In another embodiment, corresponding regions of other
Her-2 proteins (e.g. Her-2 proteins from other species) are determined by aligning the TM domains of the other Her-2 proteins and adjusting the AA ranges. For example, for human Hei-2 transcript variant 2, GenBank Accession No NM__001005862, the TM region spans AA 623-645 Thus, in this embodiment, the region of this protein corresponding to EC3 is about AA 446-622, determined by subtracting 33 from the AA numbers to account for the 33 AA difference in the extracellular border of the TM domain Similarly, the region of this protein corresponding to 1C 1 is 646-] 037, determined by subtracting 44 from the numbers to account for the 44 AA difference in the intracellular border of the TM domain In another embodiment, corresponding regions of other Hei-2 proteins are determined by alignment with the ends of the protein. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the fragment is a fragment of the extracellular domain of the Her-2 protein. In another embodiment, the fragment consists of about one-thhd to one-half of the extracellular domain of the Her-2 protein In another embodiment, the fragment consists of about one-tenth to one-fifth thereof. In another embodiment, the fragment consists of about one-fifth to one-fourth thereof. In another embodiment, the fragment consists of about one- fourth to one-third thereof. In another embodiment, the fragment consists of about one-third to one-half thereof. In another embodiment, the fragment consists of about one-half to three quarters thereof. In another embodiment, the fragment consists of about three quarters to the entire extracellular domain. In another embodiment, the fragment consists of about 5-10% thereof. In another embodiment, the fragment consists of about 10-15% thereof. In another embodiment, the fragment consists of about 15-20% thereof. In another embodiment, the fragment consists of about 20-25% thereof In another embodiment, the fragment consists of about 25-30% thereof In another embodiment, the fragment consists of about 30-35% thereof In another embodiment, the fragment consists of about 35-40% thereof In another embodiment, the fragment consists of about 45-50% thereof In another embodiment, the fragment consists of about 50-55% thereof. In another embodiment, the fragment consists of about 55-60% thereof In another embodiment, the fragment consists of about 5-15% thereof In another embodiment, the fragment consists of about 10-20% thereof. In another embodiment, the fragment consists of about 15-25% thereof In another embodiment, the fragment consists of about 20-30% thereof. In another embodiment, the fragment consists of about 25-35% thereof In another embodiment, the fragment consists of about 30-40% thereof. In another embodiment, the fragment consists of about 35-45% thereof. In another embodiment, the fragment consists of about 45-55% thereof In another embodiment, the fragment consists of about 50-60% thereof. In another embodiment, the fragment consists of about 55-65% thereof, In another embodiment, the fragment consists of about 60-70% thereof. In another embodiment, the fragment consists of about 65-75% thereof. In another embodiment, the fragment consists of about 70-80% thereof. In another embodiment, the fragment consists of about 5-20% thereof. In another embodiment, the fragment consists of about 10-25% thereof. In another embodiment, the fragment consists of about 15-30% thereof. In another embodiment, the fragment consists of about 20-35% thereof. In another embodiment, the fragment consists of about 25-40% thereof. In another embodiment, the fragment consists of about 30-45% thereof. In another embodiment, the fragment consists of about 35-50% thereof. In another embodiment, the fragment consists of about 45-60% thereof. In another embodiment, the fragment consists of about 50-65% thereof, In another embodiment, the fragment consists of about 55-70% thereof. In another embodiment, the fragment consists of about 60-759ffthereof. In another embodiment, the fragment consists of about 65-80% thereof. In another embodiment, the fragment consists of about 70-85% thereof. In another embodiment, the fragment consists of about 75-90% thereof. In another embodiment, the fragment consists of about 80-95% thereof. In another embodiment, the fragment consists of about 85-100% thereof. In another embodiment, the fragment consists of about 5-25% thereof. In another embodiment,- the fragment consists of about 10-30% thereof, In another embodiment, the fragment consists of about 15-35% thereof. In another embodiment, the fragment consists of about 20-40% thereof. In another embodiment, the fragment consists of about 30-50% thereof In another embodiment, the fragment consists of about 40-60% thereof. In another embodiment, the fragment consists of about 50-70% thereof. In another embodiment, the fragment consists of about 60-80% thereof In another embodiment, the fragment consists of about 70-90% thereof. In another embodiment, the fragment consists of about 80- 100% thereof. In another embodiment, the fragment consists of about 5-35% thereof. In another embodiment, the fragment consists of about 10-40% thereof, In another embodiment, the fragment consists of about 15-45% thereof. In another embodiment, the fragment consists of about 20-50% thereof In another embodiment, the fragment consists of about 30-60% thereof. In another embodiment, the fragment consists of about 40-70% thereof. In another embodiment, the fragment consists of about 50-80% thereof. In another embodiment, the fragment consists of about 60-90% thereof. In another embodiment, the fragment consists of about 70-100% thereof. In another embodiment, the fragment consists of about 5-45% thereof In another embodiment, the fragment consists of about 10-50% thereof In another embodiment, the fragment consists of about 20-60% thereof. In another embodiment, the fragment consists of about 30-70% thereof In another embodiment, the fragment consists of about 40-80% thereof- In another embodiment, the fragment consists of about 50-90% thereof In another embodiment, the fragment consists of about 60-100% thereof. In anothei embodiment, the fragment consists of about 5-55% thereof In another embodiment, the fragment consists of about 10-60% thereof. In another embodiment, the fragment consists of about 20-70% thereof. In another embodiment, the fragment consists of about 30-80% thereof. In another embodiment, the fiagment consists of about 40-90% thereof. In another embodiment, the fiagment consists of about 50-100% thereof In another embodiment, the fragment consists of about 5-65% thereof. In another embodiment, the fragment consists of about 10-70% thereof. In another embodiment, the fiagment consists of about 20-80% thereof In another embodiment, the fragment consists of about 30-90% thereof. In another embodiment, the fragment consists of about 40-100% thereof In another embodiment, the fragment consists of about 5-75% thereof. In another embodiment, the fragment consists of about 10-80% thereof. In another embodiment, the fragment consists of about 20-90% thereof. In another embodiment, the fragment consists of about 30-100% thereof. In another embodiment, the fragment consists of about 10-90% thereof. In another embodiment, the fragment consists of about 20- 100% thereof. In another embodiment, the fragment consists of about 10-100% thereof.
In another embodiment, the fragment consists of about 5% of the extracellular domain. In another embodiment, the fragment consists of about 6% thereof. In another embodiment, the fragment consists of about 8% thereof. In another embodiment, the fragment consists of about 10% thereof. In another embodiment, the fragment consists of about 12% thereof In another embodiment, the fragment consists of about 15% thereof. In another embodiment, the fragment consists of about 18% thereof. In another embodiment, the fragment consists of about 20% thereof. In another embodiment, the fragment consists of about 25% thereof. In another embodiment, the fragment consists of about 30% thereof In another embodiment, the fiagment consists of about 35% thereof. In another embodiment, the fragment consists of about 40% thereof. In another embodiment, the fragment consists of about 45% thereof In another embodiment, the fragment consists of about 50% thereof. In another embodiment, the fragment consists of about 55% thereof. In another embodiment, the fiagment consists of about 60% thereof. In another embodiment, the fragment consists of about 65% thereof. In another embodiment, the fragment consists of about 70% thereof. In another embodiment, the fragment consists of about 75% thereof. In another embodiment, the fragment consists of about 80% thereof. In another embodiment, the fragment consists of about 85% thereof. In another embodiment, the fragment consists of about 90% thereof. In another embodiment, the fragment consists of about 95% thereof In another embodiment, the fragment consists of about 100% thereof. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the fragment is a fragment of the intracellular domain of the Her-2 protein. In one embodiment, the fragment is from about one-third to one-half of the intracellular domain. In another embodiment, the fragment of the intracellular domain is any of the amounts, fractions, or ranges listed above for the extracellular domain. Each possibility represents a separate embodiment of the present invention
In another embodiment, the fiagment of a Her-2 protein of methods and compositions of the present invention does not include a signal sequence thereof. In one embodiment, omission of the signal sequence enables the Her-2 fragment to be successfully expressed in Listeria, due the high hydrophobicity of the signal sequence Each possibility represents a separate embodiment of the present invention
In another embodiment, the fragment of a Her-2 protein of methods and compositions of the present invention does not include a TM domain thereof. In one embodiment, omission of the TM enables the Her-2 fragment to be successfully expressed in Listeria, due the high hydrophobicity of the TM Each possibility represents a separate embodiment of the present invention.
"Does not include" refers, in one embodiment, to not including more than 1 amino acid (AA) of the signal sequence or TM domain. In another embodiment, the terms refers to not including more than 2 AA. In other embodiments, the term refers to not including more than
3, 4, 5, 6, 7, 8, 9, 1 OJ 2, 14, 16, 18, 20, 22, 24, 26, 28, or 30 AA of the sequence. In another embodiment, none of the sequence is included In anothei embodiment, most of the sequence is not included. In other embodiments, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 AA of the sequence is not included Each possibility represents a separate embodiment of the present invention. The LLO utilized in methods and compositions of the present invention is, in one embodiment, is a Listeria LLO In one embodiment, the Listeria from which the LLO is derived is Listeria monocytogenes (LM). In another embodiment, the Listeria is Listeria ivanovii. In anothei embodiment, the Listeria is Listeria welshimeri In another embodiment, the Listeria is Listeria seeligeή. In another embodiment, the LLO protein is a non-Listerial
LLO protein
In another embodiment, the LLO protein has the sequence:
MKJCIML VFITLIL VSLPIAQQTEAKDASAFNICENSISSMAPPASPPASPKTPIEKXI-IADEID KYIQGLDYNICNNVLVYHGDAVTNVPPRICGYKDGNEYIVVEKKJCKSINQNN ADIQVVN AISSLTYPGALVICANSEL VENQPD VLPVKRDSLTLSIDLPGMTNQDNKIWKNATKSNV NNAVNTLVERWNEKYAQA YPNVSAKIDYDDEMA YSESQLIAICFGTAFKA VNNSLNVN FGAISEGKMQEEVISFKQIYYNVNVNEPTRPSRFFGKAVTKEQLQALGVNAE1NPPAYISS VAYGRQVYLKLSTNSHSTKVICAAFDAAVSGKSVSGDVELTNIIICNSSFKAVIYGGSAK DEVQIIDGNLGDLRDILICKGATFNRETPGVPIA YTTNFLICDNELA VIKNNSEYIETTSICA YTDGiciNiDHSGGYVAQFNis WDEVNYDPEGNEΓVQHICNWSENNICSICLAHFTSSIYLPG
NARNINVYAICECTGLA WEWWRTVIDDRNLPLVKNRMSIWGTTLYPKYSNKVDNPIE
(GenBanlc Accession No. P13128; SEQ ID NO: 34; nucleic acid sequence is set forth in GenBank Accession No Xl 5127; SEQ ID NO: 33). The first 25 amino acids of the proprotein corresponding to this sequence are the signal sequence and are cleaved from LL-O when it is secreted by the bacterium. Thus, in this embodiment, the full length active LLO protein is 504 residues long In other embodiments, the LLO protein has a sequence set forth in GenBank Accession No DQ054588, DQ054589, AY878649, U25452, or U25452 In another embodiment, the LLO protein is a variant of an LLO protein. In another embodiment, the LLO protein is a homologue of an LLO protein. Each possibility represents a separate embodiment of the present invention.
In another embodiment, "truncated LLO" or "ΔLLO'" refers to a fragment of LLO that comprises the PEST-like domain In another embodiment, the terms refer to an LLO fragment that does not contain the activation domain at the amino terminus and does not include cystine 484. In another embodiment, the LLO fragment consists of a PEST sequence. In another embodiment, the LLO fragment comprises a PEST sequence. In another embodiment, the LLO fragment consists of about the first 441 amino acids of the LLO protein. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
In one embodiment, the PEST-like domain referred to above has the sequence set forth in SEQ ID NO: 42. In another embodiment, the PEST-like domain is any other PEST-like domain known in the art. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the LLO fragment consists of about residues 1-25. In another embodiment, the LLO fragment consists of about residues 1-50. In another embodiment, the LLO fragment consists of about residues 1-75 In another embodiment, the LLO fragment consists of about residues 1-100. In another embodiment, the LLO fragment consists of about residues 1-125. In another embodiment, the LLO fragment consists of about residues 1-150. In another embodiment, the LLO fragment consists of about residues 1 175. In another embodiment, the LLO fragment consists of about residues 1 -200. In another embodiment, the LLO fragment consists of about residues 1 -225 In another embodiment, the LLO fragment consists of about residues 1 -250. In another embodiment, the LLO fragment consists of about residues 1-275. In another embodiment, the LLO fragment consists of about residues 1 -300. In another embodiment, the LLO fragment consists of about residues 1-325. In another embodiment, the LLO fragment consists of about residues 1-350, In another embodiment, the LLO fragment consists of about residues 1-375 In another embodiment, the LLO fragment consists of about residues 1 -400. In another embodiment, the LLO fragment consists of about residues 1-425. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a vaccine comprising a recombinant polypeptide of the present invention.
In another embodiment, the present invention provides a nucleotide molecule encoding a recombinant polypeptide of the present invention. In another embodiment, the present invention provides a vaccine comprising the nucleotide molecule.
In another embodiment, the present invention provides a nucleotide molecule encoding a recombinant polypeptide of the present invention. In another embodiment, the present invention provides a vaccine comprising a nucleotide molecule or recombinant polypeptide of the present invention.
In another embodiment, the present invention provides an immunogenic composition comprising a nucleotide molecule or recombinant polypeptide of the present invention.
In another embodiment, the present invention provides a vector comprising a nucleotide molecule or recombinant polypeptide of the present invention
