WO2016154412A2 - Combination of a pd-1 antagonist and a listeria based vaccine for treating pancreatic cancer - Google Patents

Combination of a pd-1 antagonist and a listeria based vaccine for treating pancreatic cancer Download PDF

Info

Publication number
WO2016154412A2
WO2016154412A2 PCT/US2016/023969 US2016023969W WO2016154412A2 WO 2016154412 A2 WO2016154412 A2 WO 2016154412A2 US 2016023969 W US2016023969 W US 2016023969W WO 2016154412 A2 WO2016154412 A2 WO 2016154412A2
Authority
WO
WIPO (PCT)
Prior art keywords
another embodiment
cancer
fragment
tumor
survivin
Prior art date
Application number
PCT/US2016/023969
Other languages
French (fr)
Other versions
WO2016154412A9 (en
Inventor
Gregory L. BEATTY
Dawson KNOBLOCK
Robert Petit
Michael F. PRINCIOTTA
Anu Wallecha
Original Assignee
The Trustees Of The University Of Pennsylvania
Advaxis, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Trustees Of The University Of Pennsylvania, Advaxis, Inc. filed Critical The Trustees Of The University Of Pennsylvania
Publication of WO2016154412A2 publication Critical patent/WO2016154412A2/en
Publication of WO2016154412A9 publication Critical patent/WO2016154412A9/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0271Chimeric vertebrates, e.g. comprising exogenous cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/12Animals modified by administration of exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55544Bacterial toxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to combination therapies useful for the treatment of cancer.
  • the invention relates to the treatment of pancreatic cancer using an antagonist of a Programmed Death 1 protein (PD-1) in combination with a live attenuated recombinant Listeria strain comprising a fusion protein of a PEST sequence-containing polypeptide fused to a tumor-associated antigen.
  • PD-1 Programmed Death 1 protein
  • pancreatic cancer arises when cells in the pancreas begin to multiply out of control and form a mass. These cancer cells have the ability to invade other parts of the body.
  • pancreatic adenocarcinoma accounts for about 85% of cases.
  • pancreatic cancers of all types were the seventh most common cause of cancer deaths, resulting in 330,000 deaths globally.
  • pancreatic cancer is the fourth most common cause of deaths due to cancer. The disease occurs most often in the developed world, where about 70% of the new cases in 2012 originated.
  • Pancreatic adenocarcinoma typically has a very poor prognosis: after diagnosis, 25% of people survive one year and 5% live for five years.
  • pancreatic cancer For cancers diagnosed early the five-year survival rate rises to about 20%. Risk factors for pancreatic cancer include tobacco smoking, obesity, diabetes, and certain rare genetic conditions. Pancreatic cancer can be treated with surgery, radiotherapy, chemotherapy, palliative care, or a combination of these. Surgery is the only treatment that can cure the disease.
  • PD-1 is recognized as an important player in immune regulation and the maintenance of peripheral tolerance. PD-1 is moderately expressed on naive T, B and NKT cells and up-regulated by T/B cell receptor signaling on lymphocytes, monocytes and myeloid cells.
  • PD-Ll expressing tumor cells interact with PD-1 expressing T cells to attenuate T cell activation and evasion of immune surveillance, thereby contributing to an impaired immune response against the tumor.
  • monoclonal antibodies that inhibit the interaction between PD-1 and one or both of its ligands PD-Ll and PD-L2 are in clinical development for treating cancer.
  • Survivin an inhibitor of apoptosis protein, is encoded by the BIRC5 gene in humans. This protein is highly conserved across species (human and canine survivin proteins share 91.5% sequence identity). Survivin is a member of the inhibitor of apoptosis (IAP) family. The survivin protein functions to inhibit caspase activation, thereby leading to negative regulation of apoptosis or programmed cell death. Survivin is detectable in a wide range of adult tissues in humans, dogs and mice.
  • Survivin is highly expressed in most human cancers and associated with chemotherapy resistance, increased tumor recurrence, and shorter patient survival. Due to its presence in many types of cancer tumors, e.g. breast and colon cancer, lymphoma, leukemia, and melanoma, it is believed that survivin could serve as a universal target antigen for anticancer immunotherapy. Expression of survivin has also been reported in the vast majority of canine lymphomas, as well as in canine mast cell tumor, hemangiosarcoma, and transitional cell carcinoma. High levels of survivin expression in canine B cell lymphoma correlate with reduced disease-free interval (DFI) and median survival time (MST). Immunization of human patients using survivin peptides has shown some promise in the human clinics with only mild grade ⁇ / ⁇ adverse effects that include anemia, lethargy and fever. Serious adverse effects have not been reported.
  • DFI disease-free interval
  • MST median survival time
  • Listeria monocytogenes (L. monocytogenes or Lm) 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.
  • LLO listeriolysin O
  • 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. Since L. monocytogenes has access to both phagosomal and cytosolic compartments, antigens delivered by Lm can be presented in the context of both MHC I and ⁇ molecules, resulting in strong but preferentially cellular immune responses.
  • pancreatic cancer patients create a clear need for a more robust treatment of this type of cancer.
  • the present invention addresses the needs of subjects suffering from pancreatic cancer by providing a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen or immunogenic fragment thereof and an immunosuppressive molecule antagonist.
  • a method of eliciting an anti-tumor or anti-cancer immune response against a tumor or cancer in a subject comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist, thereby eliciting an anti-tumor or anti-cancer immune response in said subject.
  • a method of preventing a tumor or cancer in a subject comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration treats a tumor or a cancer in said subject.
  • the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer.
  • a method of treating a tumor or cancer in a subject comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration treats a tumor or a cancer in said subject.
  • the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer.
  • a method of delaying metastatic disease in a subject having tumor or cancer comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration delays metastatic disease in said subject.
  • a method of breaking tolerance to survivin in a subject having tumor or cancer comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration breaks tolerance to survivin in said subject.
  • a method of prolonging survival of a subject having tumor or cancer comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration prolongs survival in said subject.
  • a method of stimulating an anti-tumor immune response in a subject having tumor or cancer comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration stimulates an anti-tumor immune response in said subject.
  • said antagonist is an antibody or functional fragment thereof.
  • said immunosuppressive molecule is PD-1 B7-1, PD-L1, and PD-L2.
  • said recombinant Listeria strain comprises a nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide, wherein the said fusion polypeptide comprises a PEST sequence- containing polypeptide fused to survivin antigen or immunogenic fragment thereof.
  • said PEST-containing peptide or polypeptide is selected from the group consisting of an N-terminal non-hemolytic Listeriolysin (LLO) fragment, an N-terminal ActA fragment, and a PEST-containing amino acid sequence.
  • LLO N-terminal non-hemolytic Listeriolysin
  • said nucleic acid molecule comprises a second open reading frame encoding a metabolic enzyme, and wherein said metabolic enzyme complements a mutation in the chromosome of said recombinant Listeria strain.
  • the metabolic enzyme is an alanine racemase enzyme or a D-amino acid transferase enzyme.
  • said recombinant Listeria comprises a mutation in the actA virulence gene.
  • said recombinant attenuated Listeria is a Listeria monocytogenes.
  • said subject is a human.
  • administering a composition comprising said recombinant attenuated Listeria prevents escape mutations within a tumor or cancer, results in progression free survival, inhibiting tumor growth, inducing cancer regression, extension of progression free survival (PFS), increasing time to disease progression, or any combination thereof.
  • said tumor or cancer comprises a pancreatic tumor or pancreatic cancer.
  • said cancer is a refractory cancer.
  • said tumor is a survivin-expressing tumor.
  • said cancer is a survivin- expressing cancer.
  • Figures 1A-D Presents a representative image of Her2 staining by immunofluorescence (IF) on spontaneously arising tumor in the KPC (KrasG12D/+; Trp53R172H/+; Pdx-lCre) model ( Figure 1A).
  • Tumor cell lines derived from KPC mice showing Her2 expression by flow cytometry ( Figure IB).
  • Western blot analysis of tumor cell lines from KPC mice show Her2 expression (Figure 1C).
  • Figures 2A-C Presents a representative image of Her2 staining by immunofluorescence (IF) on spontaneously arising tumor in the KPC (KrasG12D/+; Trp53R172H/+; Pdx-lCre) model ( Figure 1A).
  • FIG. 1 Shows two tumor cell lines selected for studies: 7681. PDA, which shows minimal stromal compartment and 69. PDA with stromal compartment ( Figure 2A). Presents 14 day old established tumors (7681) that were treated weekly beginning on day 0 with Listeria vaccine constructs and wherein tumor volume was assessed at day +14 ( Figure 2B). Mice were challenged with 69. PDA tumors and treated with a single dose of vaccine on day 4 and tumor growths were monitored (Figure 2C). [0023] Figures 3A-B. Presents a treatment model schematic for prophylaxis therapy with
  • Figures 4A-D Presents flow cytometry analysis of tumor cell line for expression of surviving (intracellular analysis) ( Figure 4A).
  • Figure 4A Presents tumor growth curves for mice challenged with 69. PDA and treated beginning on day 10 with single dose of Listeria vaccine ( Figure 4B).
  • Figure 4B Presents LSL-Kras G12D/+ ;LSL-Trp53 R172H/+ ;Pdx-l-Cre (KPC) mice with established tumors that were treated with single dose of Listeria vaccines and wherein 14 days later tumor tissue was evaluated for T cell infiltration (Figure 4C).
  • Figure 4C Presents a representative IHC images of T cell infiltration 14 days after Listeria vaccine therapy (Figure 4D).
  • Figures 5A-C Presents flow cytometry analysis of tumor cell line for expression of surviving (intracellular analysis)
  • Figures 6A-D Presents the graph showing the impact of IFN-g treatment on percentage of tumor cells expressing MHC class I ( Figure 6A).
  • Figure 6B Presents the graph showing the impact of IFN-g treatment on the level of MHC expression
  • Figure 6C Presents the graph showing the impact of IFN-g treatment on percentage of tumor cells expressing PD-L1 ( Figure 6D).
  • Figures 7A-D Presents the treatment model schematic for Listeria vaccine therapy in combination with PD-1 blockade (Figure 7A).
  • Figure 7B Presents the individual mouse growth curves ( Figure 7B).
  • Figure 7C Presents the average growth curves ( Figure 7C).
  • Figure 7D Presents the overall survival ( Figure 7D). DETAILED DESCRIPTION OF THE PRESENT INVENTION
  • ADXS31-265 Listeria monocytogenes (10403S dal dat actA'
  • TILs Tumor infiltrating lymphocytes
  • a recombinant attenuated Listeria strain comprising a nucleic acid molecule encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof or an immunogenic fragment thereof fused to a PEST-containing peptide or polypeptide.
  • the nucleic acid molecule comprises a first open reading frame encoding the fusion polypeptide.
  • a PEST-containing peptide or polypeptide is a truncated Listeriolysin O (LLO) protein (tLLO).
  • LLO Listeriolysin O
  • the PEST-containing peptide or polypeptide is a fragment of LLO.
  • the PEST-containing peptide or polypeptide is an N- terminal fragment of LLO. In another embodiment, a PEST-containing polypeptide or polypeptide is a truncated ActA protein. In another embodiment, a PEST-containing peptide or polypeptide is fragment of an ActA protein. In another embodiment, the PEST-containing peptide or polypeptide is an N-terminal fragment of ActA. In another embodiment, the PEST-containing peptide or polypeptide is PEST-containing amino acid sequence. In another embodiment, the nucleic acid molecule further comprises a second open reading frame encoding a metabolic enzyme, wherein the metabolic enzyme complements a mutation in the chromosome of the recombinant Listeria strain.
  • nucleic acids or “nucleotide” may encompass a string of at least two base-sugar-phosphate 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.
  • 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.
  • 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.
  • the nucleotide is oxetane modified. In another embodiment, 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 9:353-57; and Raz NK et al Biochem Biophys Res Commun. 297: 1075-84.
  • nucleic acid derivative represents a separate embodiment as provided herein.
  • amino acid or “amino acids” is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phospho threonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine.
  • amino acid may include both D- and L-amino acids.
  • ORF open reading frame
  • the term "open reading frame” or "ORF” may encompass a portion of an organism's genome which contains a sequence of bases that could potentially encode a protein.
  • the start and stop ends of the ORF are not equivalent to the ends of the mRNA, but they are usually contained within the mRNA.
  • ORFs are located between the start-code sequence (initiation codon) and the stop- codon sequence (termination codon) of a gene.
  • a nucleic acid molecule operably integrated into a genome as an open reading frame with an endogenous polypeptide is a nucleic acid molecule that has integrated into a genome in the same open reading frame as an endogenous polypeptide.
  • endogenous may encompass an item that has developed or originated within the reference organism or arisen from causes within the reference organism.
  • endogenous refers to native.
  • fragment may encompass a protein or polypeptide that is shorter or comprises fewer amino acids than the full length protein or polypeptide.
  • a fragment is an N-terminal fragment.
  • a fragment is a C-terminal fragment.
  • a fragment is an intrasequential section of the protein or peptide.
  • a fragment is a functional fragment.
  • a fragment is an immunogenic fragment.
  • a fragment has 10-20 amino acids, while in another embodiment, a fragment has more than 5 amino acids, while in another embodiment, a fragment has 100-200 amino acids, while in another embodiment, a fragment has 100-500 amino acids, while in another embodiment, a fragment has 50-200 amino acids, while in another embodiment, a fragment has 10-250 amino acids.
  • fragment refers to a nucleic acid that is shorter or comprises fewer nucleotides than the full length nucleic acid. In one embodiment, a fragment is a 5 '-terminal fragment. In another embodiment, a fragment is a 3 '-terminal fragment.