In another embodiment, the present invention provides a recombinant form of Listeria comprising a nucleotide molecule of the present invention.
In another embodiment, the present invention provides a vaccine comprising a recombinant form of Listeria of the present invention
In another embodiment, the present invention provides a culture of a recombinant form of Listeria of the present invention
In another embodiment, the Listeria of methods and compositions of the present invention is LM. In another embodiment, the Listeria is Listeria ivanovii. In another embodiment, the Listeria is Listeria welshimeή. In another embodiment, the Listeria is Listeria seeligeri
Each type of Listeria represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a recombinant form of Listeria comprising a nucleotide molecule encoding a Her-2 protein or a fragment thereof.
In another embodiment, the present invention provides a method of inducing an anli-Her-2 immune response in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a Her-2 protein or fused to a fragment thereof, thereby inducing an anti-Her-2 immune response in a subject.
In one embodiment, the fusion protein of methods and compositions of the present invention comprises an LLO signal sequence from LLO. In another embodiment, the two molecules of the protein (the LLO fragment and the antigen) are joined directly. In another embodiment, the two molecules are joined by a short spacer peptide, consisting of one or more amino acids. In one embodiment, the spacer has no specific biological activity other than to join the proteins or to preserve some minimum distance or other spatial relationship between them. In another embodiment, the constituent amino acids of the spacer are selected to influence some property of the molecule such as the folding, net charge, or hydrophobicity Each possibility represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a method of inducing an anti-Her-2 immune response in a subject, comprising administering to the subject a recombinant nucleotide encoding a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a Her-2 protein or fused to a fragment thereof, thereby inducing an anti-Her-2 immune response in a subject.
In one embodiment, the step of administering a recombinant polypeptide or recombinant nucleotide of the present invention is performed with a recombinant form of Listeria comprising the recombinant nucleotide or expressing the recombinant polypeptide. In another embodiment, the administering is performed with a different bacterial vector. In another embodiment, the administering is performed with a viral vector In another embodiment, the administering is performed with a DNA vaccine (e.g. a naked DNA vaccine). In anothei embodiment, administration of a recombinant polypeptide of the present invention is performed by producing the protein recombinantly, then administering the recombinant protein to a subject. Each possibility represents a separate embodiment of the present invention
In another embodiment, the immune response elicited by methods and compositions of the present invention comprises a CD8 T cell-mediated response. In another embodiment, the immune response consists primarily of a CDS+ T cell-mediated response. In another embodiment, the only delectable component of the immune response is a CD8+ T cell- mediated response
In another embodiment, the immune response elicited by methods and compositions of the present' invention comprises a CD4+ T cell-mediated response. In another embodiment, the immune response consists primarily of a CD4+ T cell-mediated response. In another embodiment, the only detectable component of the immune response is a CD4+ T cell- mediated response. In another embodiment, the CD4+ T cell-mediated response is accompanied by a measurable antibody response against the antigen. In another embodiment, the CD4+ T cell-mediated response is not accompanied by a measurable antibody response against the antigen.
In another embodiment, the immune response elicited by methods and compositions of the present invention comprises an immune response to a subdominant epitope of the antigen. In another embodiment, the immune response does not comprise an immune response to a subdominant epitope. In another embodiment, the immune response consists primarily of an immune response to a subdominant epitope. In another embodiment, the only measurable component of the immune response is an immune response to a subdominant epitope.
Each type of immune response represents a separate embodiment of the present invention
Methods of measuring immune responses are well known in the art, and include, e.g. measuring suppression of tumor growth (Examples 2, 5, 8, and 9 herein), flow cytometry (FACS; Example 3), target cell lysis assays (e.g chromium release assay; Examples 4 and 6), the use of tetramers, and others Each method represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a method of impeding a growth of a
Her-2-expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her -2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
In another embodiment, the present invention provides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her-2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressiπg tumor in a subject.
In another embodiment, the present invention provides a method of breaking immune tolerance of a subject to a Her-2-expressing tumor, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the Her -2 protein or a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby breaking immune tolerance of a subject to a Her-2-expressing tumor,
In another embodiment, the present invention provides a method of impeding a growth of a Her-2-expressing rumor in a subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
In another embodiment, the present invention provides a method of shrinking a Her-2- expressing tumor in a subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-cxpressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
In another embodiment, the present invention provides a method of breaking immune tolerance of a subject to a Hei-2-expressing tumor, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen or a fragment thereof, whereby the subject mounts an immune response against the Her-2-expressing tumor, thereby breaking immune tolerance of a subject to a Her-2-expressing tumor.
In another embodiment, the present invention provides a method of improving an antigenicity of a Her-2 protein, comprising the step of fusing a nucleotide encoding an N- terminal fragment of a LLO protein to a nucleotide encoding the Her-2 protein or a fragment thereof to create a recombinant nucleotide, thereby improving an antigenicity of a Her-2 protein.
In another embodiment, a method of the present invention of improving an antigenicity of a
Her-2 protein further comprises engineering a Listeria strain to express the recombinant nucleotide In another embodiment, a different bacterial vector is used to express the recombinant nucleotide. In another embodiment, a viral vector is used to express the recombinant nucleotide In another embodiment, a DNA vaccine (e.g. a naked DNA vaccine) is used to express the recombinant nucleotide. In another embodiment, administration of the LLO-Her-2 fusion peptide encoded by the nucleotide is performed by producing the protein recombinantly, then administering the recombinant protein to a subject. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the anti-Her-2 immune response elicted by methods and compositions of the present invention comprises a first immune response against an epitope of the Her-2 protein that is present in the fragment and a second immune response to an epitope of the Her-2 protein that is not present in the fragment, as further detailed hereinbelow.
In another embodiment, the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumoi, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a fragment of the antigen or a recombinant nucleotide encoding the recombinant polypeptide, wherein the antigen has one or more dominant CDS+ T cell epitopes and wherein the fragment does not contain any of the dominant CDS+ T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen-expressing tumor
In another embodiment, the present invention provides a method of breaking immune tolerance of a subject to an antigen-expressing tumor, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding a fragment of the antigen, wherein the antigen has one or more dominant CDS+ T cell epitopes and wherein the fragment does not contain any of the dominant CD8+ T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking immune tolerance of a subject to an antigen- expressing tumor.
In another embodiment, the present invention provides a method of identifying a CD8+ T cell epitope of an antigen, comprising the steps of (a) fusing a nucleotide molecule encoding the antigen to a nucleotide molecule encoding an N-terminal fragment of a LLO protein, thereby creating a recombinant nucleotide an LLO-antigen fusion protein; (b) administering the LLO-antigen fusion to a subject; (c) isolating a CD8+ T cell from the subject; and (d) determining the epitope recognized by the CD8+ T cell; thereby identifying a CDS+ T cell epitope of an antigen. In one embodiment, the CDS+ T cell epitope is a subdominant epitope. Each possibility represents a separate embodiment of the present invention.
"Dominant CD8+ T cell epitope," in one embodiment, refers to an epitope that is recognized by over 30% of the antigen-specific CD8+ T cells that are elicited by vaccination, infection, or a malignant growth with a protein or a pathogen oi cancer cell containing the protein. In another embodiment, the term refers to an epitope recognized by over 35% of the antigen- specific CD8+ T cells that are elicited thereby. In another embodiment, the term refers to an epitope recognized by over 40% of the antigen-specific CDS+ T cells In another embodiment, the term refers to an epitope recognized by over 45% of the antigen-specific CDS+ T cells. In another embodiment, the term refers to an epitope recognized by over 50% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 55% of the antigen-specific CD8+ T cells In another embodiment, the term iefers to an epitope recognized by over 60% of the antigen-specific CDS+ T cells. In another embodiment, the term refers to an epitope recognized by over 65% of the antigen- specific CD8+ T cells. In another embodiment, the teπn refers to an epitope recognized by over 70% of the antigen-specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by over 75% of the antigen-specific CD8+ T cells. In another embodiment, the teπn refers to an epitope recognized by over 80% of the antigen-specific
CDS+ T cells. In another embodiment, the term refers to an epitope recognized by over 85% of the antigen-specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by over 90% of the antigen-specific CDS+ T cells. In another embodiment, the term refers to an epitope recognized by over 95% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 96% of the antigen- specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by over 97% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 98% of the antigen-specific CDS+ T cells.
"Subdominant CD8+ T cell epitope," in one embodiment, refers to an epitope recognized by fewer than 30% of the antigen-specific CDS+ T cells that are elicited by vaccination, infection, or a malignant growth with a protein or a pathogen or cancer cell containing the protein. In another embodiment, the term refers to an epitope recognized by fewer than 28% of the antigen-specific CDS+ T cells. In another embodiment, the term refers to an epitope recognized by over 26% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 24% of the antigen-specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by over 22% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 20% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by ovei 18% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 16% of the antigen- specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by ovei 14% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 12% of the antigen-specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by fewer than 10% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 8% of the antigen-specific CD8+ T cells. In another embodiment, the teπn refers to an epitope recognized by fewer than 6% of the antigen-specific CDS+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 5% of the antigen- specific CD8+ T cells In another embodiment, the term refers to an epitope recognized by over 4% of the antigen-specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 3% of the antigen-specific CDS+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 2% of the antigen- specific CD8+ T cells. In another embodiment, the teπn refers to an epitope recognized by fewer than 1 % of the antigen-specific CD8H T cells. In another embodiment, the term refers to an epitope recognized by fewer than 0.5% of the antigen-specific CDS+ T cells
In another embodiment, the "subdominant epitope" refers to an epitope not revealed by other methods of vaccination. For example, Ercolini et al (J Immunol 2003, 170: 4273-4280) vaccinated subjects with both neu-expressing tumor cells transfected with GM-CSF and recombinant vaccinia expressing Her-2, yet found a single dominant epitope, AA 420-429
By contrast, use of the ΔLLO-Hei-2 fusions in the experiments described herein revealed additional epitopes in addition to AA 420-429. Delivering the fusions with recombinant LM reveals yet more epitopes.
In another embodiment, the dominant epitope or subdominant epitope is dominant or subdominant, respectively, in the subject being treated. In another embodiment, the dominant epitope or subdominant epitope is dominant or subdominant in a population being treated.
Each type of the dominant epitope and subdominant epitope represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a method of breaking an immune tolerance of a subject to an antigen-expressing tumor, wherein the antigen is expressed at a detectable level on a non-tumor cell of the subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to the antigen or fused to a fragment thereof or a recombinant nucleotide encoding the recombinant polypeptide, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking an immune tolerance of a subject to an antigen-expressing tumor.
In another embodiment, the present invention provides a method of breaking an immune tolerance of a subject to an antigen-expressing tumor, wherein the antigen is expressed at a detectable level on a non-tumor cell of the subject, comprising administering to the subject a recombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding the antigen, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby breaking an immune tolerance of a subject to an antigen-expressing tumor.
"Detectable level" refers, in one embodiment, to a level delectable by a standard assay In one embodiment, the assay is an immunological assay. In one embodiment, the assay is enzyme-linked immunoassay (HLISA). In another embodiment, the assay is Western blot
(Example 1). In another embodiment, the assay is FACS (Example 3) In another embodiment, a detectable level is determined relative to the background level of a particular assay. Methods for performing each of these techniques are well known to those skilled in the art, and each technique represents a separate embodiment of the present invention.
The antigen in methods and compositions of the present invention is, in one embodiment, expressed at a detectable level on a non-rumor cell of the subject In another embodiment, the antigen is expressed at a detectable level on at least a certain percentage (e.g 0.01%, 0 03%, 0 1 %, 0 3%, 1 %, 2%, 3%, or 5%) of non-tumor cells of the subject. In one embodiment, "non-tumor cell" refers to a cell outside the body of the tumor. In another embodiment, "non-tumor cell" refers to a non-malignant cell. In another embodiment, "non- tumor cell" refers to a non-transformed cell. In another embodiment, the non-tumor cell is a somatic cell, hi another embodiment, the non-tumor cell is a germ cell Each possibility represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a method of inducing a CD8+ T cell- mediated immune response in a subject against a subdominant CD8+ T cell epitope of an antigen, comprising the steps of (a) fusing a nucleotide molecule encoding the antigen or a fragment thereof to a nucleotide molecule encoding an N-terminal fragment of a LLO protein, thereby creating a recombinant nucleotide encoding an LLO-antigen fusion protein; and (b) administering the recombinant nucleotide or the LLO-antigen fusion to the subject; thereby inducing a CDS+ T cell-mediated immune response against a subdominant CD8+ T cell epitope of an antigen.
In another embodiment, the present invention provides a method of inducing an immune response to a first epitope of an antigen, in an animal expressing the antigen on a tumor or an infectious agent, by vaccinating an animal with a recombinant Listeria expressing a fragment of the antigen, wherein the fragment used in vaccination does not include the first epitope. Rather, the fragment contains a second epitope of the same antigen, against which the animal mounts an immune response. A continuing immune response against the tumor or infectious agent results in recognition of the first epitope by epitope spreading, as shown herein
In another embodiment, the present invention provides a method of inducing an immune response to a first epitope of an antigen, in an animal expressing the antigen on a tumor or an infectious agent, by vaccinating an animal with a vaccine comprising LLO fused to a fragment of the antigen, wherein the fragment used in vaccination does not include the first epitope. Rather, the fragment contains a second epitope, against which the animal mounts an immune response A continuing immune response against the tumor or infectious agent results in recognition of the first epitope by epitope spreading, as shown herein.
In one embodiment, the immune response to the first epitope is initialed at least 2 weeks following the step of administering. In another embodiment, the immune response to the first epitope is initiated at least 2 weeks following conclusion of the step of administering. In another embodiment, the time frame is 1 week. In another embodiment, the time frame is 10 days. In another embodiment, the time frame is 17 days. In another embodiment, the time frame is 3 weeks. In another embodiment, the time frame is 4 weeks. Each possibility represents a separate embodiment of the present invention.
As provided herein, the results of the present invention further demonstrate that vaccination with recombinant antigen-expressing LM induces epitope spreading. In another embodiment, vaccination with LLO-antigen fusions, even outside the context of LM, induces epitope spreading as well. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a method of impeding a growth of an antigen-expressing tumor in a subject, comprising administering to the subject a recombinant polypeptide comprising an N-terminal fragment of a LLO protein fused to a fragment of the antigen oi a recombinant nucleotide encoding the recombinant polypeptide, wheiein the antigen has one or more dominant CDS+ T cell epitopes and wherein the fragment does not contain any of the dominant CD8+ T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby impeding a growth of an antigen- expressing tumor in a subject,
In another embodiment, the present invention provides a method of impeding a growth of an antigen-expressing tumor in a subject, comprising administering to the subject a iecombinant form of Listeria comprising a recombinant nucleotide, the recombinant nucleotide encoding a fragment of the antigen, wherein the antigen has one or more dominant CDS+ T cell epitopes and wherein the fragment docs not contain any of the dominant CDS"1 T cell epitopes, whereby the subject mounts an immune response against the antigen-expressing tumor, thereby impeding a growth of an antigen-expressing tumor in a subject
In another embodiment, the antigen of methods of the present invention is a Her -2 protein In another embodiment, the antigen is a HPV-16 E7 protein In another embodiment, the antigen is bcr/abl. In another embodiment, the antigen is HPV E6 In another embodiment, the antigen is MZ2-E. In another embodiment, the antigen is MAGE-I . In another embodiment, the antigen is MUC-I . In another embodiment, the antigen is NY/ESO-1. In another embodiment, the antigen is Wilms tumor antigen. In another embodiment, the antigen is telomerase In another embodiment, the antigen is Proteinase 3 In another embodiment, the antigen is Tyrosinase related protein 2. In another embodiment, the antigen is HIV-I Gag protein. In another embodiment, the antigen is SIV-I Gag protein. In another embodiment, the antigen is HIV-I Env protein. In another embodiment, the antigen is any other tumor antigen known in the art. In another embodiment, the antigen is any other infectious disease antigen known in the art Each possibility represents a separate embodiment of the present invention.
In other embodiments, the antigen is derived from a tumor or an infectious organism, including, but not limited to fungal pathogens, bacteria, parasites, helminths, viruses, and the like. In other embodiments, the antigen is selected from tetanus toxoid, hemagglutinin molecules from influenza virus, diphtheria toxoid, HIV gpl20, HIV gag protein, IgA protease, insulin peptide B, Spongospora subterranea antigen, vibriose antigens, Salmonella antigens, pneumococcus antigens, respiratory syncytial virus antigens, Haemophilus influenza outer membrane proteins, Helicobacter pylori urease, Neisseria meningitidis pilins, N. gonorrhoeae pilins, the melanoma-associated antigens (TRP-2, MAGE-I , MAGE-3, gp- 100, tyrosinase, MART-I , HSP-70, beta-HCG), human papilloma virus antigens El and E2 from type HPV-16, -18, -31, -33, -35 oi -45 human papilloma viruses, the tumor antigens CEA, the ras protein, mutated or otherwise, the p53 protein, mutated or otherwise, Mucl, or pSA
In other embodiments, the antigen is an antigen associated with one of the following diseases; cholera, diphtheria, Haemophilus, hepatitis A, hepatitis B, influenza, measles, meningitis, mumps, pertussis, small pox, pneumococcal pneumonia, polio, rabies, rubella, tetanus, tuberculosis, typhoid, Varicella-zoster, whooping cough3 yellow fever, the immunogens and antigens from Addison's disease, allergies, anaphylaxis, Biuton's syndrome, cancer, including solid and blood borne tumors, eczema, Hashimoto's thyroiditis, polymyositis, dermatomyositis, type 1 diabetes mellitus, acquired immune deficiency syndrome, transplant rejection, such as kidney, heart, pancreas, lung, bone, and liver transplants, Graves' disease, polyendocrine autoimmune disease, hepatitis, microscopic polyarteritis, polyarteritis nodosa, pemphigus, primary biliary cirrhosis, pernicious anemia, coeliac disease, antibody-mediated nephritis, glomerulonephritis, rheumatic diseases, systemic lupus erthematosus. rheumatoid arthritis, seronegative spondylarthritides, rhinitis,
Sjogren's syndrome, systemic sclerosis, sclerosing cholangitis, Wegener's granulomatosis, dermatitis herpetiformis, psoriasis, vitiligo, multiple sclerosis, encephalomyelitis, Guillain- Barre syndrome, myasthenia gravis, Lambert-Eaton syndrome, sclera, episclera, uveitis, chronic mucocutaneous candidiasis, urticaria, transient hypogammaglobulinemia of infancy, myeloma, X-linked hyper IgM syndrome, Wiskott-Aldrich syndrome, ataxia telangiectasia, autoimmune hemolytic anemia, autoimmune thrombocytopenia, autoimmune neutropenia, Waldenstrom's macroglobulinemia, amyloidosis, chronic lymphocytic leukemia, non-
Hodgkin's lymphoma, malarial circumsporozite protein, microbial antigens, viral antigens, autoantigens, and lesteriosis,
In other embodiments, the antigen is one of the following tumor antigens: a MAGE (Melanoma-Associated Antigen E) protein, e.g. MAGE 1, MAGE 2, MAGE 3, MAGE 4, a tyrosinase; a mutant ras protein; a mutant p53 protein; p97 melanoma antigen, a ras peptide or p53 peptide associated with advanced cancers; the HPV 16/18 antigens associated with cervical cancers, KLH antigen associated with breast carcinoma, CEA (carcinoembryonic antigen) associated with colorectal cancer, gpl OO, a MARTl antigen associated with melanoma, or the PSA antigen associated with prostate cancer.
The skilled artisan will appreciate that any of the above antigens can be fused to an LLO fragment. Each of the above antigens represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a method for suppressing formation of tumors in a host, comprising administering to the host a composition of the present invention, thereby suppressing formation of tumors in a host.
In another embodiment, the present invention provides a method for inducing formation of tumor-infiltrating CDS+ T cells in a host having cancer, comprising administering to the host a composition of the present invention, thereby inducing formation of tumor-infiltrating CD8+ T cells in a host having cancer.
In another embodiment, the present invention provides a method for inducing formation of cytotoxic T cells in a host having cancer, comprising administering to the host a composition of the present invention, thereby inducing formation of cytotoxic T cells in a host having cancer.
In another embodiment, the present invention provides a method of reducing an incidence of cancer, comprising administering a composition of the present invention. In another embodiment, the present invention provides a method of ameliorating cancer, comprising administering a composition of the present invention. Each possibility represents a separate embodiment of the present invention.
In one embodiment of methods of the present invention, the composition is administered to the cells of the subject ex vivo; in another embodiment, the composition is administered to the cells of a donor ex vivo, in another embodiment, the composition is administered to the cells of a donor in vivo, then is transferred to the subject. Each possibility represents a separate embodiment of the present invention
In one embodiment, the cancer treated by a method of the present invention is breast cancer.
In another embodiment, the cancer is a melanoma In another embodiment, the cancer is pancreatic cancer. In another embodiment, the cancer is ovaiian cancer. In another embodiment, the cancer is gastric cancel. In another embodiment, the cancer is a carcinomatous lesion of the pancreas. In another embodiment, the cancer is pulmonary adenocarcinoma. In another embodiment, the cancer is colorectal adenocarcinoma. In another embodiment, the cancer is pulmonary squamous adenocarcinoma In another embodiment, the cancer is gastric adenocarcinoma. In another embodiment, the cancer is an ovarian surface epithelial neoplasm (e.g a benign, proliferative or malignant variety thereof). In another embodiment, the cancer is an oral squamous cell carcinoma. In another embodiment, the cancer is non small-cell lung carcinoma. In another embodiment, the cancer is an endometrial carcinoma. In another embodiment, the cancer is a bladder cancer. In another embodiment, the cancer is a head and neck cancer. In another embodiment, the cancer is a prostate carcinoma. Each possibility represents a separate embodiment of the present invention.
In another embodiment of the methods of the present invention, the subject mounts an immune response against the antigen-expressing tumor or target antigen, thereby mediating the anti -tumor effects.
In another embodiment, the present invention provides an immunogenic composition for treating cancer, the composition comprising a fusion of a truncated LLO to a Her-2 protein. In another embodiment, the immunogenic composition further comprises a Listeria strain expressing the fusion Each possibility represents a separate embodiment of the present invention.
In another embodiment, the present invention provides an immunogenic composition for treating cancer, the composition comprising a Listeria strain expressing a Her-2 protein
In one embodiment, a treatment protocol of the present invention is therapeutic. In another embodiment, the protocol is prophylactic. In another embodiment, the vaccines of the present invention are used to protect people at risk for cancer because of familial genetics or other circumstances that predispose them to certain types of cancer, e g., cervical cancer in women whose husbands have papilloma virus. In another embodiment, the vaccines are used as a cancer immunotherapy after debulking of tumor growth by surgery, conventional chemotherapy or radiation treatment Following such treatments, the vaccines of the present invention are administered so that the CTL response to the tumor antigen of the vaccine destroys remaining metastases and prolongs remission from the cancer In another embodiment, vaccines of the present invention are used to effect the growth of previously established tumors and to kill existing tumor cells. Each possibility represents a separate embodiment of the present invention
In other embodiments, the vaccines and immunogenic compositions utilized in any of the methods described above have any of the characteristics of vaccines and immunogenic compositions of the present invention. Each characteristic represents a separate embodiment of the present invention.
Various embodiments of dosage ranges are contemplated by this invention. In one embodiment, in the case of vaccine vectors, the dosage is in the range of 0.001 LDso/dose. In another embodiment, the dosage is 0.002 LDso/dose. In another embodiment the dosage is 0.003 LD50/dose In another embodiment the dosage is 0 004 LD5o/dose. In another embodiment the dosage is 0 006 LDso/dose In another embodiment the dosage is 0.008
LD50/dose. In another embodiment the dosage is 0 01 LDso/dose In another embodiment the dosage is 0 02 LDso/dose. In another embodiment the dosage is 0 03 LD50/dose. In one embodiment, the dosage is 0 04 LDso/dose In another embodiment, the dosage is 0.06 LDso/dose. In another embodiment, the dosage 0.08 LDso/dose. In another embodiment, the dosage is 0.1 LDso/dose. In another embodiment, the dosage is 0.15 LDso/dose. In another embodiment, the dosage is 0 2 LDso/dose. In another embodiment, the dosage is 0.25 LD5o/dose. In another embodiment, the dosage is 0 3 LDso/dose. In another embodiment, the dosage is 0.4 LDso/dose. In another embodiment, the dosage is 0 5 LDso/dose.
In another embodiment, the dosage is 107 bacteria/dose. In another embodiment, the dosage is 1.5 x 107 bacteria/dose In another embodiment, the dosage is 2 x 107 bacteria/dose. In another embodiment, the dosage is 3 x 107 bacteria/dose, hi another embodiment, the dosage is 4 x 107 bacteria/dose. In another embodiment, the dosage is 6 x 107 bacteria/dose In another embodiment, the dosage is 8 x 107 bacteria/dose. In another embodiment, the dosage is 1 x 108 bacteria/dose. In another embodiment, the dosage is 1 5 x 108 bacteria/dose. In another embodiment, the dosage is 2 x 108 bacteria/dose. In another embodiment, the dosage is 3 x 108 bacteria/dose. In another embodiment, the dosage is 4 x 10s bacteria/dose In another embodiment, the dosage is 6 x 10 bacteria/dose. In another embodiment, the dosage is 8 x 108 bacteria/dose In another embodiment, the dosage is 1 x 109 bacteria/dose. In another embodiment, the dosage is 1 5 x 10 bacteria/dose In another embodiment, the dosage is 2 x 109 bacteria/dose. In another embodiment, the dosage is 3 x 109 bacteria/dose