  • a fragment encodes an intrasequential section of the protein.
  • a fragment has 10-20 nucleotides, while in another embodiment, a fragment has more than 5 nucleotides, while in another embodiment, a fragment has 100-200 nucleotides, while in another embodiment, a fragment has 100-500 nucleotides, while in another embodiment, a fragment has 50-200 nucleotides, while in another embodiment, a fragment has 10-250 nucleotides.
  • the term "functional" within the meaning of the invention may encompass the innate ability of a protein, peptide, nucleic acid, fragment or a variant thereof to exhibit a biological activity or function.
  • a biological function is its binding property to an interaction partner, e.g., a membrane-associated receptor, and in another embodiment, its trimerization property.
  • these biological functions may in fact be changed, e.g., with respect to their specificity or selectivity, but with retention of the basic biological function.
  • a functional fragment may encompass an immunogenic fragment and elicits an immune response when administered to a subject alone or in a strain composition provided herein.
  • a functional fragment has biological activity as will be understood by a skilled artisan and as further provided herein.
  • fused may encompass an operable linkage by covalent bonding.
  • the term encompasses recombinant fusion (of nucleic acid sequences or open reading frames thereof).
  • the term encompasses chemical conjugation.
  • an LLO sequence comprises PEST amino acid (AA) sequence.
  • the PEST amino acid sequence is a putative PEST sequence present in LLO.
  • a truncated LLO comprises a PEST amino acid (AA) sequence.
  • a PEST AA sequence comprises a truncated LLO sequence.
  • a PEST AA sequence comprises an LLO fragment sequence.
  • a PEST AA sequence comprises a truncated ActA sequence.
  • a truncated ActA sequence comprises a PEST sequence.
  • PEST AA sequence comprises an ActA fragment sequence.
  • the PEST amino acid sequence may be KENS IS S M APP AS PP AS PKTPIEKKH ADEID K (SEQ ID NO: 1). Furthermore, fusion of an antigen to other Listeria monocytogenes (Lm) PEST AA sequences from Listeria may enhance immunogenicity of the antigen.
  • Lm Listeria monocytogenes
  • truncated LLO an "N-terminal LLO fragment” or “ALLO” are used interchangeably herein and refer to a non-hemolytic fragment of LLO that comprises a putative PEST sequence.
  • the terms refer to an LLO fragment that comprises a PEST domain.
  • the LLO fragment is at least 492 amino acids (AA) long.
  • the LLO fragment is 492-528 AA long.
  • the LLO fragment consists of about the first 441 AA of the LLO protein. In another embodiment, the LLO fragment consists of about the first 420 AA of LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
  • 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 1175. 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.
  • 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.
  • the LLO fragment contains residues of a homologous LLO protein that correspond to one of the above AA ranges.
  • the residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous LLO protein has an insertion or deletion, relative to an LLO protein utilized herein, then the residue numbers can be adjusted accordingly.
  • the LLO fragment is any other LLO fragment known in the art.
  • an N-terminal LLO protein fragment of the methods and compositions disclosed herein comprises, in one embodiment, SEQ ID No: 3.
  • the LLO fragment comprises an LLO signal peptide.
  • the LLO fragment comprises SEQ ID No: 4.
  • the LLO fragment consists of SEQ ID No: 4.
  • the LLO fragment consists essentially of SEQ ID No: 4.
  • the LLO fragment corresponds to SEQ ID No: 4.
  • the LLO fragment is homologous to SEQ ID No: 4.
  • the LLO fragment is homologous to a fragment of SEQ ID No: 4.
  • ALLO The LLO fragments (ALLO) used in some of the Examples was 416 AA long (exclusive of the signal sequence). In this truncated fragment, 88 residues from the carboxy terminus (which contain the activation domain, including cysteine 484) were removed. It will be understood by those skilled in the art that any ALLO without the activation domain, and in particular without cysteine 484, are suitable for methods and compositions of the present invention. In another embodiment, fusion of a heterologous antigen, such as survivin antigen or fragment thereof to any ALLO enhances cell mediated and anti-tumor immunity of the antigen.
  • a heterologous antigen such as survivin antigen or fragment thereof to any ALLO enhances cell mediated and anti-tumor immunity of the antigen.
  • the LLO fragment lacks the cholesterol-binding domain (CBD). LLO mutants lacking the CBD are disclosed in US Patent Serial No. 8,771,702, which is incorporated by reference herein in its entirety.
  • PEST amino acid sequence PEST AA sequence
  • PEST sequence-containing polypeptide PEST sequence-containing protein
  • PEST-containing peptide or polypeptide may encompass a PEST sequence peptide or a fragment of an LLO protein or a fragment of an ActA protein.
  • PEST sequence peptides are known in the art and are described in US Patent Serial No. 7,635,479, and in US Patent Publication Serial No. 2014/0186387, both of which are hereby incorporated in their entirety herein.
  • fusion of an antigen to the PEST sequence of Listeria monocytogenes enhances cell mediated and anti-tumor immunity of the antigen.
  • fusion of an antigen to PEST- amino acid sequences from other prokaryotic organisms would be expected to have similar effect.
  • the PEST sequence is embedded within the antigenic protein.
  • fusion refers to an antigenic protein comprising both the antigen and the PEST amino acid sequence either linked at one end of the antigen or embedded within the antigen. PEST sequences derived from other prokaryotic organisms will also enhance immunogenicity of the antigen.
  • a PEST sequence of prokaryotic organisms can be identified routinely in accordance with methods such as described by Rechsteiner and Roberts (TBS 21:267-271, 1996) for L. monocytogenes.
  • PEST amino acid sequences from other prokaryotic organisms can also be identified based by this method.
  • Other prokaryotic organisms wherein PEST amino acid sequences would be expected include, but are not limited to, other Listeria species.
  • the L. monocytogenes protein ActA contains four such sequences.
  • KTEEQPS E VNTGPR SEQ ID NO: 5
  • KASVTDTSEGDLDSSMQSADESTPQPLK SEQ ID NO: 6
  • KNEE VN AS DFPPPPTDEELR SEQ ID NO: 7
  • RGGIPTS EEFS SLNS GDFTDDENS ETTEEEIDR (SEQ ID NO: 8).
  • Streptolysin O from Streptococcus sp. contains a PEST sequence.
  • Streptococcus pyogenes Streptolysin O comprises the PEST sequence KQNTASTETTTTNEQPK (SEQ ID NO: 9) at amino acids 35-51 and Streptococcus equisimilis Streptolysin O comprises the PEST sequence KQNTANTETTTTNEQPK (SEQ ID NO: 10) at amino acids 38-54.
  • the PEST sequence can be embedded within the antigenic protein.
  • fusion when in relation to PEST sequence fusions, it is meant that the antigenic protein comprises both the antigen and the PEST amino acid sequence either linked at one end of the antigen or embedded within the antigen.
  • immunogenicity or “immunogenic” may encompass the innate ability of a protein, peptide, nucleic acid, antigen or organism to elicit an immune response in an animal when the protein, peptide, nucleic acid, antigen or organism is administered to the animal.
  • enhancing the immunogenicity in one embodiment, refers to increasing the ability of a protein, peptide, nucleic acid, antigen or organism to elicit an immune response in an animal when the protein, peptide, nucleic acid, antigen or organism is administered to an animal.
  • the increased ability of a protein, peptide, nucleic acid, antigen or organism to elicit an immune response can be measured by, in one embodiment, a greater number of antibodies to a protein, peptide, nucleic acid, antigen or organism, a greater diversity of antibodies to an antigen or organism, a greater number of T-cells specific for a protein, peptide, nucleic acid, antigen or organism, a greater cytotoxic or helper T- cell response to a protein, peptide, nucleic acid, antigen or organism, and the like.
  • the construct or nucleic acid molecule is expressed from an episomal or plasmid vector, with a nucleic acid sequence encoding a PEST sequence-containing polypeptide or a PEST-sequence peptide.
  • the plasmid is stably maintained in the recombinant Listeria vaccine strain in the absence of antibiotic selection.
  • the plasmid does not confer antibiotic resistance upon the recombinant Listeria.
  • the fragment is a functional fragment.
  • the fragment is an immunogenic fragment.
  • the term "vector” may encompass a plasmid.
  • the term may encompass an integration vector capable of being transformed into the Listeria host and being incorporated in the Listeria's, chromosome in a manner that allows expression of the genes comprised by the vector.
  • the term may encompass a non-integration vector that does not integrate in the Listeria's, chromosome but instead is present in the cytoplasm of the Listeria.
  • the term may encompass a plasmid comprising an integration vector.
  • the integration vector is a site- specific integration vector.
  • transcriptional promoters e.g. those in commercially available cloning vectors
  • these functionalities are provided in, for example, the commercially available vectors known as the pUC series.
  • non-essential DNA sequences e.g. antibiotic resistance genes
  • a commercially available plasmid is used in the present invention.
  • plasmids are available from a variety of sources, for example, Invitrogen (La JoUa, CA), Stratagene (La Jolla, CA), Clontech (Palo Alto, CA), or can be constructed using methods well known in the art.
  • Another embodiment is a plasmid such as pCR2.1 (Invitrogen, La Jolla, CA), which is a prokaryotic expression vector with a prokaryotic origin of replication and promoter/regulatory elements to facilitate expression in a prokaryotic organism.
  • extraneous nucleotide sequences are removed to decrease the size of the plasmid and increase the size of the cassette that can be placed therein.
  • the LLO protein utilized to construct vaccines disclosed herein has, in another embodiment, the sequence:
  • the full length active LLO protein is 504 residues long.
  • the above LLO fragment is used as the source of the LLO fragment incorporated in a vaccine of the present invention.
  • N-terminal fragment of an LLO protein utilized in compositions and methods disclosed herein has the sequence:
  • the LLO fragment corresponds to about AA 20-442 of an LLO protein utilized herein.
  • the LLO fragment has the sequence:
  • truncated LLO refers to a fragment of LLO that comprises the PEST-amino acid sequence.
  • the term refers to an LLO fragment that comprises a PEST sequence.
  • ALLO refers to 416AA LLO fragment as defined above.
  • the terms truncated LLO, tLLO, or ALLO refer to an LLO fragment that does not contain the activation domain at the amino terminus and does not include cysteine 484. In another embodiment, the terms refer to an LLO fragment that is not hemolytic. In another embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation of the activation domain. In another embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation of cysteine 484. In another embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation at another location. In another embodiment, the LLO is rendered nonhemolytic by a deletion or mutation of the cholesterol binding domain (CBD) as detailed in US Patent No.
  • CBD cholesterol binding domain
  • the recombinant nucleotide encoding a truncated LLO protein comprises the sequence set forth in SEQ ID NO: 11: aaaaaataatgctagtttttattacacttatattag ttagtctaccaattgcgcaacaaactgaagcaaaggatgcatctgcattcaataaagaaaattcaatttcatccatggcaccaccaccagcatctccgcc tgcaagtcctaagacgccaatcgaaaagaaacacgcggatgaaatcgataagtatatacaaggattggattacaataaaaacaatgtattagtata ccacggagatgcagtgtgccgccaagaaaaggtttacaagatggaaaaagatggaaaatgaatatattgttttttttttt
  • the recombinant nucleotide has the sequence set forth in SEQ ID NO: 11. In another embodiment, the recombinant nucleotide comprises a sequence that encodes a fragment of an LLO protein.
  • the LLO fragment consists of about the first 441 AA of the LLO protein. In another embodiment, the LLO fragment consists of about the first 420 AA of LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
  • 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.
  • 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.
  • an LLO fragment disclosed herein contains residues of a homologous LLO protein that correspond to one of the above AA ranges.
  • the residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous LLO protein has an insertion or deletion, relative to an LLO protein utilized herein, then the residue numbers can be adjusted accordingly.
  • Other LLO fragments are known in the art.
  • the LLO fragment consists of about the first 441 AA of the LLO protein. In another embodiment, the LLO fragment consists of about the first 420 AA of LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
  • a homologous LLO refers to identity to an LLO sequence (e.g. to one of SEQ ID No: 2-4) of greater than 70%.
  • a homologous LLO refers to identity to one of SEQ ID No: 2-4 of greater than 72%.
  • a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 75%.
  • a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 78%.
  • a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 80%.
  • a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 82%.
  • a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 83%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 85%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 87%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 88%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 90%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 92%.
  • a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 93%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 95%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 96%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 97%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 98%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 99%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of 100%.
  • an ActA protein comprises the sequence set forth in SEQ ID NO: 12: [00104] MGLNRFMRAMMVVFITANCrriNPDIIFAATDSEDSSLNTDEWEEEKTEEQPSEVN TGPRYETAREVSSRDIKELEKSNKVRNTNKADLIAMLKEKAEKGPNINNNNSEQTENAAIN EEASGADRPAIQVERRHPGLPSDSAAEIKKRRKAIASSDSELESLTYPDKPTKVNKKKVAK ESVADASESDLDSSMQSADESSPQPLKANQQPFFPKVFKKIKDAGKWVRDKIDENPEVKK
  • the first 29 AA of the proprotein corresponding to this sequence are the signal sequence and are cleaved from ActA protein when it is secreted by the bacterium.
  • an ActA polypeptide or peptide comprises the signal sequence, AA 1-29 of SEQ ID NO: 12 above.
  • an ActA polypeptide or peptide does not include the signal sequence, AA 1-29 of SEQ ID NO: 12 above.
  • a truncated ActA protein comprises an N-terminal fragment of an ActA protein.
  • a truncated ActA protein is an N-terminal fragment of an ActA protein.