In another embodiment, the dosage is 5 x 109 bacteria/dose. In anothei embodiment, the dosage is 6 x 109 bacteria/dose. In another embodiment, the dosage is 8 x 109 bacteria/dose In another embodiment, the dosage is 1 x I O10 bacteria/dose. In another embodiment, the dosage is 1.5 x 1010 bacteria/dose. In another embodiment, the dosage is 2 x 1010 bacteria/dose. In another embodiment, the dosage is 3 x 1010 bacteria/dose. In another embodiment, the dosage is 5 x 1010 bacteria/dose. In another embodiment, the dosage is 6 x 1010 bacteria/dose. In another embodiment, the dosage is 8 x 1010 bacteria/dose. In another embodiment, the dosage is 8 x 109 bacteria/dose. In another embodiment, the dosage is 1 x 101 ' bacteria/dose. In another embodiment, the dosage is 1 5 x 10 " bacteria/dose. In another embodiment, the dosage is 2 x 101 ' bacteria/dose In another embodiment, the dosage is 3 x lθ" bacteria/dose. In another embodiment, the dosage is 5 x lθ" bacteria/dose. In another embodiment, the dosage is 6 x 101 ' bacteria/dose. In another embodiment, the dosage is 8 x lθ" bacteria/dose. Each possibility represents a separate embodiment of the present invention.
In another embodiment, in the case of recombinant polypeptides, the dosage is 1 mg/dose. In another embodiment, the dosage is 1.5 mg/dose. In another embodiment, the dosage is 2 mg/dose. In another embodiment, the dosage is 3 mg/dose. In another embodiment, the dosage is 4 mg/dose. In another embodiment, the dosage is 6 mg/dose. In another embodiment, the dosage is 8 mg/dose. In another embodiment, the dosage is 10 mg/dose. In another embodiment, the dosage is 15 mg/dose. In another embodiment, the dosage is 20 mg/dose. In another embodiment, the dosage is 30 mg/dose. In another embodiment, the dosage is 40 mg/dose In another embodiment, the dosage is 60 mg/dose In another embodiment, the dosage is 80 mg/dose. In another embodiment, the dosage is 100 mg/dose. In another embodiment, the dosage is 150 mg/dose. In another embodiment, the dosage is 200 mg/dose. In another embodiment, the dosage is 300 mg/dose In another embodiment, the dosage is 400 mg/dose In another embodiment, the dosage is 600 mg/dose. In another embodiment, the dosage is 800 mg/dose In another embodiment, the dosage is 1000 mg/dose
Each of the above doses represents a separate embodiment of the present invention.
In one embodiment, a vaccine or immunogenic composition of the present invention is administered alone to a subject In another embodiment, the vaccine or immunogenic composition is administered together with another cancer therapy Each possibility represents a separate embodiment of the present invention.
The recombinant Listeria of methods and compositions of the present invention is, in one embodiment, stably transformed with a construct encoding an antigen or an LLO-antigen fusion. In one embodiment, the construct contains a polylinker to facilitate further subcloning. Several techniques for producing recombinant Listeria are known.
In one embodiment, the construct or heterologous gene is integrated into the Listerial chromosome using homologous recombination Techniques for homologous recombination are well known in the art, and are described, for example, in Baloglu S, Boyle SM, et al, Immune responses of mice to vaccinia vims recombinants expressing either Listeria monocytogenes partial listeriolysin or Brucella abortus ribosomal L7/L12 protein. Vet
Microbiol 2005, 109(1-2): 1 1 -7; and Hang LL, Song HH, et al, Characterization of a mutant Listeria monocytogenes strain expressing green fluorescent protein. Acta Biochim Biophys Sin (Shanghai) 2005, 37(1 ): 19-24 In another embodiment, homologous recombination is performed as described in United States Patent No 6,855,320. In this case, a recombinant LM strain that expresses E7 was made by chromosomal integration of the E7 gene under the control of the hly promoter and with the inclusion of the hly signal sequence to ensure secretion of the gene product, yielding the recombinant referred to as Lm-AZ/E7. In another embodiment, a temperature sensitive plasmid is used to select the recombinants. Each technique represents a separate embodiment of the present invention.
In another embodiment, the construct or heterologous gene is integrated into the Listerial chromosome using transposon insertion. Techniques fortransposon insertion are well known in the art, and are described, inter alia, by Sun et al. (Infection and Immunity 1990, 58: 3770-3778) in the construction of DP-L967. Transposon mutagenesis has the advantage, in another embodiment, that a stable genomic insertion mutant can be formed but the disadvantage that the position in the genome where the foreign gene has been inserted is unknown.
In another embodiment, the construct or heterologous gene is integrated into the Listerial chromosome using phage integration sites (Lauer P, Chow MY et al, Construction, characterization, and use of two Listeria monocytogenes site-specific phage integration vectors. J Bacteriol 2002;184(15): 41 77-86). In certain embodiments of this method, an integrase gene and attachment site of a bacteriophage (e.g. U 153 or PSA lisleriophage) is used to insert the heterologous gene into the corresponding attachment site, which may be any appropriate site in the genome (e.g. comK or the 3' end of the arg tRNA gene). In another embodiment, endogenous prophages are cured from the attachment site utilized prior to integration of the construct or heterologous gene. In another embodiment, this method results in single-copy integrants. Each possibility represents a separate embodiment of the present invention.
In another embodiment, the construct is carried by the Listeria strain on a plasmid. Cloning of the gene into a prfA-containing vector and using this plasmid to complement a prfA(-) Listeria mutant has been used to construct DP-L2028. DP-L2028 is the influenza NP expressing strain used in the tumor protection experiments. An LM vector that expresses an E7 fusion protein has also been constructed via this technique Lm-GG/E7 was made by complementing a prfA-deletion mutant with a plasmid containing a copy of the prfA gene and a copy of the E7 gene fused to a form of the LLO (hly) gene truncated to eliminate the hemolytic activity of the enzyme, as described in United States Patent No. 6,565,852
Functional LLO is maintained by the organism via the endogenous chromosomal copy of hly.
In other embodiments, one of several approaches is taken to express the tumor antigen in Listeria In one embodiment, a fusion protein of the selected tumor antigen and a Listerial protein, such as PI-PLC, or a construct encoding same, is generated. In another embodiment, a signal sequence, of a secreted Listerial protein such as hemolysin or phospholipases, is fused to the antigen-encoding gene.
In another embodiment, the construct is contained in the Listeria strain in an episomal fashion- In another embodiment, the foreign antigen is expressed from a vector harbored by the recombinant Listeria strain. Each possibility represents a separate embodiment of the present invention.
In other embodiments, one of various promoters is used to express the antigen or fusion protein containing same. In one embodiment, an LM promoter is used, e.g. promoters for the genes hly, acta, pica, plcB and mpl, which encode the Listerial proteins hemolysin, act A, phosphotidylinositol-specific phospholipase, phospholipase C, and metalloprotease, respectively. Each possibility represents a separate embodiment of the present invention.
Panels of antigens are, in one embodiment, useful in immunotherapy against cancer to compensate for the fact that antigen-loss variants of the tumors can grow out under immune system pressure (Zhang et al, Clin Cancer Res 1998 4: 2669; Kawashima et al, Hum Immunol 1998 59: 1) Thus, in another embodiment, methods and compositions of the present invention comprise a cocktail of recombinant fusion proteins, each fusion protein comprising a different tumor associated antigen fused to a truncated LLO, or a cocktail of recombinant LM strains, each expressing a different tumor associated antigen. Each possibility represents a separate embodiment of the present invention
In another embodiment, methods and compositions of the present invention utilize a homologue of a Her-2 or LLO sequence of the present invention (e g SEQ ID No: 33, 34,
40, 41, 43, and 44). The terms "homology," "homologous," etc, when in reference to any protein or peptide, refer in one embodiment, to a percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology, and not considering any conservative substitutions as part of the sequence identity. Methods and computer programs for the alignment are well known in the art.
In another embodiment, the term "homology," when in reference to any nucleic acid sequence similarly indicates a percentage of nucleotides in a candidate sequence that are identical with the nucleotides of a corresponding native nucleic acid sequence.
Homology is, in one embodiment, determined by computer algorithm for sequence alignment, by methods well described in the art. For example, computer algorithm analysis of nucleic acid sequence homology may include the utilization of any number of software packages available, such as, for example, the BLAST, DOMAIN, BEAUTY (BLAST Enhanced Alignment Utility), GENPEPT and TREMBL packages.
In another embodiment, "homology'" refers to identity to a sequence selected from SEQ ID No: 33, 34, 40, 41 , 43, and 44 of greater than 70%, In anothei embodiment, "homology" refers to identity to a sequence selected from SEQ ID No: 33, 34, 40, 41 , 43, and 44 of greater than 72%. In another embodiment, the identity is greater than 75%. In another embodiment, the identity is greater than 78% In another embodiment, the identity is greater than 80% In another embodiment, the identity is greater than 82%. In another embodiment, the identity is greater than 83%. In another embodiment, the identity is greater than 85% In another embodiment, the identity is greater than 87%. In another embodiment, the identity is greater than 88% In another embodiment, the identity is greater than 90%. In another embodiment, the identity is greater than 92%. In another embodiment, the identity is greater than 93%. In another embodiment, the identity is greater than 95%. In another embodiment, the identity is greater than 96% In another embodiment, the identity is greater than 97%. In another embodiment, the identity is greater than 98%. In another embodiment, the identity is greater than 99%. In another embodiment, the identity is 100%. Each possibility represents a separate embodiment of the present invention.
In another embodiment, homology is determined is via determination of candidate sequence hybridization, methods of which are well described in the art (See, for example, "Nucleic Acid Hybridization" Hames, B. D., and Higgins S. T., Eds. (1985); Sambrook et al., 2001, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, N Y.; and Ausubel et al., 1989, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N. Y). For example methods of hybridization may be carried out under moderate to stringent conditions, to the complement of a DNA encoding a native caspase peptide. Hybridization conditions being, for example, overnight incubation at 42 0C in a solution comprising: 10-20 % formamide, 5 X SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7. 6), 5 X Denhardt's solution, 10 % dextran sulfate, and 20 μg/ml denatured, sheared salmon sperm DNA.
In one embodiment of the present invention, "nucleic acids" refers to a string of at least two base-sugar-phosphale combinations. The term includes, in one embodiment, DNA and RNA. "Nucleotides" refers, in one embodiment, to the monomeric units of nucleic acid polymers. RNA may be, in one embodiment, in the form of a tRNA (transfer RNA), snRNA (small nuclear RNA), rRNA (ribosomal RNA), mRNA (messenger RNA), anti-sense RNA, small inhibitory RNA (siRNA), micro RNA (miRNA) and ribozymes. The use of siRNA and miRNA has been described (Caudy AA el al, Genes & Devel 16: 2491 -96 and references cited therein). DNA may be in form of plasmid DNA, viral DNA, linear DNA, or chromosomal DNA or derivatives of these groups In addition, these forms of DNA and
RNA may be single, double, triple, or quadruple stranded The term also includes, in another embodiment, artificial nucleic acids that may contain other types of backbones but the same bases In one embodiment, the artificial nucleic acid is a PNA (peptide nucleic acid). PNA contain peptide backbones and nucleotide bases and are able to bind, in one embodiment, to both DNA and RNA molecules. In another embodiment, the nucleotide is oxetane modified.
In another embodiment, the nucleotide is modified by replacement of one or more phosphodiester bonds with a phosphorothioate bond. In another embodiment, the artificial nucleic acid contains any other variant of the phosphate backbone of native nucleic acids known in the art. The use of phosphothiorate nucleic acids and PNA are known to those skilled in the art, and are described in, for example, Neilsen PE, Curr Opin Struct Biol
9:353-57; and Raz NK et al Biochem Biophys Res Commun 297: 1075-84. The production and use of nucleic acids is known to those skilled in art and is described, for example, in Molecular Cloning, (2001), Sambrook and Russell, eds. and Methods in Enzymology: Methods for molecular cloning in eukaryotic cells (2003) Purchio and G C Fareed. Each nucleic acid derivative represents a separate embodiment of the present invention.
Protein and/or peptide homology for any amino acid sequence listed herein is determined, in one embodiment, by methods well described in the art, including immunoblot analysis, or via computer algorithm analysis of amino acid sequences, utilizing any of a number of software packages available, via established methods. Some of these packages may include the FASTA, BLAST, MPsrch or Scanps packages, and may employ the use of the Smith and Waterman algorithms, and/or global/local or BLOCKS alignments for analysis, for example. Each method of determining homology represents a separate embodiment of the present invention.
In another embodiment, the present invention provides a kit comprising a reagent utilized in performing a method of the present invention. In another embodiment, the present invention provides a kit comprising a composition, tool, or instrument of the present invention.
The terms "contacting" or "administering," in one embodiment, refer to directly contacting the cancer cell oi tumor with a composition of the present invention. In anothei embodiment, the terms refer to indirectly contacting the cancer cell or tumor with a composition of the present invention. In anothei embodiment, methods of the present invention include methods in which the subject is contacted with a composition of the present invention after which the composition is brought in contact with the cancer cell or tumor by diffusion or any othei active transport or passive transport piocess known in the art by which compounds circulate within the body. Each possibility represents a separate embodiment of the present invention.
Pharmaceutical Compositions
The pharmaceutical compositions containing vaccines and compositions of the present invention can be, in another embodiment, administered to a subject by any method known to a person skilled in the art, such as parenterally, paracancerally, transmucosally, transdermally, intramuscularly, intravenously, intra-dermally, subcutancously, intra- peritonealy, intra-ventricularly, intra-cranially, intra-vaginally or intra-tumorally.
In another embodiment of methods and compositions of the present invention, the vaccines or compositions are administered orally, and are thus formulated in a form suitable for oral administration, i.e as a solid or a liquid preparation. Suitable solid oral formulations include tablets, capsules, pills, granules, pellets and the like. Suitable liquid oral foπnulations include solutions, suspensions, dispersions, emulsions, oils and the like. In another embodiment of the present invention, the active ingredient is formulated in a capsule In accordance with this embodiment, the compositions of the present invention comprise, in addition to the active compound and the inert carrier or diluent, a hard gelating capsule.
In another embodiment, the vaccines or compositions are administered by intravenous, intra¬ arterial, or intra-muscular injection of a liquid preparation. Suitable liquid formulations include solutions, suspensions, dispersions, emulsions, oils and the like. In one embodiment, the pharmaceutical compositions are administered intravenously and are thus formulated in a form suitable for intravenous administration. In another embodiment, the pharmaceutical compositions are administered intra-arterially and are thus formulated in a form suitable for intra-arterial administration. In another embodiment, the pharmaceutical compositions are administered intra-muscularly and are thus formulated in a form suitable for intra-muscular administration.
In one embodiment, the term "treating" refers to curing a disease. In another embodiment, "treating" refers to preventing a disease. In another embodiment, "treating" refers to reducing the incidence of a disease. In another embodiment, "treating" refers to ameliorating symptoms of a disease. In another embodiment, "treating" refers to inducing remission In another embodiment, "treating" refers to slowing the progression of a disease. The terms "reducing", "suppressing" and "inhibiting" refer in another embodiment to lessening or decreasing Each possibility represents a separate embodiment of the present invention.
EXPERIMENTAL DETAILS SECTION
EXAMPLE 1
GENERATION OF L. MONOCYTOGENES STRAINS THAT SECRETE LLO FRAGMENTS FUSED TO Hcr-2 FRAGMENTS
MATERIALS AND EXPERIMENTAL METHODS
Subcloning pGG-55, the backbone of the Listeria Her-2 constructs used in the Examples, was created from pAM401. pAM401 , a shuttle vector able to replicate in both gram+ and gram" bacteria, contains a gram+ chloramphenicol resistance gene and a giam' tetracycline resistance gene ( Wirth, R et al, .1 Bacterid, 165: 831, 1986). To produce pGG-55, an hly- HPV 16 E7 fusion gene (including the hly promoter and the portion of hly encoding the first 441 amino acids of LLO; referred to below as "ΔLLO") and the pluripotent transcription factor, prfA (positive regulatory factor of listeriolysin expression) gene were cloned into pAM401 (Figure 1 ).
L monocytogenes (LM) strains Lm-ΔLLO-ECl , Lm-ΔLL0-EC2, Lm-ΔLLO-EC3, Lm- ΔLLO-IC1, and Lm-ΔLL0-IC2 each contain a plasmid expressing a fragment of rat Her-2 fused to the Listeria! hlγ gene. The following overlapping fragments of the extracellular and intracellular domains of Her-2 were cloned into the plasmid pGG-55: base pairs (bp) 74-994, (Lm-ΔLLO-ECl ; corresponding to AA 20-326 of Her-2), 923-1519 (Lm-ΔLLO-EC2; corresponding to AA 303-501), 1451 -1981 (Lm-ΔLLO-EC3; corresponding to AA 479-655), 2084-3259 (Lm-ΔLLO-ICl ; corresponding to AA 690-1081), and 3073-3796 (Lm-ΔLLO- IC2; corresponding to AA 1020-1255). The fragments are depicted in Figure 2A. The LDso of ECl, EC2, EC3, ICl , and IC2 were 1 x 108, 1 x 10°, 5 x lθ\ 1 x 108, and Ix 108, respectively.
Each Her-2 fragment was amplified by PCR from the pNINA plasmid, which contains the full-length rat Her-2 gene, using the following primers. Restriction sites (Xhol in the case of ECl, ICl, and IC2 5' primers; Spel for the 3' primers; and Sail for the EC2 and EC3 5' primers) are underlined, and the FLAG tag sequence in the EC 2 and EC3 the 3' primers are indicated by italics:
ECl: 5' primer:
CACGCGGATGAAATCGATAAGCTCGAGCCCCCCGGAATCGCGGGCAC (SEQ ID No: 1 ); 3' primer: CCGGACTAGTGACCTCTTGGTTATTCGGGGGACACACC (SEQ ID
No: 2).
EC2: 5" primer: CCGGGTCGACTGCCCCTACAACTACCTGTCTACG (SEQ IDNo: 3); 3' primer: CCGGACTAGT7TΛ CTTGTCA TCGTCGTCCTTGTA GTCCCCAC
TGTGGAGCAGGGCCTG (SEQ ID NO: 4);
EC3: 5' primer: CCGGGTCGACTGCTTTGTACACACTGTACCTTGG (SEQ ID No: 5); 3' primer: CCGGACTAGT7TΛ CTTGTCA TCGTCGTCCTTGTA GTCC
GGGCTGGCTCTCTGCTCTGC (SEQ ID NO: 6);
ICl : 5' primer: CCGGCTCGAGTATACGATGCGTAGGCTGCTGCAGG (SEQ ID No: 7); 31 primer: CCGGACTAGTAGCCAGTGGAGATCTGGGGGGCCC (SEQ ID No: 8); 1C2: 51 primer: CCGGCTCGAGGGTGACCTGGTAGACGCTGAAG (SEQ ID No: 9) and 3' primer: CCGGACTAGTTACAGGTACATCCAGGCCTAGG (SEQ ID No: 10)
Fragments were amplified by PCR and cloned into the pCR 2.1 expression system (Invitrogen, Carlsbad, CA), then excised with the delineated enzymes. The E7 gene was excised from the pGG-55 plasmid using XIw I and Spe I, then the Her-2 fragment was fusion was ligated into the E7 site (ends digested with Sal J are compatible with XJwI ends). XFL- 7, apr/A negative strain of LM, (Gunn GR et al, J Immunol 167: 647, 2001) was transfected with the plasmids by electroporation.
Bacteria Bacteria were grown in brain heart infusion medium (BD, Sparks, MD) with 50 μg/ml chloramphenicol and were frozen in 1 ml aliquots at -800C.
Western blots
ΔLLO-I-ler-2 expressing strains were grown overnight at 370C in Luria-Bertani (LB) medium with 50 microgram per milliliter (μg/ml) chloramphenicol. Supernatants were TCA precipitated and resuspended in IX LDS sample buffer (Invitrogen, San Diego, CA). 15 microliter (μl) of each sample was loaded on a 4-12% Bis-Tris SDS-PAGE gel (Invitrogen, San Diego, CA). Gels were transferred to a Immobilon-P polyvinyl idene fluoride membrane (Millipore, Billerica, MA) and blotted with a polyclonal rabbit serum recognizing iesidues 1 - 30 of LLO, followed by HRP-conjugated anti-rabbit antibody (Ameisham Pharmacia Biotech, UK).
Statistical analyses
Statistical analyses were performed using Student's t-test throughout the Examples
RESULTS
Five recombinant LM strains were constructed that express and secrete overlapping fragments of the rat Her-2 gene fused to the N-terminal portion of £ monocytogenes LLO protein (Figure 2A). The signal sequence and transmembrane domain of Her-2 were not included among the fragments due to their hydrophobicity and the inability of LM to secrete extremely hydrophobic domains. Secretion of each Her-2 fragment was confirmed by Western blot (Figure 2B). Molecular weights of the proteins Lm-ΔLLO-EC 1 Lm-ΔLLOΕC2 Lm-ΔLLO-EC3, Lm-ΔLLO-ICl, and Lm-ΔLLO-lC2 were 83, 70, 68, 92.5, and 74-kDa (lcilodalton), respectively. The strains were attenuated relative to the wild-type 10403S strain, exhibiting virulences comparable to Lm-ΔLL0-E7; namely 1x108, 5x10s, 1x109, 1x108, and 1x108 colony forming units (CFU), respectively.
EXAMPLE 2
VACCINATION WITH LLO-Her-2 CONSTRUCTS HALTS AND REVERSES GROWTH OF Her-2-EXPRESSING TUMORS
MATERIALS AND EXPERIMENTAL METHODS
Mice Six to eight week old female FVB/N mice were purchased from Charles River Laboratories
(Wilmington, MA).
Cell lines
The FVB/N syngeneic NT-2 tumor cell line, derived from a spontaneously occurring mammary tumor in an FVB/N Her-2 tiansgenic mouse (Reilly RT et al, Cane Res 60: 3569, 2000), constitutive! y expresses low levels of rat Her-2 and is tumorigenic in wild type syngeneic mice. NT-2 cells were grown in RPMI 1640 medium with 20% FCS, 10.2 mM HEPES, 2 milliniolar (mM) L-glutamine, 100 micromolar (μM) nonessential amino acids, 1 mM sodium pyruvate, 50 U (units)/ml penicillin G, 50 μg/ml streptomycin, 20 μg/ml insulin, and 2 μg/ml gentamycin at 37° C with 5% CO2.
Experimental setup
6-8 week-old FVB/N mice (n=8) were injected subcutaneously in the right flank with 2 x 106 NT-2 tumor cells in 200 μl PBS. Seven days post-tumor inoculation, palpable tumors of 4-5 mm were observed, after which mice were injected intraperitoneally with recombinant LM or PBS on days 7, 14, and 21 The shortest and longest surface diameters of the tumors were measured every 2 days with calipers. Mice were sacrificed if they reached a point at which mean tumor diameter reached 20 mm.
RESULTS
LM strains expressing ΔLLO-Her-2 fusions were compared to PBS and Lm-ΔLLO-B7 (negative controls) for theii ability to induce immunity against and reduction of tumors composed of the rat Her-2 expressing tumor line, NT-2. FVB/N mice were injected with NT- 2 tumors, then, on days 7, 14, and 21 following rumor inoculation, were administered 0 1 LD50 of recombinant LM or PBS. Injection of LM expiessing the ΔLLO-Her-2 fusions halted tumor growth aftei the first injection (Figures 2A and B); the cessation in tumor growth continued through the last timepoint, more than nine weeks after the last Her-2 vaccination. Moreover, a complete regression of tumors was subsequently observed in three out of 8 of the Lm-ΔLLO-EC2 and Lm-ΔLLO-EC3 mice and 1 of 8 of the Lm-ΔLLO-ECl and Lm- ΔLLO-IC1 mice. Additional mice from all five of the ΔLLO-Her-2 groups exhibited a reduction in tumor size. As expected, tumors grew continually in mice injected with PBS and Lm-ΔLLO-E7.
These findings demonstrate that fusions of a LLO fragment to Her-2 are capable of eliciting immunity against Her-2 -expressing tumors These findings further indicate that the elicited immunity (a) is strong enough to induces the complete regression of greater than 75% of established Her-2-expressing tumors; and (b) lasts over the course of over at least several months.
EXAMPLE 3
IMMUNE RESPONSES INDUCED BY THE Lm-LLO-Her-2 VACCINE INCLUDE
CD8+ T CELLS
MATERIALS AND EXPERIMENTAL METHODS
CD8* T cell depletion
CD8+ T cells were depleted by injection with 0 5 mg of the anti-CD8 antibody 2 43 (Saπniento M et al, J Immunol 125(6): 2665-72, 1980) on days 6, 7, 8, 1 1 , 14, 17, 20, and 23 post-tumor injection CDS+ T cell populations were reduced by greater than 95% as measured by flow cytometric analysis on day 24.
Flow cytometric analysis
Three color flow cytometry for CD8 (53-6.7, FITC conjugated), CD62 ligand (MeH 4, APC conjugated) (BD Biosciences Pharmingen, San Diego, CA), and Her-2 H-2q tetramer (PE conjugated) was performed using a FACSCalibur flow cytometer with CellQuest software (Becton Dickinson, San Jose, CA). Tetramers, provided by the NIAID Tetramer Core Facility of Emory University and the NIH AIDS Research and Reference Reagent Program, were loaded with an H-2q specific PDSLRDLSVF peptide. Splenocytes were stained at room temperature (rt) with the tetramer for one hour (hr) at 1 :200 dilution, then at 4° C with anti- CD8 and anti-CD62L antibodies for 30 minutes (min). The CD8+, CD62Llow subset was selected ("gated on"), and percentages of tetramer4 cells were compared using FlowJo software (Tree Star, Inc, Ashland, OR).
RESULTS
In order to determine the type of T cells that were mediating the observed anti-Her-2 immune responses, FVB/N mice with NT-2 tumors were depleted of CD8+ T cells, beginning 1 day prior to vaccination with Lm-ΔLLO-Her-2 vaccines, then vaccinated as described in Example 2. In the CD8+-injected mice, each of the Lm-ΔLLO-Her-2 vaccines lost effectiveness (Figures 3A and B); while in the non-depleted mice, tumor growth was controlled, as observed in Example 2.
Tetramer analysis was next used to confirm the above results. Non-tumor bearing 6-8 week- old FVB/N mice were immunized with either PBS or 0.1 LD50 Lm-ΔLLO-EC2 and boosted 21 days later. Splenocytes were harvested 5 days after the boost and were stained with an H- 2q tetramer specific for the epitope defined by amino acids 420-429 (PDSLRDLVF). A three-fold increase in tetiamer positive cells was observed in the Lm-ΔLLO-EC2- vaccinated mice (Figure 5),
These results show that CD8+ T cells are featured in the immunity elicited by the LLO-Her-2 fusion vaccines.
EXAMPLE 4
LLO-Her-2 FUSION VACCINES INDUCE IMMUNE RESPONSES TO SUB- DOMINANT CD8+ T CELL EPITOPES
MATERIALS AND EXPERIMENTAL METHODS
Cells NIH 3T3 cells, a mouse fibroblast line, were obtained from the American Type Culture Collection (ATCC). The NIH 3T3 and all the derived cells were cultured in DMEM supplemented with 10% FCS, 2 mM L-glutamine, 100 μM nonessential amino acids, 1 mM sodium pyruvate, 50 U/ml penicillin G, and 50 μg/ml streptomycin. Culture media for the 3T3-neu cell lines was supplemented with 1 mg/ml G418. Cells were grown at 37° C with
5% CO2.
T3-Her-2 tines
Briefly, wild type 3T3 cells were transduced with overlapping fragments of the rat Her-2 gene, creating nine 3T3 Her-2 fragment lines, and one 3T3 line expressing the full-length rat Her-2. Her-2 fragments were created using the following PCR primers:
Fragment 3 (bp 1-508): S'-CCGGGCCGAATTCGCAATGATC (SEQ ID NO: 1 1 ) and 3'- CCCCGAATTCCTACTGAGGGTTCCCACGGATCAA (SEQ ID NO: 12).
Fragment 2 (bp 458-886): 5'-
GACATGAAGTTGCGGCTCCCTAGTCTCACAGAGATCCTGAAG (SEQ ID NO: 13) and 3'-CCCCGAATTCCTACTCAGGGTTGTGCATGGACTC (SEQ ID NO: 14).
Fragment 3 (bp 836-1294): 5'-
GACATGAAGTTGCGGCTCCCTGCCCTCGTCACCTACAACACA (SEQ ID NO: 15) and 3'-CCCCGAATTCCTAGAGGTCACGGAGACTGTCTGG (SEQ ID NO: 16).
Fragment 4 (bp 1244-1675): 5'- GACATGAAGTTGCGGCTCCCTATCACAGGTTACCTGTACATC (SEQ E) No: 17) and
3'-CCCCGAATTCCTACTTCCATACTCGGCACTCCTC (SEQ ID NO: 18).
Fragment 5 (bp 1607-2077): 5'-
GACATGAAGTTGCGGCTCCCTACCCAGTGTGTCAACTGCAGT (SEQ ID No: 19) and 3'-CCCCGGTACCCTAGATCTTCTGTCTCCTTCGTTT (SEQ ID NO: 20)
Fragment 6 (bp 2009-2476): 5'-
GACATGAAGTTGCGGCTCCCTGGCGTCCTGCTGTTCCTGATC (SEQ ID No: 21 ) and 3'-CCCCGGTACCCTAACCTCGGTGTTCTCGGACATG (SEQ ID NO: 22).
Fragment 7 (bp 2405-2872), 5'- GACATGAAGTTGCGGCTCCCTTCCACAGTACAGCTGGTGACA(SEQIDNO:23) and3'-CCCCGGTACCCTAGCAGATTGGAGGCTGAGGTAG(SEQIDNO:24)
Fragment 8 '(bp 2801-3271), 5'- GACATGAAGTTGCGGCTCCCTGATGGAATCCCAGCCCGGGAG(SEQIDNO:25) and3'-CCCCGGTACCCTACCCTTCCGAGGGAGCCAGTGG(SEQIDNO: 26).
Fragment 9 (bp 3203-3796), 5'- GACATGAAGTTGCGGCTCCCTGAGCTGACACTGGGCCTGGAG(SEQIDNO:27) and3'-CCCCGGTACCCTATACAGGTACATCCAGGCCTAG(SEQIDNO:28).
Fragments 1 -9 span amino acids 1 -165, 148-291 , 274-426, 410-553, 531 -687, 655-820, 797- 952, 929-1085, 1063-1255 of Hei-2, respectively. Each fragment was ligated into the pcDNA3.1 mammalian transfection vector, which contains a cytomegalovirus (CMV) promoter (Invitrogen, Carlsbad, CA), at the multicloning site. Constructs were transfected into 3T3 cells using electroporation (20 μg/1 x 107 cells) or Lipofectamine (1 5 μg/3 x 105 cells; Life Technologies). Several clones of each fragment were isolated by limiting dilution. Expression of Her-2 fragments in the clones was determined by RT-PCR
Chromium release assay
FVB/N mice were immunized with 0 1 LD50 of each of the Lm-ΔLLO-Her-2 vaccines Splenocytes were harvested 9 days later and cultured for four days with irradiated (20,000 rads) NT-2 tumor cells at a 100:1 ratio of splenocytes to tumor cells with 20 U/ml IL-2 (Roche, Indianapolis, IN). Splenocytes were then used as effector cells in a standard 51Cr release assay. Target cells were labeled with chiomium-51 (51Cr) and cultured for four hours with splenocytes at effector :target ratios of 200: 1 , 100:1, 50: 1 , and 25: 1 in triplicate. Following the incubation, 100 μl of supernatant was assayed for 51Cr release. The percent specific lysis was determined as [(experimental counts pei minute - spontaneous counts per minute)/( total counts per minute - spontaneous counts per minute)] x 100. "Total counts per minute"' refers to the total number of counts in the target cell population, measured by lysing the cells after labeling and counting the label. In other words, this is the maximum amount of 51Cr that could be released