  • a truncated ActA protein comprises the sequence set forth in SEQ ID NO: 13:
  • the ActA fragment comprises the sequence set forth in SEQ ID NO: 13.
  • a truncated ActA protein comprises the sequence set forth in SEQ ID NO: 14: MGLNRFMRAMMVVFITANCITINPDIIFAATDSEDSSLNTDEWEEEKTEEQPSE VNTGPRYETAREVSSRDIKELEKSNKVRNTNKADLIAMLKEKAEKG.
  • the ActA fragment is any other ActA fragment known in the art.
  • the recombinant nucleotide encoding a truncated ActA protein comprises the sequence set forth in SEQ ID NO: 15:atgcgtgcgatgatggtggttttcattac tgccaattgcattacgattaaccccgacataatatttgcagcgacagatagcgaagattctagtctaaacacagatgaatgggaagaagaaaaaac agaagagcaaccaagcgaggtaaatacgggaccaagatacgaaactgcacgtgaagtaagttcacgtgatattaaagaactagaaaaatcgaa taaagtgagaaatacgaacaaagcagacctaatagcaatgttgaaagaaaaaaaggtccaaatatcaataataatacaacagt
  • the recombinant nucleotide has the sequence set forth in SEQ ID NO: 15. In another embodiment, the recombinant nucleotide comprises other sequences that encode a fragment of an ActA protein.
  • truncated ActA or "AActA” refers to a fragment of ActA that comprises at least one PEST sequence. In another embodiment, the terms refer to an ActA fragment that comprises more than one PEST sequence.
  • the N-terminal ActA protein fragment of methods and compositions disclosed herein comprises, in one embodiment, SEQ ID No: 13.
  • the ActA fragment comprises an ActA signal peptide.
  • the ActA fragment comprises SEQ ID No: 14.
  • the ActA fragment consists approximately of SEQ ID No: 13 or SEQ ID No: 14.
  • the ActA fragment consists essentially of SEQ ID No: 13 or SEQ ID No: 14.
  • the ActA fragment corresponds to SEQ ID No: 13 or SEQ ID No: 14.
  • the ActA fragment is homologous to SEQ ID No: 13 or SEQ ID No: 14.
  • the ActA fragment is homologous to a fragment of SEQ ID No: 13 or SEQ ID No: 14.
  • a PEST- sequence is another PEST- AA sequence derived from a prokaryotic organism.
  • the PEST- sequence may be other PEST- sequences known in the art.
  • the ActA fragment consists of about the first 100 AA of the ActA protein.
  • the ActA fragment consists of about residues 1-25. In another embodiment, the ActA fragment consists of about residues 1-50. In another embodiment, the ActA fragment consists of about residues 1-75. In another embodiment, the ActA fragment consists of about residues 1-100. In another embodiment, the ActA fragment consists of about residues 1-125. In another embodiment, the ActA fragment consists of about residues 1-150. In another embodiment, the ActA fragment consists of about residues 1-175. In another embodiment, the ActA fragment consists of about residues 1-200. In another embodiment, the ActA fragment consists of about residues 1-225. In another embodiment, the ActA fragment consists of about residues 1-250.
  • the ActA fragment consists of about residues 1-275. In another embodiment, the ActA fragment consists of about residues 1-300. In another embodiment, the ActA fragment consists of about residues 1-325. In another embodiment, the ActA fragment consists of about residues 1-338. In another embodiment, the ActA fragment consists of about residues 1-350. In another embodiment, the ActA fragment consists of about residues 1-375. In another embodiment, the ActA fragment consists of about residues 1-400. In another embodiment, the ActA fragment consists of about residues 1-450. In another embodiment, the ActA fragment consists of about residues 1-500. In another embodiment, the ActA fragment consists of about residues 1-550. In another embodiment, the ActA fragment consists of about residues 1-600.
  • the ActA fragment consists of about residues 1-639. In another embodiment, the ActA fragment consists of about residues 30-100. In another embodiment, the ActA fragment consists of about residues 30-125. In another embodiment, the ActA fragment consists of about residues 30- 150. In another embodiment, the ActA fragment consists of about residues 30-175. In another embodiment, the ActA fragment consists of about residues 30-200. In another embodiment, the ActA fragment consists of about residues 30-225. In another embodiment, the ActA fragment consists of about residues 30-250. In another embodiment, the ActA fragment consists of about residues 30-275. In another embodiment, the ActA fragment consists of about residues 30-300. In another embodiment, the ActA fragment consists of about residues 30-325.
  • the ActA fragment consists of about residues 30-338. In another embodiment, the ActA fragment consists of about residues 30-350. In another embodiment, the ActA fragment consists of about residues 30-375. In another embodiment, the ActA fragment consists of about residues 30-400. In another embodiment, the ActA fragment consists of about residues 30-450. In another embodiment, the ActA fragment consists of about residues 30-500. In another embodiment, the ActA fragment consists of about residues 30-550. In another embodiment, the ActA fragment consists of about residues 1-600. In another embodiment, the ActA fragment consists of about residues 30-604.
  • an ActA fragment disclosed herein contains residues of a homologous ActA protein that correspond to one of the above AA ranges.
  • the residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous ActA protein has an insertion or deletion, relative to an ActA protein utilized herein, then the residue numbers can be adjusted accordingly.
  • Other ActA fragments are known in the art.
  • a homologous ActA refers to identity of an ActA sequence (e.g. to one of SEQ ID No: 12) of greater than 70%.
  • a homologous ActA refers to identity to one of SEQ ID No: 12 of greater than 72%.
  • a homologous refers to identity to one of SEQ ID No: 12 of greater than 75%.
  • a homologous refers to identity to one of SEQ ID No: 12 of greater than 78%.
  • a homologous refers to identity to one of SEQ ID No: 12 of greater than 80%.
  • a homologous refers to identity to one of SEQ ID No: 12 of greater than 82%.
  • a homologous refers to identity to one of SEQ ID No: 12 of greater than 83%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 85%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 87%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 88%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 greater than 90%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12of greater than 92%.
  • a homologous refers to identity to one of SEQ ID No: 12 of greater than 93%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 95%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 96%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 97%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 98%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 99%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of 100%.
  • Homology may be determined by a 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 the sequences disclosed herein of greater than 68%. In another embodiment, “homology” refers to identity to a sequence selected from the sequences disclosed herein of greater than 70%. In another embodiment, “homology” refers to identity to a sequence selected from the sequences disclosed herein 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%.
  • 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%.
  • LLO protein, Act A protein, or a fragment thereof that are provided for in the present invention need not be that which is set forth exactly in the sequences set forth herein, but rather that other alterations, modifications, or changes can be made that retain the functional characteristics of an LLO or ActA protein fused to an antigen as set forth elsewhere herein.
  • the present invention utilizes an analog of an LLO protein, ActA protein, or fragment thereof. Analogs differ, in another embodiment, from naturally occurring proteins or peptides by conservative AA sequence differences or by modifications which do not affect sequence, or by both.
  • Constantly modified variants may encompass substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity or other desired property of the protein, such as antigen affinity and/or specificity.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)).
  • substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity.
  • homology is determined 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. J., 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 °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 .
  • a recombinant Listeria strain disclosed herein lacks antibiotic resistance genes.
  • the recombinant Listeria strain disclosed herein comprises a plasmid comprising a nucleic acid encoding an antibiotic resistance gene.
  • the recombinant Listeria strain disclosed herein comprises a plasmid that does not encode an antibiotic resistance gene.
  • a recombinant Listeria disclosed herein is capable of escaping the phagolysosome.
  • a polypeptide disclosed herein is a fusion protein comprising an additional polypeptide selected from the group consisting of: non-hemolytic LLO protein or an N- terminal fragment thereof, a PEST sequence, or an ActA protein or a fragment thereof.
  • an additional polypeptide is fused to the survivin antigen.
  • an additional polypeptide is functional.
  • a fragment of an additional polypeptide is immunogenic.
  • an additional polypeptide is immunogenic.
  • a nucleic acid sequence encoding survivin or an antigenic portion thereof is integrated in frame with LLO in the Listeria chromosome.
  • an integrated nucleic acid sequence encoding survivin or an antigenic portion thereof is integrated in frame with ActA at the actA locus.
  • the chromosomal nucleic acid encoding ActA is replaced by a nucleic acid molecule comprising a sequence encoding an antigen.
  • the antigen is survivin or a fragment thereof.
  • the survivin is human survivin.
  • antigenic portion thereof, a fragment thereof and “immunogenic portion thereof in regard to a protein, peptide or polypeptide are used interchangeably herein and may encompass a protein, polypeptide, peptide, including recombinant forms thereof comprising a domain or segment that leads to the mounting of an immune response when present in, or, in some embodiments, detected by, a host, either alone, or in the context of a fusion protein, as described herein.
  • a nucleic acid molecule disclosed herein comprises a first open reading frame encoding a recombinant polypeptide comprising a survivin or an antigenic fragment thereof.
  • the recombinant polypeptide further comprises a truncated LLO protein, a truncated ActA protein or PEST sequence peptide fused to a survivin or an antigenic portion thereof.
  • the truncated LLO protein is an N-terminal LLO or fragment thereof.
  • the truncated ActA protein is an N-terminal ActA protein or fragment thereof.
  • a nucleic acid molecule disclosed herein further comprises a second open reading frame encoding a metabolic enzyme.
  • the metabolic enzyme complements a mutation in the chromosome of the recombinant Listeria strain.
  • the metabolic enzyme encoded by the second open reading frame is an alanine racemase enzyme ⁇ dal).
  • the metabolic enzyme encoded by the second open reading frame is a D-amino acid transferase enzyme (dat).
  • the Listeria strains disclosed herein comprise a mutation in the endogenous dal/ dat genes.
  • the Listeria lacks the dal/dat genes.
  • the Listeria lacks the dal/dat and actA genes.
  • a nucleic acid molecule of the methods and compositions disclosed herein is operably linked to a promoter/regulatory sequence.
  • the first open reading frame of methods and compositions disclosed herein is operably linked to a promoter/regulatory sequence.
  • the second open reading frame of methods and compositions disclosed herein is operably linked to a promoter/regulatory sequence.
  • each of the open reading frames is operably linked to a promoter/regulatory sequence.
  • operably linked may encompass a transcriptional and translational regulatory nucleic acid that is positioned relative to any coding sequences in such a manner that transcription is initiated. Generally, this will mean that the promoter and transcriptional initiation or start sequences are positioned 5' to the coding region.
  • operably linked refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • the term "metabolic enzyme” may encompass an enzyme involved in synthesis of a nutrient required by the host bacteria.
  • the term refers to an enzyme required for synthesis of a nutrient required by the host bacteria.
  • the term refers to an enzyme involved in synthesis of a nutrient utilized by the host bacteria.
  • the term refers to an enzyme involved in synthesis of a nutrient required for sustained growth of the host bacteria.
  • the enzyme is required for synthesis of the nutrient.
  • a recombinant Listeria is an attenuated auxotrophic strain.
  • a recombinant auxotrophic strain comprises strains described in US Patent No. 8,114,414, which is incorporated by reference herein in its entirety.
  • the attenuated strain is Lm dal ⁇ -)dat ⁇ -) (Lmdd).
  • the attenuated strains is Lm dal(-)dat(-)AactA (LmddA).
  • LmddA is based on a Listeria vaccine vector which is attenuated due to the deletion of virulence gene actA and retains the plasmid for a survivin antigen fusion polypeptide in vivo and in vitro by complementation of dal gene.
  • an attenuated strain disclosed herein is ADXS31-265, which comprises a pAdv 265.5 plasmid (SEQ ID NO: 16).
  • the attenuated strain is LmAactA.
  • the attenuated strain is LmAprfA.
  • the attenuated strain is LmAplcB.
  • the attenuated strain is LmAplcA.
  • the attenuated strain is LmAinlA.
  • the attenuated strain is LmAinlB.
  • the attenuated strain is LmAinlC.
  • the strain is the double mutant or triple mutant of any of the above-mentioned strains. In another embodiment, this strain exerts a strong adjuvant effect which is a property of Listeria-based vaccines. In another embodiment, this strain is constructed from the EGD Listeria backbone. In another embodiment, the strain used in the invention is a Listeria strain that expresses a nonhemolytic LLO or a fragment thereof.
  • a Listeria strain disclosed herein is an auxotrophic mutant.
  • the Listeria strain is deficient in a gene encoding a vitamin synthesis gene.
  • the Listeria strain is deficient in a gene encoding pantothenic acid synthase.
  • a Listeria strain disclosed herein is deficient in an amino acid (AA) metabolism enzyme.
  • the generation of auxotrophic strains of Listeria deficient in D-alanine may be accomplished in a number of ways that are well known to those of skill in the art, including deletion mutagenesis, insertion mutagenesis, and mutagenesis which results in the generation of frameshift mutations, mutations which cause premature termination of a protein, or mutation of regulatory sequences which affect gene expression.
  • mutagenesis can be accomplished using recombinant DNA techniques or using traditional mutagenesis technology using mutagenic chemicals or radiation and subsequent selection of mutants.
  • deletion mutants are preferred because of the accompanying low probability of reversion of the auxotrophic phenotype.
  • mutants of D-alanine which are generated according to the protocols presented herein may be tested for the ability to grow in the absence of D-alanine in a simple laboratory culture assay.
  • those mutants which are unable to grow in the absence of this compound are selected for further study.
  • a metabolic enzyme complements a mutation in the chromosome of the recombinant bacterial strain.
  • an endogenous metabolic gene is mutated in the chromosome.
  • an endogenous metabolic gene is deleted from the chromosome.
  • a metabolic enzyme is an amino acid metabolism enzyme.