RESULTS CTL assays were perfonned in order to confirm that each of the Lm-ΔLLO-Her-2 vaccines elicit anti-Her-2 CD8"1 T cell immune responses. Mice were vaccinated with each of the ΔLLO-Hei-2 fusion vaccines, and splenocytes were isolated and tested for ability to induce lysis, as measured by 51Cr release assay, of 3T3 cells transduced with full length rat Her-2 (B), using wild-type 3T3 cells as a negative control (A). Each vaccine induced an anti-Her-2
CTL response, as evidenced by significant levels of lysis at E:T ratios of 200:1 or 100:1 Splenocytes from PBS-vaccinated mice, an additional negative control, induced only background levels of lysis (Figure 6). These findings provide further evidence that each of the ΔLLO-Her-2 fusion vaccines induces anti-Her-2 CTL immune responses.
To delineate the epitopes recognized by the CTL elicited by each vaccine, a panel of 3T3 cells expressing each of 9 Her-2 fragments were used as target cells in the above-described lysis assay, following vaccination with the coπesponding ΔLLO-Her-2 fusion vaccine. Each vaccine elicited a CTL response to at least one Her-2 fragment. For several of the Her-2 fragments, levels of lysis over background reached statistical significance (p < 0.05). Based on a combination of the negative and positive results, regions of Her-2 containing sub- dominant epitopes were delineated (Table 1 ). Thus, vaccination with the Lm-ΔLLO-Her-2 fragment vaccines revealed sub-dominant epitopes within Her-2 protein.
Table 1 Regions of Her-2/neu with potential H-2q epitopes, based on percent specific lysis in the CTL assay. Percent (specific lysis - background lysis by splenocytes from PBS- vaccinated mice) is depicted for the E:T ratios of 200:1 and 100:1 Assays were perfonned in triplicate; results of a representative experiment are shown. *Denotes statistically significant lysis above background (p < 0.05).
Figure imgf000046_0001
Figure imgf000047_0001
EXAMPLE 5
FUSION TO LLO AND DELIVERY BY LM ENHANCES THE IMMUNOGENICITY
OF Her-2 MATERIALS AND EXPERIMENTAL METHODS
DNA vaccines
DNA vaccines were constructed using pcDNA 3.1. Hei-2 and the ECl fragment were amplified by PCR using the following primers:
Full length, unfused Her-2: 5' CCGGGCTAGCATGGTCATCATGGAGCTGGCCGG (Nhe / site underlined; SEQ ID No: 29) and 3'
CCQQGMnJCTTACTTGTCATCGTCGTCCTTGTAGTCTCAT ACAGGTACAJCCA GGCC (EcoRV site underlined, FLAG lag in italics, stop codon in bold; SEQ ID No: 30). The above 5' primer was also used for amplifying unfused ECl, and the 3' primer for amplifying ΔLLO - full length Her-2.
ΔLLO-full length Her-2: 5' CCGGGTCGACATGGTCATCATGGAGCTGGCCGG (Sal I site underlined; SEQ ID No: 31). This primer was also used for amplifying ΔLLO-EC1.
Unfused ECl : 3' ccGGGAjAic TTA CTTGTCA TCGTCGTCCTTGTA GΓCTCAGACCTCTTGGTTAT
TCGGGGG {EcoR V site underlined, FLAG tag in italics, stop codon in bold; SEQ ID No: 32). This primer was also used for amplifying unfused ECl fused to ΔLLO.
Fragments were cloned into the multicloning site of pcDNA3.1, and used to transform Escherichia coli. Bacteria were grown in Luria-Bertani media (BD, Sparks, MD) with 50 micrograms per milliliter (μg/ml) ampicillin.
Tumor regression experiments
Tumor regression experiments were performed as described in Example 2, except that 7 x 105 NT-2 cells were utilized, and vaccinations were administered on days 3, 10, and 18. DNA vaccines (50 μg each of the pcDNA plasmid + and the GM-CSF plasmid or GM-CSF alone) were administered intra-muscularly and Lm administered intraperitoneally.
RESULTS
Several factors were present in the Lm-ΔLLO-Her-2 vaccines that may have contributed to the recognition of sub-dominant epitopes: (a) delivery by LM; (b) fusion of the target antigen to ΔLLO; (c) breaking Her-2 into fragments. To determine which one or more of these factors contributed to the recognition of sub-dominant epitopes, as evidence by enhanced anti-Hei-2 immune responses, mice were vaccinated with (a) pcDNA 3.1 -full length Her-2 (a DNA vaccine; "pcDNA neu"); (b) pcDNA 3.1 ΔLLO-full length Her-2 (pcDNA LLO-neu); (c) pcDNA 3.1-ECl (pcDNA ECl); (d) pcDNA 3 1-ΔLLO-EC1 (pcDNA LLO-ECl); or (e) Lm-ΔLLO-EC 1 , and a tumor regression expei iment was performed. GM-CSF was included with the DNA vaccines because of its ability to enhance the efficacy of DNA vaccines (McKay PF, Barouch DH et al, Eur J Immunol 2004 Apr;34(4): 101 1 -20.).
As depicted in Figure 7A, the best control of tumor growth was observed with Lm-ΔLLO- EC 1 ; 2/8 of the mice never developed palpable tumors; the tumor completely regressed in another two; and the other 4 mice exhibited slower (retarded) tumor growth than the mock- vaccinated controls. In the case of pcDNA ΔLLO-EC1 , one mouse never developed a tumor, and several of the other mice exhibited retarded tumor growth. Fusion to ΔLLO enhanced immunogenicity of ECl , as seen by comparison of pcDNA ECl vs. pcDNA ΔLLO-EC1 and pcDNA neu vs. pcDNA ΔLLO-neu (Figures 6B-C); vaccines in which the antigen was unfused exhibited tumors that grew at the same rate as mock-vaccinated controls An enhancing effect of dividing Her-2 into smaller fragments, in the case of the ΔLLO fusions, is seen from a comparison of pcDNA ΔLLO-neu vs pcDNA ΔLLO-EC1 , in which the latter group exhibited superior tumor control (Figure 7D). No effect was seen for the unfused antigens, as seen from a comparison of pcDNA neu vs. pcDNA ECl (Figure 7E).
EXAMPLE 6 LLO-Her-2 DNA VACCINES INDUCE IMMUNE RESPONSES TO SUB-DOMINANT
CD8* T CELL EPITOPES
Immune responses Io the DNA vaccines were further analyzed by measuring lysis of 3T3 cells transduced with Her-2 fragments, as described in Example 4 T cells from mice vaccinated with pcDNA 3.1 -ΔLLO-Her-2 or pcDN A 3.1 -ΔLLO-EC 1 lysed cells from more
3T3-Her-2 target cell groups than did the corresponding non-fused DNA vaccines (Table 2)
Table 2 Regions of Her-2/neu with potential epitopes based on DNA vaccinations of wt FVB mice. * denotes statistically significant lysis above background (p < 0.05) Percent specific lysis was calculated as % = 100 x ((experimental lysis - spontaneous lysis)/(total lysis - spontaneous lysis)).
Figure imgf000050_0001
Thus, fusion of an LLO fragment to full length Her-2 or a Her-2 fragment resulted in a broadening of the regions recognized by CTLs. Accordingly, based on the results of Examples 5-6, subdominant CD8+ antigen epitopes can be revealed by either (a) expression by LM; (b) fusion of the antigen to an LLO fragment; or (c) dividing the antigen into smaller fragments. EXAMPLE 7
VACCINATION WITH LLO-Her-2 INDUCES EPITOPE SPREADING MATERIALS AND EXPERIMENTAL METHODS
FVB/N are injected with NT-2 tumors, then vaccinated with each of the LM-ΔLLO-Hei-2 fragment strains, as described in Example 2, or with DNA ΔLLO-Her-2 fragment vaccines, as described in Example 5. Lymphocytes are isolated from the draining lymph nodes of the tumor site at various time points following vaccination. Epitopes recognized by the lymphocytes are determined by a lysis assay, using 3T3 cells expressing each of 9 Her -2 fragments, as described in Example 4
RESULTS
The induction of tumor regression at more than one month after the final boost is in contrast with observations using other types of vaccines, and suggests that the number of T cell subsets is being expanded by epitope spreading. For example, antigen presenting cells such as dendritic cells may be acquiring pieces of the dying cells, traveling to the draining lymph nodes, and presenting Her-2 epitopes not present in the vaccine fragment, resulting in a broadening of the and Her-2 CDS+ T cell response. To confirm this conclusion, mice are injected with NT-2 tumors, then vaccinated with each of the LM-ΔLLO-Her-2 fragment strains oi DNA ΔLLO-Her-2 fragment vaccines. Lymphocytes are isolated tumor draining lymph nodes at various time points, and the epitopes recognized by the lymphocytes are determined. Emergence is observed of reactivity to epitopes not present in the vaccine fragment. This broadening of the T cell response approximately correlates with temporally tumor regression.
This result demonstrates that vaccination with recombinant antigen-expressing LM induces epitope spreading In addition, vaccination with LLO-antigen fusions, even outside the context of LM, induces epitope spreading as well.
EXAMPLE 8
VACCINATION WITH LLO-Her-2 OVERCOMES IMMUNE TOLERANCE TO A SELF ANTIGEN MATERIALS AND EXPERIMENTAL METHODS
Rat Her-2/neu transgenic mice were provided by Dr. William Muller. Young, virgin HER-
2/neu transgenic mice that had not spontaneously developed tumors were injected with 5 x 104 NT-2 cells. Because the transgenic mouse is profoundly tolerant to HER-2/neu, the minimum dose required for tumoi growth in 100% of animals is much lower than wild-type mice (Reilly RT, Gottlieb MB el al, Cancer Res. 2000 JuI l ;60(13): 3569-76). NT-2 cells were injected into the subcutaneous space of the flank. Mice received 0.] LD5O of the
Listeria vaccine when 4-5 mm palpable tumors were detected (typically on day 7 after tumor implantation) and weekly thereafter, for an additional 4 weeks.
RESULTS
The rat Her-2/neu gene differs from the rat neu by 5-6% of amino acid residues, and thus is immunogenic in the mouse (Nagata Y, Furugen R et al, J Immunol. 159: 1336-43). A transgenic mouse that ovei expresses rat Her-2/neu under the transcriptional control of the Mouse Mammary Tumor Virus (MMTV) promoter and enhancer is immunologically tolerant to rat Her-2/neu. These mice spontaneously develop breast cancer The MMTV promoter also operates in hematopoietic cells, rendering the mice profoundly tolerant to HER-2/neu. This, this mouse is considered to be stringent model for human breast cancer and in general for tumors expressing antigens, such as Her-2/neu, that are expressed at low levels in normal tissue (Muller W I. (] 991 ) Expression of activated oncogenes in the murine mammary gland: transgenic models for human breast cancer. Cane Metastasis Rev 10: 217- 27)
6-8 week-old HER-2/neu transgenic mice were injected with NT-2 cells, then immunized with each of the LM-ΔLLO-Her-2 vaccines, or with PBS or ΔLLO-E7 (negative controls). While most control mice had to be sacrificed by day 42 because of their tumor burden, tumor growth was controlled in all of the vaccinated mice (Figure 8).
Thus, the ΔLM-LLO-Her-2 and Usleria-based Hei-2 vaccines are able to break tolerance to self antigen expressed on a tumor cell, as evidenced by their ability to induce the regression of established NT-2 tumors. EXAMPLE 9
LLO-HER-2 VACCINES CONTROL SPONTANEOUS TUMOR GROWTH IN
HER-2/NEU TRANSGENIC MICE
MATERIALS AND EXPERIMENTAL METHODS
ΔLM-LLO-Her-2 vaccines were administered in the following amounts: Lm-LLO-ECl : 1 x
10 Λ 7 cfu; Lm- Lm-LLO-EC2: 5 x 10 Λ 7 cfu; LLO-EC3: 1 x 10 Λ8 cfu; Lm-LLO-lC2: 1 x 10 Λ 7 cfu; Lm-LLO-ICl : 1 x 10 Λ 7.
RESULTS
The ΔLM-LLO-Her-2 vaccines were also evaluated for ability to prevent spontaneous tumor growth in the Her-2/neu transgenic mice. The transgenic mice (n=12 per vaccine group) were immunized 5 times with 0.1 LD50 of one of the vaccine strains, beginning at age 6 weeks and continuing once every three weeks. Mice were monitored for tumor formation in the mammary glands. By week 35, all of the control mice (PBS or Lm-LLO-NY-ESO-I- immunized) had developed tumors. By contrast, 92% of the Lm-LLO-ICl group were tumor free, as were 50% of the mice Lm-LLO-EC2, Lm-LLO-EC 1 , and Lm-LL0-IC2, and 25% of the mice immunized with Lm-LLO-EC3 (Figure 9).
This finding confirm the results of the previous Example, showing that ΔLM-LLO-Her-2 and Listeria-based Her-2 vaccines are able to break tolerance to self antigens.
EXAMPLE 10
GENERATION OF LLO-HER-2 VACCINES CONTAINING FRAGMENTS OF
HUMAN HER-2 PROTEIN
A similar strategy was used to express human Her-2/neu as was used for rat Her-2 (Example 1). The full-length HER-2 gene was split into five fragments, constituting overlapping fragments of the extracellular domain, (EC-I, EC-2 and EC -3) and the cytoplasmic domain (IC-I and IC-2) (Figure 10). Hydrophobic regions were not included in the constructs. These sequences differed from the rat sequences slightly due to the small disimilarities between the two sequences. The human fragments corresponding to the rat fragments were 22-326, 303- 501, 479-652, 677-1081 , and 1020-1255.
The human HER-2 sequences are isolated from a human breast cell cancer line e g. SK-BR3 (ATCC) by reverse transcription PCR (RT-PCR) using standard molecular biology methods For example, total cellular RNA is isolated using the RNeasy Minipreparation® kit (Qiagen) and a cDNA pool is generated (Titan-One-Tube PCR system®, Roche) using an oligo-dT primer The HER-2 sequences of interest are specifically amplified by the second step PCR using the following primers:
Figure imgf000054_0001
Restriction sites Xhol (5') and Spel (3') are added to allow for subsequent cloning into the Listeria vaccine vector, and an ochre stop codon is included in the reverse primers to terminate translation of the fusion protein. There are no Xhol or Spel sequences located in these fragments of the human HER-2 gene. The PCR products are purified (Qiaquick® PCR Purification Kit, Qiagen) and cloned into intermediate E coli vector pCR2.1 TOPO® (Invitrogen) After transformation of TOP 10 cells (Invitrogen), plasmid containing colonies are identified by PCR using primers Ml 3forward and M 13reverse (Invitrogen), One positive clone for each construct is giown up, plasmid DNA will be prepared (Qiafilter Midipreparation®, Qiagen) and the sequence of the HER-2/neu insert is verified by sequencing.
In order to introduce the HER-2/neu antigen sequences into the Listeria vector, pLLO-E7 is digested completely with Xhol and partially with Spel, thereby removing the E7 gene. The HER-2/neu sequences are digested with the same enzymes, all digests are separated by agarose gel electrophoresis, and the pLLO-E7 vector and the HER-2/neu insert sequences are purified (Qiaquick). The HER-2/neu antigen sequences are ligated into pLLO-E7, and the ligation mix is transformed into d-alanine racemase-deficient E. coli strain MB2159 by electroporalion. Colonies are tested by PCR for the presence of the HER-2/neu sequence and expanded in Luria Broth (LB) media, and plasmid DNA is prepared, then the sequence verified by restriction digestion with EcoRI, Smal, Ncol or with HindIII which yields a specific band pattern for each construct Plasmids are transformed into Listeria strain Lm(DA-) by electroporation, and individual clones are grown up in LB media containing 50 μg/ml streptomycin. The presence and sequence of the plasmid is again verified by restriction analysis. Expression and secretion of LLO-HER-2/neu fusion proteins is verified by Western blot of TCA-precipitated culture media, using a polyclonal PEST sequence- specific antibody.
EXAMPLE Il
TESTING OF LLO-HER-2 VACCINES CONTAINING FRAGMENTS OF HUMAN
HER-2 PROTEIN
The LLO-human Her-2 strains are tested for immunogenicity in mice and in human volunteers. Next, the vaccine strains are tested for their ability to protect mice against a challenge with tumor cells expressing human Her-2, as described in the above Examples.
Successful strains are administered to humans having Her-2-expressing tumors, and tested for their ability to induce tumor regression. In addition, the strains are tested for their ability to protect human subjects at risk for developing Her-2-expressing cancer, due to genetic or environmental factors. The vaccine strains are found to be immunogenic and to exhibit substantial anti-tumor activity.

Claims

WHAT IS CLAIMED IS:
1. A method of breaking an immune tolerance of a subject to an antigen-expressing tumor, comprising administering to said subject a recombinant polypeptide comprising an N-terminal fragment of a listeriolysin (LLO) protein fused to a fragment of said antigen or a recombinant nucleotide encoding said recombinant polypeptide, wherein said antigen has one or more dominant CDS+ T cell epitopes and wherein said fragment does not contain said any of said dominant CD8"1 T cell epitopes, whereby said subject mounts an immune response against said antigeπ- expressing tumor, thereby breaking an immune tolerance of a subject to an antigen- expressing tumor.
2 A method of breaking an immune tolerance of a subject to an antigen-expressing tumor, comprising administering to said subject a recombinant form of Listeria comprising a recombinant nucleotide, said recombinant nucleotide encoding a fragment of said antigen, wherein said antigen has one or more dominant CD8+ T cell epitopes and wherein said fragment does not contain said any of said dominant CDS+ T cell epitopes, whereby said subject mounts an immune response against said antigen-expressing tumor, thereby breaking an immune tolerance of a subject to an antigen-expressing tumor.
3. The method of either of claims 1 or 2, wherein said antigen is expressed at a detectable level on a non-tumor cell of said subject.
4. The method of either of claims 1 or 2, wherein at least part of said immune response is a CD8+ T cell response directed against an epitope of said antigen that is not any of said dominant CD8+ T cell epitopes.
5. A method of inducing a CD8+ T cell-mediated immune response in a subject against a subdominant CD8+ T cell epitope of an antigen, comprising administering to said subject a recombinant peptide comprising a fusion of an N-terminal fragment of a listeriolysin (LLO) protein to antigen or a fragment thereof or administering to said subject a nucleotide molecule encoding said recombinant peptide, thereby inducing a CD8+ T cell-mediated immune response against a subdominant CD8+ T cell epitope of an antigen.
6. A method of inducing a CD8+ T cell-mediated immune response in a subject against a subdominant CD8+ T cell epitope of an antigen, comprising administering to said subject a recombinant form of Listeria comprising a recombinant nucleotide, said iecombinant nucleotide encoding a said antigen or a fragment thereof, thereby inducing a CDS+ T cell-mediated immune response against a subdominant CDS+ T cell epitope of an antigen
J. The method of any of claims 1, 2, 3, 4, 5, or 6, wherein said fragment has a length of 150-420 amino acids.
8. The method of any of claims 1 , 2, 3, 4, 5, or 6, wherein said fragment has a length of one-sixth to one-half of said antigen.
9 The method of either of claims 1 , 3, 4, or 5, wherein the step of administering is performed with a recombinant form of Listeria comprising said recombinant nucleotide or expressing said recombinant polypeptide.
10. The method of any of claims 1, 2, 3, 4, 5, or 6, wherein said antigen is Her-2, bcr/abl, HPV E6, HPV E7, MZ2-E, MAGE-I and MUC-I, NY/ESO-1, Wilms tumor antigen, lelomerase, Proteinase 3, or Tyrosinase related protein 2.
1 1. A recombinant polypeptide compiising an N-terminal fragment of a listeriolysin (LLO) protein fused to a fragment of a Her-2 protein, said fragment of a Her-2 protein having a length of 150 to 420 amino acids.
12 The recombinant polypeptide of claim 1 1, wherein said Her-2 piotein is a human Her-2 protein.
13. The recombinant polypeptide of claim 1 1 , wherein said fragment of a Her-2 protein consists of amino acids 20-326, 303-501 , or 479-655,
14 The recombinant polypeptide of claim 1 1 , wherein said fragment of a Her-2 protein consists of amino acids 690-1081 or 1020-1255 of said Hei-2 protein.
15. The recombinant polypeptide of claim 1 1, wherein said fragment of a Her-2 protein consists of one-third to one-half of the extracellular domain of said Her-2 protein
16. The recombinant polypeptide of claim 1 1, wherein said fiagment of a Her -2 protein consists of one-third to one-half of the intracellular domain of said Her-2 protein.
17. The recombinant polypeptide of claim 1 1 , wherein said fragment of a Her-2 protein does not include a signal sequence of said Her-2 protein
18. The recombinant polypeptide of claim 1 1 , wherein said fiagment of a Her-2 protein does not include a transmembrane domain of Her-2 protein
19. A nucleotide molecule encoding the recombinant polypeptide of claim 1 1.
20. A recombinant form of Listeria comprising the nucleotide molecule of claim 19.
21. A recombinant form of Listeria comprising a nucleotide molecule encoding a fragment of a Her-2 protein, wherein said fragment of a Her-2 protein has a length of 150 to 420 amino acids.
22. The recombinant form of Listeria of claim 21, wherein said Her-2 protein is a human Her-2 protein.
23. A vaccine comprising the recombinant polypeptide of claim 11 , the nucleotide molecule of claim 19, the recombinant form of Listeria of claim 20, or the recombinant form of Listeria of claim 21
24. A method of inducing an anti-Hei-2 immune response in a subject, comprising administering to said subject the recombinant polypeptide of claim 11 or a recombinant nucleotide encoding said recombinant polypeptide, thereby inducing an anti-Her-2 immune response in a subject.
25. A method of impeding a growth of a Her-2-expressing tumor in a subject, comprising administering to said subject the recombinant polypeptide of claim 1 1 or a recombinant nucleotide encoding said recombinant polypeptide, whereby said subject mounts an immune response against said Her-2-expressing tumor, thereby impeding a growth of a Hei-2-expressing tumor in a subject.
26. A method of shrinking a Her-2-expressing tumor in a subject, comprising administering to said subject the recombinant polypeptide of claim 22 or a recombinant nucleotide encoding said recombinant polypeptide, whereby said subject mounts an immune response against said Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
27 The method of any of claims 24, 25, or 26, whereby said step of administering is performed with a recombinant form of Listeria comprising said recombinant nucleotide or expressing said recombinant polypeptide.
28. A recombinant form of Listeria comprising a recombinant nucleotide, said recombinant nucleotide encoding a fragment of a Her-2 protein, wherein said fragment of a Her-2 protein has a length of 150 to 420 amino acids.
29. The recombinant form of Listeria of claim 31, wherein said fragment of a Heτ-2 protein has a length of 150 to 420 amino acids.
30 The recombinant form of Listeria of claim 31, wherein said Her-2 protein is a human Her-2 protein
31. The recombinant form of Listeria of claim 31 , wherein said fragment of a Her-2 protein consists of one-third to one-half of the extracellular domain of said Her-2 protein.
32. The recombinant form of Listeria of claim 54, wherein said fragment of a Her-2 protein consists of one-third to one-half of the intracellular domain of said Her-2 protein,
33 A method of impeding a growth of a Her-2-expressing tumor in a subject, comprising administering to said subject the recombinant form of Listeria of claim 31 , whereby said subject mounts an immune response against said Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
34. A method of shrinking a Her-2-expressing tumor in a subject, comprising administering to said subject the recombinant form of Listeria of claim 54, whereby said subject mounts an immune response against said Her-2-expressing tumor, thereby impeding a growth of a Her-2-expressing tumor in a subject.
35. The method of any of claims 24, 25, 26, 33, or 34, wherein said immune response comprises a CDS+ T cell-mediated response.
36. The method of any of claims 24, 25, 26, 33, or 34, whereby said immune response against said Hεr-2-expressing tumor comprises an immune response to a subdominant epitope of said Her-2 protein.
37. A recombinant polypeptide comprising a fragment of a Her-2 protein, wherein said fragment consists of amino acids 20-326, 303-501, or 479-655 of said Her-2 protein.
38. A recombinant L monocytogenes strain expressing the recombinant polypeptide of claim 37
39. A recombinant polypeptide comprising a fragment of a Her-2 protein, wherein said fragment consists of amino acids 690-1081 or 1020-1255 of said Her-2 protein.
40. A recombinant L monocytogenes strain expressing the recombinant polypeptide of claim 39.
PCT/US2005/032682 2004-09-24 2005-09-14 Listeria-based and llo-based vaccines WO2006036550A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
DK05811815.9T DK1804831T3 (en) 2004-09-24 2005-09-14 Listeria LLO-BASED AND VACCINES BASED
EP05811815.9A EP1804831B1 (en) 2004-09-24 2005-09-14 Listeria-based and llo-based vaccines
CA002581331A CA2581331A1 (en) 2004-09-24 2005-09-14 Listeria-based and llo-based vaccines
JP2007533537A JP2008514199A (en) 2004-09-24 2005-09-14 Listeria-based vaccines and LLO-based vaccines
EP15182979.3A EP2980101B1 (en) 2004-09-24 2005-09-14 Vaccine comprising an n-terminal, pest sequence comprising fragment of listeriolysin (llo) protein fused to a her-2 antigen.
AU2005289957A AU2005289957A1 (en) 2004-09-24 2005-09-14 Listeria-based and LLO-based vaccines
ES05811815.9T ES2566392T3 (en) 2004-09-24 2005-09-14 Listeria-based and LLO-based vaccines

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US10/949,667 US7794729B2 (en) 1994-11-08 2004-09-24 Methods and compositions for immunotherapy of cancer
US10/949,667 2004-09-24
US11/223,945 US7820180B2 (en) 2004-09-24 2005-09-13 Listeria-based and LLO-based vaccines
US11/223,945 2005-09-13

Publications (2)

Publication Number Publication Date
WO2006036550A2 true WO2006036550A2 (en) 2006-04-06
WO2006036550A3 WO2006036550A3 (en) 2006-08-10

Family

ID=36119378

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/032682 WO2006036550A2 (en) 2004-09-24 2005-09-14 Listeria-based and llo-based vaccines

Country Status (9)

Country Link
US (1) US7820180B2 (en)
EP (2) EP1804831B1 (en)
JP (1) JP2008514199A (en)
AU (1) AU2005289957A1 (en)
CA (1) CA2581331A1 (en)
DK (1) DK1804831T3 (en)
ES (1) ES2566392T3 (en)
HK (1) HK1221232A1 (en)
WO (1) WO2006036550A2 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1939217A3 (en) * 2004-10-15 2008-09-03 Augusto Amici Plasmids coding for p185neu protein sequence variants and therapeutic uses thereof
EP2056849A2 (en) * 2006-08-04 2009-05-13 The Trustees of the University of Pennsylvania Methods and compositions for treating ige-mediated diseases
EP2061800A2 (en) * 2006-08-15 2009-05-27 The Trustees of the University of Pennsylvania Compositions comprising hmw-maa and fragments thereof, and methods of use thereof
US7588930B2 (en) 2000-03-29 2009-09-15 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
EP2128173A1 (en) 2005-05-31 2009-12-02 Ralf Jochem Therapeutic compound for preventing and fighting bone metastases
EP2155243A2 (en) * 2007-05-10 2010-02-24 The Trustees of the University of Pennsylvania Compositions and methods comprising klk3, psca, or folh1 antigen
US7700344B2 (en) 2001-03-26 2010-04-20 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
JP2010532313A (en) * 2007-03-08 2010-10-07 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Compositions and methods for the treatment of cervical cancer
US7935804B2 (en) 2006-03-01 2011-05-03 Aduro Biotech Engineered Listeria and methods of use thereof
WO2012054294A1 (en) * 2010-10-19 2012-04-26 Merial Limited Her2 dna vaccine as adjunct treatment for cancers in companion animals
US8268326B2 (en) 2006-08-15 2012-09-18 The Trustees Of The University Of Pennsylvania Compositions comprising HMW-MAA and fragments thereof, and methods of use thereof
US8771702B2 (en) 2001-03-26 2014-07-08 The Trustees Of The University Of Pennsylvania Non-hemolytic LLO fusion proteins and methods of utilizing same
US8778329B2 (en) 2009-03-04 2014-07-15 The Trustees Of The University Of Pennsylvania Compositions comprising angiogenic factors and methods of use thereof
US8791237B2 (en) 1994-11-08 2014-07-29 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of non-hodgkins lymphoma
WO2014139672A1 (en) * 2013-03-11 2014-09-18 Icon Genetics Gmbh Her2/neu cancer vaccine
US8956621B2 (en) 1994-11-08 2015-02-17 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cervical dysplasia
EP2980101A1 (en) 2004-09-24 2016-02-03 The Trustees of The University of Pennsylvania Vaccine comprising an n-terminal, pest sequence comprising fragment of listeriolysin (llo) protein fused to a her-2 antigen
US9644212B2 (en) 2008-05-19 2017-05-09 Advaxis, Inc. Dual delivery system for heterologous antigens
US9650639B2 (en) 2008-05-19 2017-05-16 Advaxis, Inc. Dual delivery system for heterologous antigens
US9943590B2 (en) 2010-10-01 2018-04-17 The Trustees Of The University Of Pennsylvania Use of Listeria vaccine vectors to reverse vaccine unresponsiveness in parasitically infected individuals
US10016617B2 (en) 2009-11-11 2018-07-10 The Trustees Of The University Of Pennsylvania Combination immuno therapy and radiotherapy for the treatment of Her-2-positive cancers
US10058599B2 (en) 2012-03-12 2018-08-28 Advaxis, Inc. Suppressor cell function inhibition following Listeria vaccine treatment
US10064898B2 (en) 2011-03-11 2018-09-04 Advaxis, Inc. Listeria-based adjuvants
US10143734B2 (en) 2014-02-18 2018-12-04 Advaxis, Inc. Biomarker directed multi-target immunotherapy
US10258679B2 (en) 2014-04-24 2019-04-16 Advaxis, Inc. Recombinant Listeria vaccine strains and methods of producing the same
US10900044B2 (en) 2015-03-03 2021-01-26 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11179339B2 (en) 2017-09-19 2021-11-23 Advaxis, Inc. Compositions and methods for lyophilization of bacteria or listeria strains
US11897927B2 (en) 2016-11-30 2024-02-13 Advaxis, Inc. Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7662396B2 (en) * 2001-03-26 2010-02-16 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
US6051237A (en) * 1994-11-08 2000-04-18 The Trustees Of The University Of Pennsylvania Specific immunotherapy of cancer using a live recombinant bacterial vaccine vector
US20070264279A1 (en) * 1994-11-08 2007-11-15 Claudia Gravekamp Compositions and methods comprising a MAGE-b antigen
US7794729B2 (en) * 1994-11-08 2010-09-14 The Trustees Of The University Of Pennsylvania Methods and compositions for immunotherapy of cancer
WO2003102168A1 (en) * 2002-05-29 2003-12-11 The Regents Of The University Of California Attenuated listeria spp. and methods for using the same
US20070207171A1 (en) * 2006-03-01 2007-09-06 Cerus Corporation Engineered listeria and methods of use thereof
US8277797B2 (en) 2006-11-22 2012-10-02 The Regents Of The University Of California Interferon-β production modulating Listeria strains and methods for using same
US20110111481A1 (en) 2007-06-29 2011-05-12 Chiang Li ENABLING THE USE OF LONG dsRNA FOR GENE TARGETING IN MAMMALIAN AND OTHER SELECTED ANIMAL CELLS
US9017660B2 (en) 2009-11-11 2015-04-28 Advaxis, Inc. Compositions and methods for prevention of escape mutation in the treatment of Her2/neu over-expressing tumors
US9084747B2 (en) * 2009-11-11 2015-07-21 Advaxis, Inc. Compositions and methods for prevention of escape mutation in the treatment of HER2/NEU over-expressing tumors
US10702614B2 (en) 2011-02-15 2020-07-07 Albert Einstein College Of Medicine Radiobacteria for therapy of cancer
EP3288572A4 (en) * 2015-04-28 2018-12-05 Albert Einstein College of Medicine, Inc. Treatment of cancer using recall antigens delivered by attenuated bacteria
WO2017048714A1 (en) * 2015-09-15 2017-03-23 Advaxis, Inc. Manufacturing method of an immunotherapeutic formulation comprising a recombinant listeria strain
JP2019522991A (en) * 2016-07-05 2019-08-22 アドバクシス, インコーポレイテッド Listeria-based immunogenic compositions containing Wilms tumor protein antigen and methods of use thereof
CN111349645B (en) * 2018-12-24 2022-05-17 上海若泰医药科技有限公司 Method for improving safety of non-integrated attenuated listeria vaccine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6565852B1 (en) 1994-11-08 2003-05-20 The Trustees Of The University Of Pennsylvania Methods and compositions for immunotherapy of cancer
US6855320B2 (en) 2000-03-29 2005-02-15 The Trustees Of The University Of Pennsylvania Fusion of non-hemolytic, truncated form of listeriolysin O to antigens to enhance immunogenicity