  • a metabolic enzyme catalyzes a formation of an amino acid used for a cell wall synthesis in the recombinant Listeria strain.
  • a metabolic enzyme is an alanine racemase enzyme.
  • a metabolic enzyme is a D-amino acid transferase enzyme.
  • an episomal expression vector may encompass a nucleic acid vector which may be linear or circular, and which is usually double- stranded in form.
  • an episomal expression vector comprises a gene of interest.
  • the inserted gene of interest is not interrupted or subjected to regulatory constraints which often occur from integration into cellular DNA.
  • the presence of the inserted heterologous gene does not lead to rearrangement or interruption of the cell's own important regions.
  • episomal vectors persist in multiple copies in the bacterial cytoplasm, resulting in amplification of the gene of interest, and, in another embodiment, viral trans-acting factors are supplied when necessary.
  • the episomal expression vectors of the methods and compositions as disclosed herein may be delivered to cells in vivo, ex vivo, or in vitro by any of a variety of the methods employed to deliver DNA molecules to cells.
  • the vectors may also be delivered alone or in the form of a pharmaceutical composition that enhances delivery to cells of a subject.
  • an auxotrophic Listeria strain disclosed herein comprises an episomal expression vector comprising a metabolic enzyme that complements the auxotrophy of the auxotrophic Listeria strain.
  • 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.
  • the episomal expression vector lacks an antibiotic resistance marker.
  • an antigen, for example, a survivin antigen, of the methods and compositions as disclosed herein is fused to a polypeptide comprising a PEST sequence.
  • the polypeptide comprising a PEST sequence is a truncated LLO.
  • the polypeptide comprising a PEST sequence is ActA.
  • a Listeria strain disclosed herein is deficient in an AA metabolism enzyme.
  • the Listeria strain is deficient in a D-glutamic acid synthase gene.
  • the Listeria strain is deficient in the dat gene.
  • the Listeria strain is deficient in the dal gene.
  • the Listeria strain is deficient in the dga gene.
  • the Listeria strain is deficient in a gene involved in the synthesis of diaminopimelic acid (DAP).
  • the Listeria strain is deficient in a gene involved in the synthesis of Cysteine synthase A (CysK).
  • the gene is vitamin-B 12 independent methionine synthase.
  • the gene is trpA.
  • the gene is trpB.
  • the gene is trpE.
  • the gene is asnB.
  • the gene is gltD.
  • the gene is gltB.
  • the gene is leuA.
  • the gene is argG.
  • the gene is thrC.
  • the Listeria strain is deficient in one or more of the genes described hereinabove.
  • a Listeria strain disclosed herein is deficient in a synthase gene.
  • the gene is an AA synthesis gene.
  • the gene is folP.
  • the gene is dihydrouridine synthase family protein.
  • the gene is ispD.
  • the gene is ispF.
  • the gene is phosphoenolpyruvate synthase.
  • the gene is hisF.
  • the gene is hisH.
  • the gene is flil.
  • the gene is ribosomal large subunit pseudouridine synthase.
  • the gene ispD.
  • the gene is bifunctional GMP synthase/glutamine amidotransferase protein.
  • the gene is cobS.
  • the gene is cobB.
  • the gene is cbiD.
  • the gene is uroporphyrin- III C- methyltransferase/ uroporphyrinogen-III synthase.
  • the gene is cobQ.
  • the gene is uppS.
  • the gene is truB.
  • the gene is dxs.
  • the gene is mvaS.
  • the gene is dapA.
  • the gene is ispG.
  • the gene is folC. In another embodiment, the gene is citrate synthase. In another embodiment, the gene is argj. In another embodiment, the gene is 3-deoxy-7-phosphoheptulonate synthase. In another embodiment, the gene is indole-3-glycerol-phosphate synthase. In another embodiment, the gene is anthranilate synthase/ glutamine amidotransferase component. In another embodiment, the gene is metiB. In another embodiment, the gene is menaquinone- specific isochorismate synthase. In another embodiment, the gene is phosphoribosylformylglycinamidine synthase I or II.
  • the gene is phosphoribosylaminoimidazole-succinocarboxamide synthase.
  • the gene is carB. In another embodiment, the gene is car A. In another embodiment, the gene is thy A. In another embodiment, the gene is mgsA. In another embodiment, the gene is aroB. In another embodiment, the gene is hepB. In another embodiment, the gene is rluB. In another embodiment, the gene is ilvB. In another embodiment, the gene is ilvN. In another embodiment, the gene is alsS. In another embodiment, the gene is fabF. In another embodiment, the gene is fabH. In another embodiment, the gene is pseudouridine synthase.
  • the gene is pyrG. In another embodiment, the gene is truA. In another embodiment, the gene is pabB. In another embodiment, the gene is an atp synthase gene (e.g. atpC, atpD-2, aptG, atpA-2, etc).
  • the gene is phoP. In another embodiment, the gene is aroA. In another embodiment, the gene is aroC. In another embodiment, the gene is aroD. In another embodiment, the gene is plcB.
  • a Listeria strain disclosed herein is deficient in a peptide transporter.
  • the gene is ABC transporter/ ATP-binding/permease protein.
  • the gene is oligopeptide ABC transporter/ oligopeptide-binding protein.
  • the gene is oligopeptide ABC transporter/ permease protein.
  • the gene is zinc ABC transporter/ zinc-binding protein.
  • the gene is sugar ABC transporter.
  • the gene is phosphate transporter.
  • the gene is ZIP zinc transporter.
  • the gene is drug resistance transporter of the EmrB/QacA family.
  • the gene is sulfate transporter.
  • the gene is proton-dependent oligopeptide transporter. In another embodiment, the gene is magnesium transporter. In another embodiment, the gene is formate/nitrite transporter. In another embodiment, the gene is spermidine/putrescine ABC transporter. In another embodiment, the gene is Na/Pi-cotransporter. In another embodiment, the gene is sugar phosphate transporter. In another embodiment, the gene is glutamine ABC transporter. In another embodiment, the gene is major facilitator family transporter. In another embodiment, the gene is glycine betaine/L-proline ABC transporter. In another embodiment, the gene is molybdenum ABC transporter. In another embodiment, the gene is techoic acid ABC transporter. In another embodiment, the gene is cobalt ABC transporter.
  • the gene is ammonium transporter. In another embodiment, the gene is amino acid ABC transporter. In another embodiment, the gene is cell division ABC transporter. In another embodiment, the gene is manganese ABC transporter. In another embodiment, the gene is iron compound ABC transporter. In another embodiment, the gene is maltose/maltodextrin ABC transporter. In another embodiment, the gene is drug resistance transporter of the BcrlCflA family. In another embodiment, the gene is a subunit of one of the above proteins.
  • nucleic acid molecule that is used to transform the Listeria in order to arrive at a recombinant Listeria.
  • the nucleic acid disclosed herein used to transform Listeria lacks a virulence gene.
  • the nucleic acid molecule is integrated into the Listeria genome and carries a non- functional virulence gene.
  • the virulence gene is mutated in the recombinant Listeria.
  • the nucleic acid molecule is used to inactivate the endogenous gene present in the Listeria genome.
  • the virulence gene is an actA gene, an inlA gene, and MB gene, an inlC gene, inlJ gene, a plbC gene, a bs/z gene, or a /?r 3 ⁇ 4 gene. It is to be understood by a skilled artisan, that the virulence gene can be any gene known in the art to be associated with virulence in the recombinant Listeria.
  • a live attenuated Listeria disclosed herein is a recombinant Listeria.
  • a recombinant Listeria disclosed herein comprises a mutation of a genomic internalin C (MB) gene.
  • the recombinant Listeria comprises a mutation or a deletion of a genomic actA gene and a genomic internalin B gene.
  • translocation of Listeria to adjacent cells is inhibited by the deletion of the actA gene and/or the inlB gene, which are involved in the process, thereby resulting in unexpectedly high levels of attenuation with increased immunogenicity and utility as a strain backbone.
  • the term "attenuation,” may encompass a diminution in the ability of the bacterium to cause disease in an animal.
  • the pathogenic characteristics of the attenuated Listeria strain have been lessened compared with wild-type Listeria, although the attenuated Listeria is capable of growth and maintenance in culture.
  • the lethal dose at which 50% of inoculated animals survive is preferably increased above the LD 50 of wild-type Listeria by at least about 10-fold, more preferably by at least about 100-fold, more preferably at least about 1,000 fold, even more preferably at least about 10,000 fold, and most preferably at least about 100,000-fold.
  • An attenuated strain of Listeria is thus one which does not kill an animal to which it is administered, or is one which kills the animal only when the number of bacteria administered is vastly greater than the number of wild type non-attenuated bacteria which would be required to kill the same animal.
  • An attenuated bacterium should also be construed to mean one which is incapable of replication in the general environment because the nutrient required for its growth is not present therein. Thus, the bacterium is limited to replication in a controlled environment wherein the required nutrient is provided.
  • the attenuated strains disclosed herein are therefore environmentally safe in that they are incapable of uncontrolled replication.
  • a Listeria strain disclosed herein is an inlA mutant, an inlB mutant, an inlC mutant, an inlJ mutant, prfA mutant, actA mutant, a dal/dat mutant, a prfA mutant, a plcB deletion mutant, or a double mutant lacking both plcA and plcB.
  • the Listeria comprises a deletion or mutation of these genes individually or in combination.
  • the Listeria disclosed herein lack each one of genes.
  • the Listeria disclosed herein lack at least one and up to ten of any gene provided herein, including the act A, prfA, and dal/dat genes.
  • the prfA mutant is a D133V prfA mutant.
  • the metabolic gene, the virulence gene, etc. is lacking in a chromosome of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is lacking in the chromosome and in any episomal genetic element of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is lacking in the genome of the virulence strain. In one embodiment, the virulence gene is mutated in the chromosome. In another embodiment, the virulence gene is deleted from the chromosome. In another embodiment, the metabolic gene, the virulence gene, etc. is mutated in a chromosome of the Listeria strain.
  • the metabolic gene, virulence gene, etc. is mutated in the chromosome and in any episomal genetic element of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is mutated in the genome of the virulence strain. In another embodiment, the virulence gene is deleted from the chromosome.
  • a recombinant Listeria strain disclosed herein is attenuated.
  • the recombinant Listeria lacks the actA virulence gene.
  • the recombinant Listeria lacks the prfA virulence gene.
  • the recombinant Listeria lacks the inlB gene.
  • the recombinant Listeria lacks both, the actA and inlB genes.
  • the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA gene.
  • the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous inlB gene.
  • the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous inlC gene. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA and inlB genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA, MB, and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA, inlB, and inlC genes.
  • the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA, MB, and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation in any single gene or combination of the following genes: actA, dal, dat, MB, inlC, prfA, plcA, plcB.
  • mutants include any type of mutation or modification to the sequence (nucleic acid or amino acid sequence), and includes a deletion mutation, a truncation, an inactivation, a disruption, or a translocation. These types of mutations are readily known in the art.
  • auxotrophic bacteria comprising a plasmid encoding a metabolic enzyme or a complementing gene provided herein
  • transformed auxotrophic bacteria are grown on a media that will select for expression of the amino acid metabolism gene or the complementing gene.
  • a bacteria auxotrophic for D-glutamic acid synthesis is transformed with a plasmid comprising a gene for D-glutamic acid synthesis, and the auxotrophic bacteria will grow in the absence of D-glutamic acid, whereas auxotrophic bacteria that have not been transformed with the plasmid, or are not expressing the plasmid encoding a protein for D-glutamic acid synthesis, will not grow.
  • a bacterium auxotrophic for D- alanine synthesis will grow in the absence of D-alanine when transformed and expressing the plasmid disclosed herein if the plasmid comprises an isolated nucleic acid encoding an amino acid metabolism enzyme for D-alanine synthesis.
  • Such methods for making appropriate media comprising or lacking necessary growth factors, supplements, amino acids, vitamins, antibiotics, and the like are well known in the art, and are available commercially (Becton-Dickinson, Franklin Lakes, NJ).
  • the bacteria are propagated in the presence of a selective pressure. Such propagation comprises growing the bacteria in media without the auxotrophic factor.
  • the presence of the plasmid expressing an amino acid metabolism enzyme in the auxotrophic bacteria ensures that the plasmid will replicate along with the bacteria, thus continually selecting for bacteria harboring the plasmid.
  • the skilled artisan when equipped with the present disclosure and methods herein will be readily able to scale-up the production of the Listeria vaccine vector by adjusting the volume of the media in which the auxotrophic bacteria comprising the plasmid are growing.
  • auxotroph strains and complementation systems are adopted for the use with the methods and compositions disclosed herein.
  • a recombinant Listeria strain disclosed herein expresses a recombinant polypeptide.
  • a recombinant Listeria strain comprises a plasmid that encodes a recombinant polypeptide.
  • a recombinant nucleic acid disclosed herein is in a plasmid in the recombinant Listeria strain provided herein.
  • the plasmid is an episomal plasmid that does not integrate into the recombinant Listeria strain's chromosome.
  • the plasmid is an integrative plasmid that integrates into the Listeria strain's chromosome.
  • the plasmid is a multicopy plasmid.
  • a recombinant Listeria strain disclosed herein comprises a nucleic acid molecule encoding a fusion protein comprising a tumor associated antigen.
  • a tumor associated antigen comprises a survivin polypeptide or a fragment thereof.
  • a survivin polypeptide comprises a human survivin.
  • a survivin polypeptide is a human survivin.
  • a survivin polypeptide comprises a mouse survivin.
  • the recombinant Listeria strain disclosed herein comprises a nucleic acid molecule encoding a fusion protein comprising a survivin protein or a fragment thereof.