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816253A (en) 1977-12-08 1989-03-28 Likhite Vilas V Novel mutant strain of Listeria monocytogenes and its use in production of IgM antibodies and as an immunotherapeutic agent
CA1156953A (en) 1979-06-08 1983-11-15 Michael A. Kessick Lime addition to heavy crude oils prior to coking
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
FR2508455A1 (en) * 1981-06-30 1982-12-31 Sanofi Sa PROCESS FOR THE PREPARATION OF (THIENYL-2) - AND (THIENYL-3) -2-ETHYLAMINE DERIVATIVES
US5262177A (en) 1986-02-07 1993-11-16 Oncogen Recombinant viruses encoding the human melanoma-associated antigen
US4777239A (en) 1986-07-10 1988-10-11 The Board Of Trustees Of The Leland Stanford Junior University Diagnostic peptides of human papilloma virus
US5171665A (en) 1989-04-17 1992-12-15 Oncogen Monoclonal antibody to novel antigen associated with human tumors
EP0474727B1 (en) * 1989-05-19 1997-07-23 Genentech, Inc. Her2 extracellular domain
US5830702A (en) 1990-10-31 1998-11-03 The Trustees Of The University Of Pennsylvania Live, recombinant listeria monocytogenes and production of cytotoxic T-cell response
US5342774A (en) 1991-05-23 1994-08-30 Ludwig Institute For Cancer Research Nucleotide sequence encoding the tumor rejection antigen precursor, MAGE-1
FR2686896B1 (en) 1992-01-31 1995-01-06 Pasteur Institut MUTANT ATTENUE OF LISTERIA MONOCYTOGENES; RECOMBINANT STRAIN OF LISTERIA MONOCYTOGENES, USE AS HETEROLOGOUS VECTORS OF VACCINE ANTIGENS AND USE AS VACCINE OR DIAGNOSTIC COMPOSITION.
US5633234A (en) 1993-01-22 1997-05-27 The Johns Hopkins University Lysosomal targeting of immunogens
US7794729B2 (en) 1994-11-08 2010-09-14 The Trustees Of The University Of Pennsylvania Methods and compositions for immunotherapy of cancer
US7662396B2 (en) 2001-03-26 2010-02-16 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
US7820180B2 (en) 2004-09-24 2010-10-26 The Trustees Of The University Of Pennsylvania Listeria-based and LLO-based vaccines
US5681570A (en) 1995-01-12 1997-10-28 Connaught Laboratories Limited Immunogenic conjugate molecules
US6017705A (en) 1995-03-14 2000-01-25 Ludwig Institute For Cancer Research Isolated nucleic acid molecules which are members of the MAGE-B family and uses thereof
US5877159A (en) 1995-05-03 1999-03-02 University Of Maryland At Baltimore Method for introducing and expressing genes in animal cells and live invasive bacterial vectors for use in the same
US5824538A (en) 1995-09-06 1998-10-20 The United States Of America As Represented By The Secretary Of The Army Shigella vector for delivering DNA to a mammalian cell
DE19541450C2 (en) 1995-11-07 1997-10-02 Gsf Forschungszentrum Umwelt Gene construct and its use
US6479258B1 (en) 1995-12-07 2002-11-12 Diversa Corporation Non-stochastic generation of genetic vaccines
US5858682A (en) 1996-08-02 1999-01-12 Pharmingen E2A/pbx1 fusion protein specific monoclonal antibodies
JP2002513287A (en) 1997-04-18 2002-05-08 ゲゼルシャフト フュア バイオテクノロギッシェ フォーシュンク エム ベー ハー(ゲー ベー エフ) Salmonella strain
US5891432A (en) 1997-07-29 1999-04-06 The Immune Response Corporation Membrane-bound cytokine compositions comprising GM=CSF and methods of modulating an immune response using same
US6833135B1 (en) 1997-08-06 2004-12-21 Laboratorio Medinfar Produtos Farmaceuticos, Lda. DNA integration into “Mycobacterium spp.” genome by trans-complementation using a site-specific integration system
EP0902086A1 (en) 1997-08-22 1999-03-17 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Tuberculosis vaccine
US6099848A (en) 1997-11-18 2000-08-08 The Trustees Of The University Of Pennsylvania Immunogenic compositions comprising DAL/DAT double-mutant, auxotrophic, attenuated strains of Listeria and their methods of use
US6306404B1 (en) 1998-07-14 2001-10-23 American Cyanamid Company Adjuvant and vaccine compositions containing monophosphoryl lipid A
US6818002B2 (en) 1999-02-02 2004-11-16 Samuel Shiber Vessel cleaner and barrier
WO2000049158A2 (en) 1999-02-18 2000-08-24 Compugen Ltd. Psa and klk-2 splicing variants
DE19949594A1 (en) 1999-10-14 2001-04-26 Deutsches Krebsforsch Recombinant attenuated listeria for immunotherapy
CA2404164A1 (en) 2000-03-29 2001-10-04 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing immunogenicity of antigens
JP5053499B2 (en) 2000-06-30 2012-10-17 地方独立行政法人東京都健康長寿医療センター Preventive and therapeutic agents for demyelinating diseases
US7700344B2 (en) 2001-03-26 2010-04-20 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
WO2003008537A2 (en) 2001-04-06 2003-01-30 Mannkind Corporation Epitope sequences
WO2003065787A2 (en) 2002-02-06 2003-08-14 Johns Hopkins University School Of Medicine Methods and compositions for the targeting of a systemic immune response to specific organs or tissues
US7425449B2 (en) 2002-04-30 2008-09-16 The Regents Of The University Of California Site specific Listeria integration vectors and methods for using the same
AU2003259109A1 (en) 2002-07-12 2004-02-02 The Johns Hopkins University Mesothelin vaccines and model systems
JP2006518191A (en) 2003-01-09 2006-08-10 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルベニア Compositions, methods, and kits for enhancing the immunogenicity of bacterial vaccine vectors
AU2004246999B2 (en) 2003-02-06 2010-06-10 Aduro Biotech, Inc., Listeria attenuated for entry into non-phagocytic cells, vaccines comprising the listeria, and methods of use thereof
CA2577306A1 (en) 2004-08-13 2006-02-16 The Trustees Of The University Of Pennsylvania Methods for constructing antibiotic resistance free vaccines
JP2008516614A (en) 2004-10-18 2008-05-22 メディミューン,インコーポレーテッド High cell density method for Listeria growth
JP2008523098A (en) * 2004-12-10 2008-07-03 エモリー・ユニバーシテイ 2 'and 3'-substituted cyclobutyl nucleoside analogs for the treatment of viral infection and abnormal cell proliferation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6565852B1 (en) 1994-11-08 2003-05-20 The Trustees Of The University Of Pennsylvania Methods and compositions for immunotherapy of cancer
US6855320B2 (en) 2000-03-29 2005-02-15 The Trustees Of The University Of Pennsylvania Fusion of non-hemolytic, truncated form of listeriolysin O to antigens to enhance immunogenicity

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Molecular Cloning", 2001
"Nucleic Acid Hybridization", 1985
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1989, GREEN PUBLISHING ASSOCIATES AND WILEY INTERSCIENCE
BALOGLU S; BOYLE SM ET AL.: "Immune responses of mice to vaccinia virus recombinants expressing either Listeria monocytogenes partial listeriolysin or Brucella abortus ribosomal L7/L12 protein", VET MICROBIOL, vol. 109, no. 1-2, 2005, pages 11 - 7, XP004970581, DOI: doi:10.1016/j.vetmic.2005.04.011
BARGMANN CI ET AL., NATURE, vol. 319, 1986, pages 226
CAUDY AA ET AL., GENES & DEVEL, vol. 16, pages 2491 - 96
ERCOLINI ET AL., J IMMUNOL, vol. 170, 2003, pages 4273 - 4280
JIANG LL; SONGHH ET AL.: "Characterization of a mutant Listeria monocytogenes strain expressing green fluorescent protein", ACTA BIOCHIM BIOPHYS SIN (SHANGHAI, vol. 37, no. 1, 2005, pages 19 - 24, XP055175888, DOI: doi:10.1093/abbs/37.1.19
KAWASHIMA ET AL., HUM IMMUNOL, vol. 59, 1998, pages 1
KING CR ET AL., SCIENCE, vol. 229, 1985, pages 974
LAUER P; CHOW MY ET AL.: "Construction, characterization, and use of two Listeria monocytogenes site-specific phage integration vectors", J BACTERIOL, vol. 184, no. 15, 2002, pages 4177 - 86, XP002971584, DOI: doi:10.1128/JB.184.15.4177-4186.2002
NEILSEN PE, CURR OPIN STRUCT BIOL, vol. 9, pages 353 - 57
PURCHIO; G. C. FAREED: "Methods in Enzymology: Methods for molecular cloning in eukaryotic cells", 2003
RAZ NK ET AL., BIOCHEM BIOPHYS RES COMMUN., vol. 297, no. 1, pages 075 - 84
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 2001, COLD SPRING HARBOR PRESS
See also references of EP1804831A4
SUN ET AL., INFECTION AND IMMUNITY, vol. 58, 1990, pages 3770 - 3778
WIRTH, R ET AL., J BACTERIOL, vol. 165, 1986, pages 831
ZHANG ET AL., CLIN CANCER RES, vol. 4, 1998, pages 2669

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8791237B2 (en) 1994-11-08 2014-07-29 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of non-hodgkins lymphoma
US8956621B2 (en) 1994-11-08 2015-02-17 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cervical dysplasia
US8114414B2 (en) 1994-11-08 2012-02-14 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cervical cancer
US9549973B2 (en) 2000-03-27 2017-01-24 The Trustees Of The University Of Pennsylvania Compositions and methods comprising KLK3 or FOLH1 antigen
US9012141B2 (en) 2000-03-27 2015-04-21 Advaxis, Inc. Compositions and methods comprising KLK3 of FOLH1 antigen
US7588930B2 (en) 2000-03-29 2009-09-15 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
US7655238B2 (en) 2000-03-29 2010-02-02 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
US7635479B2 (en) 2000-03-29 2009-12-22 The Trustees Of The University Of Pennsylvania Composition and methods for enhancing immunogenecity of antigens
US7700344B2 (en) 2001-03-26 2010-04-20 The Trustees Of The University Of Pennsylvania Compositions and methods for enhancing the immunogenicity of antigens
US9499602B2 (en) 2001-03-26 2016-11-22 Advaxis, Inc. Non-hemolytic LLO fusion proteins and methods of utilizing same
US8771702B2 (en) 2001-03-26 2014-07-08 The Trustees Of The University Of Pennsylvania Non-hemolytic LLO fusion proteins and methods of utilizing same
EP2980101A1 (en) 2004-09-24 2016-02-03 The Trustees of The University of Pennsylvania Vaccine comprising an n-terminal, pest sequence comprising fragment of listeriolysin (llo) protein fused to a her-2 antigen
EP1939217A3 (en) * 2004-10-15 2008-09-03 Augusto Amici Plasmids coding for p185neu protein sequence variants and therapeutic uses thereof
EP2128173A1 (en) 2005-05-31 2009-12-02 Ralf Jochem Therapeutic compound for preventing and fighting bone metastases
US8580939B2 (en) 2006-03-01 2013-11-12 Aduro Biotech Engineered Listeria and methods of use thereof
US7935804B2 (en) 2006-03-01 2011-05-03 Aduro Biotech Engineered Listeria and methods of use thereof
US10166276B2 (en) 2006-05-02 2019-01-01 The Trustees Of The University Of Philadelphia Compositions and methods for treatment of non-hodgkins lymphoma
EP2056849A4 (en) * 2006-08-04 2010-09-08 Univ Pennsylvania Methods and compositions for treating ige-mediated diseases
EP2056849A2 (en) * 2006-08-04 2009-05-13 The Trustees of the University of Pennsylvania Methods and compositions for treating ige-mediated diseases
EP3284478A1 (en) * 2006-08-15 2018-02-21 The Trustees of the University of Pennsylvania Compositions comprising hmw-maa and fragments thereof for treating cancer
US8241636B2 (en) 2006-08-15 2012-08-14 The Trustees Of The University Of Pennsylvania Compositions comprising HMW-MAA and fragments thereof, and methods of use thereof
US8268326B2 (en) 2006-08-15 2012-09-18 The Trustees Of The University Of Pennsylvania Compositions comprising HMW-MAA and fragments thereof, and methods of use thereof
EP2061800A4 (en) * 2006-08-15 2011-05-04 Univ Pennsylvania Compositions comprising hmw-maa and fragments thereof, and methods of use thereof
EP2977456A1 (en) * 2006-08-15 2016-01-27 The Trustees of The University of Pennsylvania Compositions comprising hmw-maa and fragments thereof for treating cancer
EP2061800A2 (en) * 2006-08-15 2009-05-27 The Trustees of the University of Pennsylvania Compositions comprising hmw-maa and fragments thereof, and methods of use thereof
JP2010532313A (en) * 2007-03-08 2010-10-07 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Compositions and methods for the treatment of cervical cancer
EP2859899A1 (en) * 2007-05-10 2015-04-15 The Trustees of the University of Pennsylvania Compositions comprising KLK3
EP2155243A4 (en) * 2007-05-10 2011-07-27 Univ Pennsylvania Compositions and methods comprising klk3, psca, or folh1 antigen
US11446369B2 (en) 2007-05-10 2022-09-20 Advaxis, Inc. Compositions and methods comprising KLK3 or FOLH1 antigen
EP2155243A2 (en) * 2007-05-10 2010-02-24 The Trustees of the University of Pennsylvania Compositions and methods comprising klk3, psca, or folh1 antigen
US9644212B2 (en) 2008-05-19 2017-05-09 Advaxis, Inc. Dual delivery system for heterologous antigens
US9650639B2 (en) 2008-05-19 2017-05-16 Advaxis, Inc. Dual delivery system for heterologous antigens
US10189885B2 (en) 2008-06-23 2019-01-29 The Trustees Of The University Of Pennsylvania Non-hemolytic LLO fusion proteins and methods of utilizing same
US9408898B2 (en) 2009-03-04 2016-08-09 The Trustees Of The University Of Pennsylvania Compositions comprising angiogenic factors and methods of use thereof
US9981024B2 (en) 2009-03-04 2018-05-29 The Trustees Of The University Of Pennsylvania Compositions comprising angiogenic factors and methods of use thereof
US10695410B2 (en) 2009-03-04 2020-06-30 The Trustees Of The University Of Pennsylvania Compositions comprising angiogenic factors and methods of use thereof
US8778329B2 (en) 2009-03-04 2014-07-15 The Trustees Of The University Of Pennsylvania Compositions comprising angiogenic factors and methods of use thereof
US9919038B2 (en) 2009-03-04 2018-03-20 The Trustees Of The University Of Pennsylvania Compositions comprising angiogenic factors and methods of use thereof
US10016617B2 (en) 2009-11-11 2018-07-10 The Trustees Of The University Of Pennsylvania Combination immuno therapy and radiotherapy for the treatment of Her-2-positive cancers
US9943590B2 (en) 2010-10-01 2018-04-17 The Trustees Of The University Of Pennsylvania Use of Listeria vaccine vectors to reverse vaccine unresponsiveness in parasitically infected individuals
US9040053B2 (en) 2010-10-19 2015-05-26 Merial, Inc. Her2 DNA vaccine as adjunct treatment for cancers in companion animals
WO2012054294A1 (en) * 2010-10-19 2012-04-26 Merial Limited Her2 dna vaccine as adjunct treatment for cancers in companion animals
AU2011318369B2 (en) * 2010-10-19 2014-10-02 Boehringer Ingelheim Animal Health USA Inc. Her2 DNA vaccine as adjunct treatment for cancers in companion animals
US10064898B2 (en) 2011-03-11 2018-09-04 Advaxis, Inc. Listeria-based adjuvants
US10058599B2 (en) 2012-03-12 2018-08-28 Advaxis, Inc. Suppressor cell function inhibition following Listeria vaccine treatment
AU2014231214B2 (en) * 2013-03-11 2018-01-18 Icon Genetics Gmbh HER2/neu cancer vaccine
US10052370B2 (en) 2013-03-11 2018-08-21 Icon Genetics Gmbh HER2/Neu cancer vaccine
WO2014139672A1 (en) * 2013-03-11 2014-09-18 Icon Genetics Gmbh Her2/neu cancer vaccine
US10143734B2 (en) 2014-02-18 2018-12-04 Advaxis, Inc. Biomarker directed multi-target immunotherapy
US10258679B2 (en) 2014-04-24 2019-04-16 Advaxis, Inc. Recombinant Listeria vaccine strains and methods of producing the same
US10900044B2 (en) 2015-03-03 2021-01-26 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11702664B2 (en) 2015-03-03 2023-07-18 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11897927B2 (en) 2016-11-30 2024-02-13 Advaxis, Inc. Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof
US11179339B2 (en) 2017-09-19 2021-11-23 Advaxis, Inc. Compositions and methods for lyophilization of bacteria or listeria strains

Also Published As

Publication number Publication date
US20060093582A1 (en) 2006-05-04
AU2005289957A1 (en) 2006-04-06
WO2006036550A3 (en) 2006-08-10
ES2566392T3 (en) 2016-04-12
EP2980101B1 (en) 2019-06-05
US7820180B2 (en) 2010-10-26
EP1804831A4 (en) 2009-09-02
EP1804831A2 (en) 2007-07-11
EP1804831B1 (en) 2015-12-30
HK1221232A1 (en) 2017-05-26
JP2008514199A (en) 2008-05-08
DK1804831T3 (en) 2016-04-04
CA2581331A1 (en) 2006-04-06
EP2980101A1 (en) 2016-02-03

Similar Documents

Publication Publication Date Title
US7820180B2 (en) Listeria-based and LLO-based vaccines
US11446369B2 (en) Compositions and methods comprising KLK3 or FOLH1 antigen
US11702664B2 (en) Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US9084747B2 (en) Compositions and methods for prevention of escape mutation in the treatment of HER2/NEU over-expressing tumors
AU2012296474B2 (en) Compositions and methods for prevention of escape mutation in the treatment of Her2/neu over-expressing tumors
US20080124354A1 (en) Methods for administering tumor vaccines
DK2977456T3 (en) Compositions comprising HMW-MAA and fragments thereof for the treatment of cancer
US20110223187A1 (en) Live listeria-based vaccines for central nervous system therapy
JP6329211B2 (en) Use of recombinant Listeria strains in the manufacture of a medicament for delaying the onset of brain tumors or breast tumors
WO2007059030A2 (en) Llo-encoding dna/nucleic acid vaccines and methods comprising same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2581331

Country of ref document: CA

Ref document number: 2005289957

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2007533537

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2005289957

Country of ref document: AU

Date of ref document: 20050914

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005289957

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005811815

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005811815

Country of ref document: EP