  • survivin or an antigenic portion thereof comprises a tumor specific antigen.
  • the human survivin protein comprises the amino sequence as set forth in SEQ ID NO: 17 GAPTLPPAWQPFLKDHRISTFKNWPF
  • the survivin protein is a homologue of SEQ ID No: 17. In another embodiment, the survivin protein is a variant of SEQ ID No: 17. In another embodiment, the survivin protein is a fragment of SEQ ID No: 17.
  • the mouse survivin protein comprises the amino sequence as set forth in SEQ ID NO: 18 MGAPALPQIWQLYLKNYRIATFKNWPF
  • the survivin protein is a homologue of SEQ ID No: 18. In another embodiment, the survivin protein is a variant of SEQ ID No: 17. In another embodiment, the survivin protein is a fragment of SEQ ID No: 18.
  • a survivin fragment disclosed herein consists of about residues 1- 25. In another embodiment, the survivin fragment consists of about residues 1-50. In another embodiment, the survivin fragment consists of about residues 1-75. In another embodiment, the survivin fragment consists of about residues 1-100. In another embodiment, the survivin fragment consists of about residues 1-125. In another embodiment, the survivin fragment consists of full length survivin. In another embodiment, the survivin fragment is lacking a signal sequence.
  • a survivin disclosed herein fragment contains residues of a homologous survivin protein that correspond to one of the above AA ranges.
  • the residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous survivin protein has an insertion or deletion, relative to an survivin protein utilized herein, then the residue numbers can be adjusted accordingly.
  • Other survivin fragments are known in the art.
  • a homologous survivin has identity to a survivin sequence (e.g. to one of SEQ ID No: 17 or SEQ ID No: 18) of greater than 70%.
  • a homologous survivin refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 72%.
  • a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 75%.
  • a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 78%.
  • a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 80%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 82%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 83%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 85%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 87%.
  • a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 88%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 90%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 92%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 93%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 95%.
  • a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 96%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 97%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 98%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 99%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of 100%.
  • the human survivin protein is encoded by a nucleotide molecule having the sequence SEQ ID NO: 19: ggtgccccgacgttgcccctgcctggcagccctttctc aaggaccaccgcatctctacattcaagaactggcccttcttggagggctgcgccccggagcggatggccgaggctggcttcatccact gccccactgagaacgagccagacttggcccagtgtttctctgcttcaaggagctggaaggctgggagccagatgacgaccccatagaggaac ataaaaagcattcgtccggttgcgctttcttgtcaagaagcagtttgaagaatta
  • the survivin protein is encoded by a homologue of SEQ ID No: 19. In another embodiment, the survivin protein is encoded by a variant of SEQ ID No: 19. In another embodiment, the survivin protein is encoded by an isomer of SEQ ID No: 19. In another embodiment, the survivin protein is encoded by a fragment of SEQ ID No : 19.
  • the survivin protein is a mouse survivin protein is encoded by a nucleotide molecule having the sequence SEQ ID NO: 20: tcgagggagctccggcg ctgccccagatctggcagctgtacctcaagaactaccgcatcgccaccttcaagaactggcccttcctggaggactgcgcctgcgccccagagc gaatggcggaggctggcttcatccactgccctaccgagaacgagcctgatttggcccagtgtttttttctgcttttaaggaattggaaggctgggaac cccgatgacaacccgatagaggagcatagaaagcactcccctggctgcgcttcctcactgtcaagaagcagatggaagaactagcactccctggct
  • the survivin protein is encoded by a homologue of SEQ ID No: 20.
  • the survivin protein is encoded by a variant of SEQ ID No: 20.
  • the survivin protein is encoded by an isomer of SEQ ID No: 20.
  • the survivin protein is encoded by a fragment of SEQ ID No: 20.
  • the survivin protein of the methods and compositions disclosed herein is a human survivin protein (SEQ ID NO: 19).
  • the survivin protein is a mouse survivin protein (SEQ ID NO: 20).
  • the survivin protein is a rat survivin protein.
  • the survivin protein is a primate survivin protein.
  • the survivin protein is a canine survivin protein.
  • the survivin protein is a survivin protein of human or other animal species or combinations thereof known in the art.
  • a nucleic acid sequence encoding a canine survivin protein is set forth in SEQ ID NO: 21:
  • nucleic acid encoding a canine survivin protein is set forth in GenBank Accession No. AB095108.1.
  • a nucleic acid encoding a survivin protein is a homolog of SEQ ID NO: 21.
  • nucleic acid sequence is a variant of SEQ ID NO: 21.
  • an amino acid sequence encoding a canine survivin protein is set forth in SEQ ID NO: 22:
  • nucleic acid encoding a canine survivin protein is set forth in GenBank Accession No. BAC22748.2.
  • an amino acid sequence encoding a survivin protein is a homolog of SEQ ID NO: 22.
  • amino acid sequence is a variant of SEQ ID NO: 22.
  • an amino acid sequence of survivin disclosed herein has 99% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 95% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 90% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 85% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 80% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 75% identity with that of human survivin amino acid sequence.
  • an amino acid sequence of survivin disclosed herein has 99% identity with that of mouse survivin amino acid sequence.
  • the amino acid sequence of survivin has 95% identity with that of mouse survivin amino acid sequence.
  • the amino acid sequence of survivin has 90% identity with that of mouse survivin amino acid sequence.
  • the amino acid sequence of survivin has 85% identity with that of mouse survivin amino acid sequence.
  • the amino acid sequence of survivin has 80% identity with that of mouse survivin amino acid sequence.
  • the amino acid sequence of survivin has 75% identity with that of mouse survivin amino acid sequence.
  • an amino acid sequence of survivin disclosed herein has 99% identity with that of a canine survivin amino acid sequence.
  • the amino acid sequence of survivin has 95% identity with that of canine survivin amino acid sequence.
  • the amino acid sequence of survivin has 90% identity with that of canine survivin amino acid sequence.
  • the amino acid sequence of survivin has 85% identity with that of canine survivin amino acid sequence.
  • the amino acid sequence of survivin has 80% identity with that of canine survivin amino acid sequence.
  • the amino acid sequence of survivin has 75% identity with that of canine survivin amino acid sequence.
  • a survivin protein that is the source of a survivin antigen for use in the methods and compositions as disclosed herein is a Baculoviral IAP Repeat Containing 5 Protein.
  • the survivin protein is API4 protein.
  • the survivin protein is EPR-1 protein.
  • the survivin protein is Apoptosis Inhibitor 4 protein.
  • the survivin protein is IAP4 protein.
  • the survivin protein is Survivin Variant 3 Alpha.
  • a survivin protein or fragment thereof of the methods and compositions disclosed herein does not include a signal sequence thereof. In another embodiment, omission of the signal sequence enables the survivin fragment to be successfully secreted in Listeria.
  • nucleic acids encoding the recombinant polypeptides disclosed herein also encode a signal peptide or sequence.
  • the fusion protein of methods and compositions disclosed herein comprises an LLO signal sequence.
  • a heterologous antigen may be expressed in Listeria through the use of a signal sequence, such as a Listerial signal sequence, for example, the hemolysin signal sequence or the ActA signal sequence.
  • foreign genes can be expressed downstream from a L. monocytogenes promoter without creating a fusion protein.
  • the signal peptide is bacterial (Listerial or non-Listerial).
  • the signal peptide is native to the bacterium.
  • the signal peptide is foreign to the bacterium.
  • the signal peptide is a signal peptide from Listeria monocytogenes, such as a secAl signal peptide.
  • the signal peptide is an Usp45 signal peptide from Lactococcus lactis, or a Protective Antigen signal peptide from Bacillus anthracis.
  • the signal peptide is a secA2 signal peptide, such the p60 signal peptide from Listeria monocytogenes.
  • the recombinant nucleic acid molecule optionally comprises a third polynucleotide sequence encoding p60, or a fragment thereof.
  • the signal peptide is a Tat signal peptide, such as a B. subtilis Tat signal peptide (e.g., PhoD).
  • the signal peptide is in the same translational reading frame encoding the recombinant polypeptide.
  • the survivin protein is a splice variant 1 survivin protein. In another embodiment, the survivin protein is a splice variant 2 survivin protein. In another embodiment, the survivin protein is a splice variant 3 survivin protein. In another embodiment, the survivin protein is a transcript variant 1 survivin protein. In another embodiment, the survivin protein is a transcript variant 2 survivin protein. In another embodiment, the survivin protein is a transcript variant 3 survivin protein. In another embodiment, the survivin protein is a transcript variant 4 survivin protein. In another embodiment, the survivin protein is a transcript variant 5 survivin protein. In another embodiment, the survivin protein is a transcript variant 6 survivin protein. In another embodiment, the survivin protein is a transcript variant 7 survivin protein.
  • the survivin protein that is the source of a survivin peptide of methods and compositions as disclosed herein is a human survivin protein.
  • the survivin protein is a primate survivin protein.
  • the survivin protein is a canine survivin protein.
  • Survivin proteins of other species are known in the art.
  • a sequence useful in the composition and methods as disclosed herein may be a homologue of a particular LLO sequence or N-terminal fragment thereof.
  • a sequence useful in the composition and methods as disclosed herein may be a homologue of an antigenic polypeptide, which in one embodiment, is survivin or a functional fragment thereof.
  • a homolog of a polypeptide and, in one embodiment, the nucleic acid encoding such a homolog, disclosed herein maintains the functional characteristics of the parent polypeptide.
  • a homolog of an antigenic polypeptide disclosed herein maintains the antigenic characteristic of the parent polypeptide.
  • a sequence useful in the composition and methods as disclosed herein may be a homologue of any sequence described herein.
  • a homologue shares at least 70% identity with a particular sequence.
  • a homologue shares at least 72% identity with i a . particular sequence. In another embodiment, a homologue shares at least 75% identity with a particular sequence. In another embodiment, a homologue shares at least 78% identity with a particular sequence. In another embodiment, a homologue shares at least 80% identity with a particular sequence. In another embodiment, a homologue shares at least 82% identity with a particular sequence. In another embodiment, a homologue shares at least 83% identity with a particular sequence. In another embodiment, a homologue shares at least 85% identity with a particular sequence. In another embodiment, a homologue shares at least 87% identity with a particular sequence. In another embodiment, a homologue shares at least 88% identity with a particular sequence.
  • a homologue shares at least 90% identity with a particular sequence. In another embodiment, a homologue shares at : least 90.5% ) identity with a particular sequence. In another embodiment, a homologue shares at least 91% identity with a particular sequence. In another embodiment, a homologue shares at least 92% identity with a particular sequence. In another embodiment, a homologue shares at least 93% identity with a particular sequence. In another embodiment, a homologue shares at least 95% identity with a particular sequence. In another embodiment, a homologue shares at least 96% identity with a particular sequence. In another embodiment, a homologue shares at least 97% identity with a particular sequence. In another embodiment, a homologue shares at least 98% identity with a particular sequence. In another embodiment, a homologue shares at least 99% identity with a particular sequence. In another embodiment, a homologue shares 100% identity with a particular sequence.
  • antigenic polypeptide may encompass a polypeptide, peptide or recombinant peptide as described hereinabove that is foreign to a host and leads to the mounting of an immune response when present in, or, in some embodiments, detected by, a host.
  • an "antigenic polypeptide” may encompass a polypeptide, a peptide, a recombinant polypeptide, or a recombinant peptide as described herein that is processed and presented on MHC class I and/or class II molecules present in a subject's cells leading to the mounting of an immune response when present in, or, in another embodiment, detected by, the host.
  • the antigen may be foreign to the host.
  • the antigen might be present in the host but the host does not elicit an immune response against it because of immunologic tolerance.
  • a recombinant Listeria strain of the methods and compositions as disclosed herein comprise a nucleic acid molecule operably integrated into the Listeria genome as an open reading frame with an endogenous ActA sequence.
  • a recombinant Listeria strain of the methods and compositions disclosed herein comprise an episomal expression vector comprising a nucleic acid molecule encoding fusion protein comprising an antigen fused to an ActA or a truncated ActA.
  • the expression and secretion of the antigen is under the control of an ActA promoter and ActA signal sequence and it is expressed as fusion to 1- 233 amino acids of ActA (truncated ActA or tActA).
  • the truncated ActA consists of the first 390 amino acids of the wild type ActA protein as described in US Patent Serial No. 7,655,238, which is incorporated by reference herein in its entirety.
  • the truncated ActA is an ActA-NlOO or a modified version thereof (referred to as ActA-NlOO*) in which a PEST motif has been deleted and containing the nonconservative QDNKR substitution as described in US Patent Publication Serial No. 2014/0186387.
  • an attenuated auxotrophic Listeria vaccine strain is based on a Listeria vaccine vector which is attenuated due to the deletion of virulence gene actA and retains the plasmid for survivin expression in vivo and in vitro by complementation of dal gene.
  • the Listeria strain expresses and secretes a survivin protein fused to the first 441 amino acids of listeriolysin O (LLO).
  • LLO listeriolysin O
  • the Listeria is a dal/dat/actA Listeria having a mutation in the dal, dat and actA endogenous genes.
  • the mutation is a deletion, a truncation or an inactivation of the mutated genes.
  • the expression and secretion of the fusion protein LLO-survivin from the attenuated auxotrophic strain disclosed herein that expresses a survivin antigen/LLO fusion protein after an in vitro passage is comparable to that of the Lm-LLO- survivin in TCA precipitated cell culture supernatants after 8 hours of in vitro growth.
  • the dal/dat/actA mutant Listeria strain is highly attenuated and has a better safety profile than previous Listeria vaccine generation, as it is more rapidly cleared from the spleens of the immunized mice.
  • the dal/dat/actA mutant Listeria strain results in a longer delay of tumor onset in transgenic animals than Listeria vaccines based on more virulent antibiotic resistant strains (see US Publication No. 2011/0142791, which is incorporated by reference herein in its entirety).
  • the dal/dat/actA mutant Listeria strain causes a significant decrease in intra-tumoral T regulatory cells (Tregs).
  • the lower frequency of Tregs in tumors treated with LmddA vaccines result in an increased intratumoral CD8/Tregs ratio, suggesting that a more favorable tumor microenvironment can be obtained after immunization with LmddA vaccines.
  • the present invention provides a recombinant polypeptide comprising an N-terminal fragment of an LLO protein fused to a survivin protein or to a fragment thereof. In one embodiment, the present invention provides a recombinant polypeptide consisting of an N- terminal fragment of an LLO protein fused to a survivin protein or fused to a fragment thereof. [00187] In one embodiment, the present invention provides a recombinant polypeptide comprising an N-terminal fragment of an ActA protein fused to a survivin protein or to a fragment thereof. In another embodiment, the present invention provides a recombinant polypeptide consisting of an N- terminal fragment of an ActA protein fused to a survivin protein or fused to a fragment thereof.
  • a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are fused directly to one another.
  • the genes encoding a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are fused directly to one another.
  • a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are operably attached via a linker peptide.
  • a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are attached via a heterologous peptide.
  • a PEST-containing polypeptide or peptide is N-terminal to the survivin antigen or immunogenic fragment thereof.
  • a PEST-containing polypeptide or peptide is expressed and used alone, i.e., in unfused form.
  • a PEST-containing polypeptide or peptide is the N-terminal-most portion of the fusion protein.
  • a PEST- containing polypeptide or peptide is an N-terminal LLO protein fragment.
  • a truncated LLO is truncated at the C-terminal to arrive at an N-terminal LLO.
  • a PEST-containing polypeptide or peptide is the tLLO.
  • a PEST-containing polypeptide or peptide is an ActA polypeptide or a fragment thereof. In another embodiment, a PEST-containing polypeptide or peptide is an N-terminal fragment of ActA. In another embodiment, a PEST-containing amino acid sequence is a PEST peptide.
  • the present invention provides a recombinant polypeptide comprising survivin or a fragment thereof fused to a PEST amino acid sequence.
  • a recombinant polypeptide comprises survivin or a fragment thereof fused to 1-2 PEST amino acid sequences.
  • a recombinant polypeptide comprises survivin or a fragment thereof fused to 2-3 PEST amino acid sequences.
  • a recombinant polypeptide comprises survivin or a fragment thereof fused to 3-4 PEST amino acid sequences.
  • polypeptide may encompass, in another embodiment, a peptide or polypeptide of any length.
  • a peptide or recombinant peptide as disclosed herein has one of the lengths enumerated above for an HMW-MAA fragment.
  • peptide refers to native peptides (either degradation products, synthetically synthesized peptides or recombinant peptides) and/or peptidomimetics (typically, synthetically synthesized peptides), such as peptoids and semipeptoids which are peptide analogs, which may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells.
  • Methods for preparing peptidomimetic compounds are well known in the art and are specified, for example, in Quantitative Drug Design, C.A. Ramsden Gd., Chapter 17.2, F. Choplin Pergamon Press (1992), which is incorporated by reference as if fully set forth herein. Further details in this respect are provided hereinunder.
  • Trp, Tyr and Phe may be substituted for synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
  • the peptides disclosed herein may also include one or more modified amino acids or one or more non-amino acid monomers (e.g. fatty acids, complex carbohydrates etc).
  • 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.
  • a construct or nucleic acid molecule disclosed herein 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 virus recombinants expressing either Listeria monocytogenes partial listeriolysin or Brucella abortus ribosomal L7/L12 protein. Vet Microbiol 2005, 109(1-2): 11-7); and Jiang LL, Song HH, et al., (Characterization of a mutant Listeria monocytogenes strain expressing green fluorescent protein.
  • recombination site or "site- specific recombination site” may encompass a sequence of bases in a nucleic acid molecule that is recognized by a recombinase (along with associated proteins, in some cases) that mediates exchange or excision of the nucleic acid segments flanking the recombination sites.
  • the recombinases and associated proteins are collectively referred to as “recombination proteins” see, e.g., Landy, A., (Current Opinion in Genetics & Development) 3:699-707; 1993).
  • a construct or nucleic acid molecule disclosed herein is integrated into the Listerial chromosome using transposon insertion.
  • Techniques for transposon 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 nucleic acid molecule 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 Bacterid 2002; 184(15): 4177-86).
  • an integrase gene and attachment site of a bacteriophage e.g. U153 or PSA listeriophage
  • 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).
  • 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.
  • the present invention further comprises a phage based chromosomal integration system for clinical applications, where a host strain that is auxotrophic for essential enzymes, including, but not limited to, d-alanine racemase can be used, for example Lm dal ⁇ -)dat ⁇ -).
  • a phage integration system based on PSA is used (Lauer, et al., 2002 J Bacterid, 184:4177-4186).
  • the current invention enables the establishment of a phage based chromosomal integration system that does not require selection with antibiotics. Instead, an auxotrophic host strain can be complemented.
  • phage expression vector or “phagemid” may encompass any phage-based recombinant expression system for the purpose of expressing a nucleic acid sequence of the methods and compositions as disclosed herein in vitro or in vivo, constitutively or inducibly, in any cell, including prokaryotic, yeast, fungal, plant, insect or mammalian cell.
  • a phage expression vector typically can both reproduce in a bacterial cell and, under proper conditions, produce phage particles.
  • the term includes linear or circular expression systems and encompasses both phage-based expression vectors that remain episomal or integrate into the host cell genome.
  • conjugation is used to introduce genetic material and/or plasmids into bacteria.
  • Methods for conjugation are well known in the art, and are described, for example, in Nikodinovic J et al (A second generation snp-derived Escherichia coli-Streptomyces shuttle expression vector that is generally transferable by conjugation. Plasmid. 2006 Nov;56(3):223-7) and Auchtung JM et al (Regulation of a Bacillus subtilis mobile genetic element by intercellular signaling and the global DNA damage response. Proc Natl Acad Sci U S A. 2005 Aug 30; 102(35): 12554-9).
  • Antibiotic resistance genes are used in the conventional selection and cloning processes commonly employed in molecular biology and vaccine preparation.
  • Antibiotic resistance genes contemplated in the present invention include, but are not limited to, gene products that confer resistance to ampicillin, penicillin, methicillin, streptomycin, erythromycin, kanamycin, tetracycline, cloramphenicol (CAT), neomycin, hygromycin, gentamicin and others well known in the art.
  • Plasmids and other expression vectors useful in the present invention are described elsewhere herein, and can include such features as a promoter/regulatory sequence, an origin of replication for gram negative and gram positive bacteria, an isolated nucleic acid encoding a fusion protein and an isolated nucleic acid encoding an amino acid metabolism gene. Further, an isolated nucleic acid encoding a fusion protein and an amino acid metabolism gene will have a promoter suitable for driving expression of such an isolated nucleic acid.
  • Promoters useful for driving expression in a bacterial system include bacteriophage lambda, the bla promoter of the beta-lactamase gene of pBR322, and the CAT promoter of the chloramphenicol acetyl transferase gene of pBR325.
  • prokaryotic promoters include the major right and left promoters of 5 bacteriophage lambda (PL and PR), the trp, recA, lacZ, lad, and gal promoters of E. coli, the alpha-amylase (Ulmanen et al, 1985. J. Bacterid. 162:176-182) and the S28-specific promoters of B.
  • subtilis (Gilman et al, 1984 Gene 32: 11- 20), the promoters of the bacteriophages of Bacillus (Gryczan, 1982, In: The Molecular Biology of the Bacilli, Academic Press, Inc., New York), and Streptomyces promoters (Ward et al, 1986, Mol. Gen. Genet. 203:468- 478). Additional prokaryotic promoters contemplated in the present invention are reviewed in, for example, Glick (1987, J. Ind. Microbiol. 1:277-282); Cenatiempo, (1986, Biochimie, 68:505-516); and Gottesman, (1984, Ann. Rev. Genet. 18:415-442).
  • promoter/regulatory elements contemplated in the present invention include, but are not limited to the Listerial prfA promoter, the Listerial hly promoter, the Listerial p60 promoter and the Listerial actA promoter (GenBank Acc. No. NC_003210) or fragments thereof.
  • a plasmid of methods and compositions disclosed herein comprises a gene encoding a fusion protein.
  • subsequences are cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments are then, in another embodiment, ligated to produce the desired DNA sequence.
  • DNA encoding the antigen is produced using DNA amplification methods, for example polymerase chain reaction (PCR), as discussed below
  • DNA encoding the fusion protein or the recombinant protein disclosed herein is cloned using DNA amplification methods such as polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • the gene for non-hemolytic LLO is PCR amplified, using a sense primer comprising a suitable restriction site and an antisense primer comprising another restriction site, e.g. a non-identical restriction site to facilitate cloning.
  • a sense primer comprising a suitable restriction site
  • an antisense primer comprising another restriction site, e.g. a non-identical restriction site to facilitate cloning.
  • Ligation of the non-hemolytic LLO and antigen sequences and insertion into a plasmid or vector produces a vector encoding non-hemolytic LLO joined to a terminus of the antigen.
  • the two molecules are joined either directly or by a short spacer introduced by the restriction site.
  • Fusion proteins comprising the survivin antigen or immunogenic fragment thereof may be prepared by any suitable method, including, for example, cloning and restriction of appropriate sequences or direct chemical synthesis. Alternatively, subsequences may be cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments may then be ligated to produce the desired DNA sequence.
  • DNA encoding the antigen can be produced using DNA amplification methods, for example polymerase chain reaction (PCR). First, the segments of the native DNA on either side of the new terminus are amplified separately.
  • the 5' end of the one amplified sequence encodes the peptide linker, while the 3' end of the other amplified sequence also encodes the peptide linker. Since the 5' end of the first fragment is complementary to the 3' end of the second fragment, the two fragments (after partial purification, e.g. on LMP agarose) can be used as an overlapping template in a third PCR reaction.
  • the amplified sequence will contain codons, the segment on the carboxy side of the opening site (now forming the amino sequence), the linker, and the sequence on the amino side of the opening site (now forming the carboxyl sequence).
  • the antigen is ligated into a plasmid.
  • a nucleic acid sequences encoding the fusion or recombinant proteins are transformed into a variety of host cells, including E. coli, other bacterial hosts, such as Listeria, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cells lines and myeloma cell lines.
  • the recombinant fusion protein gene will be operably linked to appropriate expression control sequences for each host.
  • Promoter/ regulatory sequences are described in detail elsewhere herein.
  • the plasmid further comprises additional promoter regulatory elements, as well as a ribosome binding site and a transcription termination signal.
  • control sequences will include a promoter and an enhancer derived from e.g. immunoglobulin genes, SV40, cytomegalovirus, etc., and a polyadenylation sequence.
  • sequences include splice donor and acceptor sequences.
  • “Stably maintained” refers to maintenance of a nucleic acid molecule or plasmid in the absence of selection (e.g. antibiotic selection) for 10 generations, without detectable loss.
  • the period is 15 generations.
  • the period is 20 generations.
  • the period is 25 generations.
  • the period is 30 generations.
  • the period is 40 generations.
  • the period is 50 generations.
  • the period is 60 generations.
  • the period is 80 generations.
  • the period is 100 generations.
  • the period is 150 generations.
  • the period is 200 generations.
  • the period is 300 generations.
  • the period is 500 generations. In another embodiment, the period is more than 500 generations.
  • the nucleic acid molecule or plasmid is maintained stably in vitro (e.g. in culture). In another embodiment, the nucleic acid molecule or plasmid is maintained stably in vivo. In another embodiment, the nucleic acid molecule or plasmid is maintained stably both in vitro and in vitro.
  • the recombinant Listeria strain of methods and compositions disclosed herein is, in another embodiment, a recombinant Listeria monocytogenes strain. In another embodiment, the Listeria strain is a recombinant Listeria seeligeri strain. In another embodiment, the Listeria strain is a recombinant Listeria grayi strain.
  • the Listeria strain is a recombinant Listeria ivanovii strain. In another embodiment, the Listeria strain is a recombinant Listeria murrayi strain. In another embodiment, the Listeria strain is a recombinant Listeria welshimeri strain. In another embodiment, the Listeria strain is a recombinant strain of another Listeria species.
  • Attenuated Listeria strains such as Lm ddta-actA mutant (Brundage et al, 1993, Proc. Natl. Acad. Sci., USA, 90: 11890-11894), L. monocytogenes delta-plcA (Camilli et al, 1991, J. Exp. Med., 173:751-754), or delta-actA, ddta-inlB (Brockstedt et 5 al, 2004, PNAS, 101:13832-13837) are used in the present invention.
  • Attenuated Listeria strains are constructed by introducing one or more attenuating mutations, as will be understood by one of average skill in the art when equipped with the disclosure herein.
  • examples of such strains include, but are not limited to Listeria strains auxotrophic for aromatic amino acids (Alexander et al, 1993, Infection and Immunity 10 61:2245-2248) and mutant for the formation of lipoteichoic acids (Abachin et al, 2002, Mol. Microbiol. 43:1-14) and those attenuated by a lack of a virulence gene (see examples herein).
  • a recombinant Listeria strain disclosed herein has been passaged through an animal host.
  • the passaging maximizes efficacy of the strain as a vaccine vector.
  • the passaging stabilizes the immunogenicity of the Listeria strain.
  • the passaging stabilizes the virulence of the Listeria strain.
  • the passaging increases the immunogenicity of the Listeria strain.
  • the passaging increases the virulence of the Listeria strain.
  • the passaging removes unstable sub-strains of the Listeria strain.
  • the passaging reduces the prevalence of unstable sub-strains of the Listeria strain.
  • the Listeria strain contains a genomic insertion of the gene encoding the antigen-containing recombinant peptide.
  • the Listeria strain carries a plasmid comprising the gene encoding the antigen-containing recombinant peptide.
  • the passaging is performed as described herein. In another embodiment, the passaging is performed by other methods known in the art.
  • a recombinant nucleic acid disclosed herein is operably linked to a promoter/regulatory sequence that drives expression of the encoded peptide in the Listeria strain.
  • Promoter/regulatory sequences useful for driving constitutive expression of a gene are well known in the art and include, but are not limited to, for example, the Phi y A, VactA, and p60 promoters of Listeria, the Streptococcus bac promoter, the Streptomyces griseus sgiA promoter, and the B. thuringiensis phaZ promoter.
  • inducible and tissue specific expression of the nucleic acid encoding a peptide disclosed herein is accomplished by placing the nucleic acid encoding the peptide under the control of an inducible or tissue specific promoter/regulatory sequence.
  • tissue specific or inducible promoter/regulatory sequences which are useful for his purpose include, but are not limited to the MMTV LTR inducible promoter, and the SV40 late enhancer/promoter.
  • a promoter that is induced in response to inducing agents such as metals, glucocorticoids, and the like, is utilized.
  • the invention includes the use of any promoter/regulatory sequence, which is either known or unknown, and which is capable of driving expression of the desired protein operably linked thereto.
  • heterologous encompasses a nucleic acid, amino acid, peptide, polypeptide, or protein derived from a different species than the reference species.
  • a Listeria strain expressing a heterologous polypeptide in one embodiment, would express a polypeptide that is not native or endogenous to the Listeria strain, or in another embodiment, a polypeptide that is not normally expressed by the Listeria strain, or in another embodiment, a polypeptide from a source other than the Listeria strain.
  • heterologous may be used to describe something derived from a different organism within the same species.
  • the heterologous antigen is expressed by a recombinant strain of Listeria, and is processed and presented to cytotoxic T-cells upon infection of mammalian cells by the recombinant strain.
  • the heterologous antigen expressed by Listeria species need not precisely match the corresponding unmodified antigen or protein in the tumor cell or infectious agent so long as it results in a T-cell response that recognizes the unmodified antigen or protein which is naturally expressed in the mammal.
  • the term heterologous antigen may be referred to herein as "antigenic polypeptide", “heterologous protein”, “heterologous protein antigen”, “protein antigen”, “antigen”, and the like.
  • a heterologous antigen disclosed herein is a survivin polypeptide or antigenic portion thereof.
  • the two molecules of the fusion 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.
  • the two molecules of the protein (for example, the LLO fragment and the antigen) are synthesized separately or unfused.
  • the two molecules of the protein are synthesized separately from the same nucleic acid.
  • the two molecules are individually synthesized from separate nucleic acids.
  • the present invention provides a composition comprising an antagonist of immunosuppressive signaling molecules.
  • immunosuppressive signaling molecules mediate resistance of tumors to a host' s immune response.
  • an immunosuppressive signaling molecule comprises a CD28/CTLA-4 receptor superfamily.
  • an immunosuppressive signaling molecule comprises a B7 receptor family.
  • an immunosuppressive molecule comprises PD- 1. In another embodiment, an immunosuppressive molecule comprises PD-L1. In another embodiment, an immunosuppressive molecule comprises PD-L2. In another embodiment, an immunosuppressive molecule comprises B7- 1.
  • an antagonist disclosed herein is any chemical compound or biological molecule that blocks interaction of an immunosuppressive molecules with these molecules' receptors.
  • an antagonist is a small molecule.
  • the antagonist is an antibody.
  • the antagonist is a monoclonal antibody (mAb).
  • the antagonist is a polyclonal antibody.
  • the antagonist is an antigen-binding antibody fragment.
  • a composition disclosed herein comprises an antagonist antibody.
  • antibody may encompass any form of immunoglobulin molecule that exhibits the desired biological or binding activity.
  • monoclonal antibodies including full length monoclonal antibodies
  • polyclonal antibodies include full length monoclonal antibodies
  • multispecific antibodies e.g. , bispecific antibodies
  • humanized human antibodies
  • chimeric antibodies camelized single domain antibodies.
  • Parental antibodies are antibodies obtained by exposure of an immune system to an antigen prior to modification of the antibodies for an intended use, such as humanization of an antibody for use as a human therapeutic.
  • the term "antibody” encompasses not only intact polyclonal or monoclonal antibodies, but also, unless otherwise specified, any antigen binding portion thereof that competes with the intact antibody for specific binding, fusion proteins comprising an antigen binding portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site.
  • Antigen binding portions include, for example, Fab, Fab', F(ab') 2 , Fd, Fv, domain antibodies (dAbs, e.g., shark and camelid antibodies), fragments including complementarity determining regions (CDRs), single chain variable fragment antibodies (scFv), maxibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.
  • An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG 2 , IgG 3 , IgG 4 , IgAi and IgA 2 .
  • the heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. [00220] It will be appreciated by a skilled artisan that the term "antibody fragment" or
  • antibody binding fragment may encompass to antigen binding fragments of antibodies, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full- length antibody, e.g. fragments that retain one or more CDR regions.
  • antibody binding fragments include, but are not limited to, Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and multispecific antibodies formed from antibody fragments.
  • CDR complementarity determining region(s) in an immunoglobulin variable region, defined using the Kabat numbering system, unless otherwise indicated.
  • Kabat as used herein means an immunoglobulin alignment and numbering system pioneered by Elvin A. Kabat ((1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.).
  • Monoclonal antibody or
  • mAb or “Mab” may encompass a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts.
  • conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • a monoclonal antibody to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the monoclonal antibody may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731.
  • a mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region.
  • the human constant region is selected from the group consisting of IgGl, IgG2, IgG3 and IgG4 constant regions, and in some embodiments, the human constant region is an IgGl or IgG4 constant region.
  • the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab') 2 , scFv and Fv fragments.
  • Chimeric antibody may encompass an antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • a particular species e.g., human
  • another species e.g., mouse
  • Human antibody may encompass an antibody that comprises human immunoglobulin protein sequences only.
  • a human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell.
  • mouse antibody or rat antibody refer to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
  • Humanized antibody may encompass forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • an immunosuppressive antagonist molecule disclosed herein comprises PD-1 antagonists.
  • PD-1 antagonists interact with PD-1 and block binding of human PD-L1 expressed on a cancer cell to human PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also block binding of human PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1.
  • Alternative names or synonyms for PD-1 include: PDCD1, PD1, CD279 and SLEB2.
  • Exemplary human PD-1 amino acid sequences can be found in NCBI Locus No.: NP_005009.
  • Examples of mAbs that bind to human PD-1, and useful in the treatment method, medicaments and uses of the present invention, are described in US7488802, US7521051, US8008449, US8354509, US8168757, WO2004/004771, WO2004/072286, WO2004/056875, and US2011/0271358.
  • Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses disclosed herein include:, nivolumab (BMS-936558), a human IgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No.
  • the immunosuppressive antagonists molecules disclosed herein comprise PD-L1 antagonists.
  • PD-L1 antagonists interact with PD-L1 and block binding of human PD-1 expressed on an immune cell (T cell, B cell or NKT cell) to human PD-L1 expressed on a cancer cell.
  • Alternative names or synonyms for PD-L1 include: PDCD1L1, PDL1, B7H1, B7-4, CD274 and B7-H.
  • Exemplary human PD-L1 amino acid sequence can be found in NCBI Locus No.: NP_054862.
  • variable regions may encompass the segment of IgG chains which is variable in sequence between different antibodies. It extends to Kabat residue 109 in the light chain and 113 in the heavy chain.
  • a "variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • the variable regions of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope.
  • CDRs complementarity determining regions
  • both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al; National Institutes of Health, Bethesda, Md. ; 5 th ed.; NIH Publ. No. 91-3242 (1991); Kabat (1978) Adv. Prot. Chem. 32: 1-75; Kabat, et al, (1977) J. Biol. Chem. 252:6609-6616; Chothia, et al, (1987) J Mol. Biol. 196:901- 917 or Chothia, et al, (1989) Nature 342:878-883.
  • hypervariable region may encompass the amino acid residues of an antibody that are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from a "complementarity determining region" or "CDR" ⁇ i.e. CDRL1, CDRL2 and CDRL3 in the light chain variable domain and CDRH1, CDRH2 and CDRH3 in the heavy chain variable domain).
  • CDR complementarity determining region
  • FR framework residues refers to variable domain residues other than the hypervariable region residues defined herein as CDR residues.
  • FR may encompass the immunoglobulin variable regions excluding the CDR regions.
  • antagonists comprise PD-L2 antagonists.
  • PD-L2 antagonists interact with PD-L2 and block binding of human PD-1 expressed on an immune cell (T cell, B cell or NKT cell) to human PD-L2 expressed on a cancer cell.
  • Alternative names or synonyms for PD-L2 include: PDCD1L2, PDL2, B7-DC, Btdc and CD273.
  • Exemplary human PD-L2 amino acid sequences can be found in NCBI Locus No.: NP_079515.
  • antagonists comprise B7-1 antagonists.
  • PD-1 antagonists useful in any of the treatment methods, medicaments and uses disclosed herein include an immunoadhesin that specifically binds to human PD-1 or human PD-Ll, e.g. a fusion protein containing the extracellular or PD-1 binding portion of PD-Ll or PD-L2 fused to a constant region such as an Fc region of an immunoglobulin molecule.
  • immunoadhesion molecules that specifically bind to PD-1 are described in WO2010/027827 and WO2011/066342.
  • AMP-224 also known as B7-DCIg
  • B7-DCIg a PD-L2-FC fusion protein and binds to human PD-1.
  • other PD-1/PD-L1 binding antagonists include a small molecule or a biomolecule that blocks or competes with critical binding or signaling of PD-1 and its ligand or signaling to the effector cell after this binding.
  • an antibody that "specifically binds to" a specified target protein may encompass an antibody that exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity.
  • An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g. without producing undesired results such as false positives.
  • Antibodies or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins.
  • an antibody is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human PD-1, human PD-L1, human PD-L2, or human B7-1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human PD-1, human PD-L1, human PD-L2, or human B7-1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • the PD- 1 antagonist is a monoclonal antibody, or an antigen binding fragment thereof.
  • the PD-1 antagonist is a monoclonal antibody, or antigen binding fragment thereof, which specifically binds to human PD- 1 and comprises (a) a heavy chain variable region, and (b) a light chain variable.
  • the PD-1 antagonist is a monoclonal antibody which specifically binds to human PD-1.
  • the PD-1 antagonist is a monoclonal antibody which specifically binds to human PD-1.
  • PD-L1 expression or "PD-L2" expression as used herein means any detectable level of expression of the designated PD-L protein on the cell surface or of the designated PD-L mRNA within a cell or tissue.
  • PD-L protein expression may be detected with a diagnostic PD-L antibody in an IHC assay of a tumor tissue section or by flow cytometry.
  • PD-L protein expression by tumor cells may be detected by PET imaging, using a binding agent (e.g., antibody fragment, antibody and the like) that specifically binds to the desired PD-L target, e.g., PD-L1 or PD-L2.
  • a binding agent e.g., antibody fragment, antibody and the like
  • One approach employs a simple binary end-point of positive or negative for PD- LI expression, with a positive result defined in terms of the percentage of tumor cells that exhibit histologic evidence of cell-surface membrane staining.
  • a tumor tissue section is counted as positive for PD-Ll expression if IHC staining is observed in at least 1%, and preferably 5% of total tumor cells.
  • a pancreatic tumor sample is designated as having weak PD-Ll expression if 1% to 49% of the total tumor cells in the sample exhibit membrane staining and is designated as having strong PD-Ll expression if at least 50% of the tumor cells in the sample exhibit membrane staining, in each case as determined by IHC assay using the antibody 22C3 described in WO2014/100079.
  • PD-Ll expression in the tumor tissue section is quantified in the tumor cells as well as in infiltrating immune cells, which predominantly comprise lymphocytes.
  • the percentage of tumor cells and infiltrating immune cells that exhibit membrane staining are separately quantified as ⁇ 5%, 5 to 9%, and then in 10% increments up to 100%.
  • PD-Ll expression is counted as negative if the score is ⁇ 5% score and positive if the score is > 5%.
  • PD-Ll expression in the immune infiltrate is reported as a semi-quantitative measurement called the adjusted inflammation score (AIS), which is determined by multiplying the percent of membrane staining cells by the intensity of the infiltrate, which is graded as none (0), mild (score of 1, rare lymphocytes), moderate (score of 2, focal infiltration of tumor by lymphohistiocytic aggregates), or severe (score of 3, diffuse infiltration).
  • AIS adjusted inflammation score
  • a tumor tissue section is counted as positive for PD-Ll expression by immune infiltrates if the AIS is > 5.
  • the level of PD-L mRNA expression may be compared to the mRNA expression levels of one or more reference genes that are frequently used in quantitative RT-PCR, such as ubiquitin C.
  • a level of PD-Ll expression (protein and/or mRNA) by malignant cells and/or by infiltrating immune cells within a tumor is determined to be "overexpressed” or “elevated” based on comparison with the level of PD-Ll expression (protein and/ or mRNA) by an appropriate control.
  • a control PD-Ll protein or mRNA expression level may be the level quantified in nonmalignant cells of the same type or in a section from a matched normal tissue.
  • PD-Ll expression in a tumor sample is determined to be elevated if PD-Ll protein (and/or PD-Ll mRNA) in the sample is at least 10%, 20%, or 30% greater than in the control.
  • compositions comprising a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to an additional polypeptide, and further comprising a PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and wherein administering the composition to a subject having a survivin-expressing cancer treats, ameliorates, or induces regression of the cancer.
  • a pharmaceutical composition comprising a nucleic acid encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to an additional polypeptide and further comprising a PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist.
  • a pharmaceutical composition comprising a recombinant Listeria vaccine strain comprising a nucleic acid encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to an additional polypeptide, the composition further comprising a PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist.
  • the additional polypeptide is a PEST- containing peptide or polypeptide.
  • the additional polypeptide is an N- terminal Listeriolysin O (LLO) fragment thereof.
  • the additional polypeptide is an N-terminal ActA fragment thereof.
  • the additional polypeptide is a PEST-containing amino acid sequence.
  • treatment may encompass contacting of an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human, subject, cell, tissue, organ, or biological fluid.
  • Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell.
  • Administration and “treatment” also may encompass in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell.
  • subject includes any organism, preferably an animal, more preferably a mammal ⁇ e.g., rat, mouse, dog, cat, rabbit) and most preferably a human.
  • composition may encompass a therapeutically effective amount of the active ingredient or ingredients comprising the Listeria strain, an immunosuppressive molecule antagonist, or both together with a pharmaceutically acceptable carrier or diluent.
  • the terms "therapeutically effective amount”, in reference to the treatment of tumor may encompass an amount capable of invoking one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, slowing down and complete growth arrest; (2) reduction in the number of tumor cells; (3) reduction in tumor size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of tumor cell infiltration into peripheral organs; (5) inhibition (i.e., reduction, slowing down or complete stopping) of metastasis; (6) enhancement of anti-tumor immune response, which may, but does not have to, result in the regression or rejection of the tumor; and/or (7) relief, to some extent, of one or more symptoms associated with the disorder.
  • a "therapeutically effective amount" of a vaccine disclosed herein for purposes of treatment of tumor may be determined empirically and in a routine manner.
  • compositions may be used interchangeably. It is also to be understood that administration of such compositions enhances an immune response, or increase a T effector cell to regulatory T cell ratio or elicit an anti-tumor immune response, as further provided herein.
  • a nucleic acid molecule of the anti-tumor composition disclosed herein further comprises a second open reading frame encoding a metabolic enzyme, wherein the metabolic enzyme complements a mutation in the chromosome of the recombinant Listeria strain.
  • an immune response elicited by the methods and compositions disclosed herein comprises an immune response to at least one 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 at least one subdominant epitope.
  • the only measurable component of the immune response is an immune response to at least one subdominant epitope.
  • the immune response against the survivin-expressing cancer comprises an immune response to at least one dominant epitope of the survivin protein.
  • a composition disclosed herein comprises a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprises recombinant Listeria strain comprising a nucleic acid molecule, wherein the nucleic acid molecule comprises a first open reading frame encoding a polypeptide, wherein the polypeptide comprises a PEST-containing peptide or polypeptide fused to a heterologous antigen, wherein the PEST-containing peptide or polypeptide comprises an N-terminal fragment of Listeriolysin O (LLO), an N-terminal fragment of ActA, or a PEST-containing amino acid sequence, and wherein the heterologous antigen comprises a survivin antigen or an immunogenic fragment thereof.
  • LLO Listeriolysin O
  • administration of compositions disclosed herein increase the number of antigen- specific T cells.
  • administration of compositions activates co- stimulatory receptors on T cells.
  • administration of compositions induces proliferation of memory and/or effector T cells.
  • administration of compositions increases proliferation of T cells.
  • administration of compositions disclosed herein negate tumor immunosuppressive signaling.
  • administration of compositions suppresses PD-1 mediated immunosuppressive signaling.
  • administration of compositions counters tumor mediated attenuation of T-cell activation.
  • administration of compositions neutralizes tumor's immune surveillance evasion mechanisms.
  • administration of compositions disclosed herein increase the number of active antigen-specific T cells.
  • administration of compositions enhances activation of co- stimulatory receptors on T cells.
  • administration of compositions induces proliferation of memory and/or effector T cells.
  • administration of compositions increases proliferation of T cells.
  • compositions disclosed herein may be used in methods disclosed hereinin order to elicit an enhanced anti-tumor T cell response in a subject, in order to inhibit tumor-mediated immunosuppression in a subject, or for increasing the ratio or T effector cells to regulatory T cells (Tregs) in the spleen and tumor of a subject, or any combination thereof.
  • Tregs regulatory T cells
  • a composition comprising a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and a recombinant attenuated a Listeria strain described hereinabove, further comprises an adjuvant.
  • the adjuvant utilized in methods and compositions disclosed herein is, in another embodiment, a granulocyte/macrophage colony- stimulating factor (GM-CSF) protein.
  • the adjuvant comprises a GM-CSF protein.
  • the adjuvant is a nucleotide molecule encoding GM-CSF.
  • the adjuvant comprises a nucleotide molecule encoding GM-CSF.
  • the adjuvant is saponin QS21. In another embodiment, the adjuvant comprises saponin QS21. In another embodiment, the adjuvant is monophosphoryl lipid A. In another embodiment, the adjuvant comprises monophosphoryl lipid A. In another embodiment, the adjuvant is SBAS2. In another embodiment, the adjuvant comprises SBAS2. In another embodiment, the adjuvant is an unmethylated CpG-containing oligonucleotide. In another embodiment, the adjuvant comprises an unmethylated CpG-containing oligonucleotide. In another embodiment, the adjuvant is an immune- stimulating cytokine. In another embodiment, the adjuvant comprises an immune- stimulating cytokine.
  • the adjuvant is a nucleotide molecule encoding an immune- stimulating cytokine. In another embodiment, the adjuvant comprises a nucleotide molecule encoding an immune- stimulating cytokine. In another embodiment, the adjuvant is or comprises a quill glycoside. In another embodiment, the adjuvant is or comprises a bacterial mitogen. In another embodiment, the adjuvant is or comprises a bacterial toxin. Other adjuvants are known in the art.
  • oligonucleotide is interchangeable with the term “nucleic acid”, and may encompass a molecule, which may include, but is not limited to, prokaryotic sequences, eukaryotic mRNA, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic (e.g., mammalian) DNA, and even synthetic DNA sequences.
  • the term may encompass sequences that include any of the known base analogs of DNA and RNA.
  • the present invention provides a composition comprising a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria strain expressing the antigen.
  • the antigen is survivin or an antigenic portion thereof.
  • the PD-1 antagonist is an anti-PD-1 antibody or an antigen binding fragment thereof.
  • PD-L1 antagonist is an anti-PD-Ll antibody or an antigen binding fragment thereof.
  • PD-L2 antagonist is an anti-PD-L2 antibody or an antigen binding fragment thereof.
  • B7-1 antagonist is an anti-B7-l antibody or an antigen binding fragment thereof.
  • the present invention provides immunogenic compositions comprising PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising recombinant Listeria provided herein.
  • the present invention provides pharmaceutical compositions comprising a PD- 1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising recombinant Listeria provided herein.
  • the fusion protein comprises the survivin antigen or immunogenic fragment thereof and an additional polypeptide.
  • the additional polypeptide disclosed herein is an N-terminal Listeriolysin O (LLO) fragment.
  • the additional polypeptide is an N-terminal ActA fragment.
  • the additional polypeptide is a PEST-containing amino acid sequence.
  • an additional polypeptide or an additional adjuvant polypeptide augments antigen presentation and immunity in a similar fashion to LLO.
  • PD-L1 antagonist is an anti-PD-Ll antibody or an antigen binding fragment thereof.
  • PD-L2 antagonist is an anti-PD-L2 antibody or an antigen binding fragment thereof.
  • B7-1 antagonist is an anti-B7-l antibody or an antigen binding fragment thereof.
  • the PD-1 antagonist blocks interaction of PD-1 with any of its ligands.
  • the PD-1 antagonist blocks the interaction of PD-1 with PD-Ll, PD-L2, or both.
  • the PD-Ll antagonist blocks the interaction of PD-Ll with PD-1, B7-1, or both.
  • the PD-L2 antagonist blocks the interaction of PD-L2 with PD-1.
  • the B7-1 antagonist blocks the interaction of B7-1 with PD-Ll.
  • compositions comprising PD-1 antagonist, PD- Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria of the present invention.
  • compositions comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-lantagonist comprising a recombinant attenuated Listeria expressing a fusion polypeptide of the present invention.
  • the composition further comprises formulation buffer suitable for administration to a subject.
  • a pharmaceutical composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria of the present invention.
  • a composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist comprising a recombinant attenuated Listeria expressing a fusion polypeptide of the present invention.
  • the pharmaceutical composition further comprises formulation buffer suitable for administration to a subject.
  • an immunogenic composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria of the present invention.
  • a composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist comprising a recombinant attenuated Listeria expressing a fusion polypeptide of the present invention.
  • said immunogenic composition further comprises formulation buffer suitable for administration to a subject.
  • the combination therapy may also comprise one or more additional therapeutic agents.
  • the additional therapeutic agent may be, e.g., a chemo therapeutic, a biotherapeutic agent (including but not limited to antibodies to VEGF, VEGFR, EGFR, Her2/neu, other growth factor receptors, CD20, CD40, CD-40L, CTLA-4, OX-40, GITR, 4- IBB, and ICOS), an immunogenic agent (for example, attenuated cancerous cells, tumor antigens, antigen presenting cells such as dendritic cells pulsed with tumor derived antigen or nucleic acids, immune stimulating cytokines (for example, IL-2, IFNa2, GM-CSF), and cells transfected with genes encoding immune stimulating cytokines such as but not limited to GM-CSF).
  • a chemo therapeutic including but not limited to antibodies to VEGF, VEGFR, EGFR, Her2/neu, other growth factor receptors, CD20, CD40, CD-40L, CTLA-4, OX
  • Bio therapeutic agent may encompass a biological molecule, such as an antibody or fusion protein, that blocks ligand / receptor signaling in any biological pathway that supports tumor maintenance and/or growth or suppresses the anti-tumor immune response.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (
  • calicheamicin especially calicheamicin gammall and calicheamicin phill, see, e.g., Agnew, Chem. Intl. Ed. Engl., 33: 183- 186 (1994); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino- doxorubicin,
  • paclitaxel and doxetaxel paclitaxel and doxetaxel; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • platinum analogs such as cisplatin and carboplatin
  • vinblastine platinum
  • anti-hormonal agents that act to regulate or inhibit hormone action on tumors
  • SERMs selective estrogen receptor modulators
  • aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate, exemestane, formestane, fadrozole, vorozole, letrozole, and anastrozole
  • anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin
  • pharmaceutically acceptable salts, acids or derivatives of any of the above such as anti- estrogens and selective estrogen receptor modulators (SER
  • chemotherapeutic agent may encompass a chemical or biological substance that can cause death of cancer cells, or interfere with growth, division, repair, and/or function of cancer cells.
  • Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytoxic/antitumor antibiotics, topisomerase inhibitors, photosensitizers, anti-estrogens and selective estrogen receptor modulators (SERMs), anti- progesterones,estrogen receptor down-regulators (ERDs), estrogen receptor antagonists, leutinizing hormone-releasing hormone agonists, anti-androgens, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, anti-sense oligonucleotides that inhibit expression of genes implicated in abnormal cell proliferation or tumor growth.
  • Chemotherapeutic agents useful in the treatment methods disclosed herein include
  • Each therapeutic agent in a combination therapy of the invention may be administered either alone or in a medicament (also referred to herein as a pharmaceutical composition) which comprises the therapeutic agent and one or more pharmaceutically acceptable carriers, excipients and diluents, according to standard pharmaceutical practice.
  • the term "pharmaceutically acceptable carrier” may encompass any inactive substance that is suitable for use in a formulation for the administration to a human of a PD-1 antagonist and/or a live-attenuated Listeria strain that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells or a live-attenuated Listeria monocytogenes strain bioengineered, by transforming it with an expression vector to express a survivin antigen or immunogenic fragment thereof fused to a tLLO or ADXS31-265 ⁇ 10403S da ] dat ('] actA ('] pADV265) strain.
  • Transforming may encompass engineering a bacterial cell to take up a plasmid or other heterologous DNA molecule.
  • transforming refers to engineering a bacterial cell to express a gene of a plasmid or other heterologous DNA molecule.
  • a method of treating a tumor or cancer in a subject comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration treats a tumor or a cancer in said subject.
  • the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer.
  • an immunogenic composition as described above, for treating cancer.
  • an immunogenic composition used in a method disclosed herein comprises a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprises a Listeria strain expressing a fusion polypeptide as described throughout, wherein the fusion polypeptide comprises a survivin antigen.
  • the term “treating” may encompass curing a disease. In another embodiment, “treating” may encompass preventing a disease. In another embodiment, “treating” may encompass reducing the incidence of a disease. In another embodiment, “treating” may encompass ameliorating symptoms of a disease. In another embodiment, “treating” may encompass increasing performance free survival or overall survival of a patient. In another embodiment, “treating” may encompass stabilizing the progression of a disease. In another embodiment, “treating” may encompass inducing remission. In another embodiment, “treating” may encompass slowing the progression of a disease. The terms “reducing”, “suppressing” and “inhibiting” refer to lessening or decreasing.
  • treating may encompass both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or lessen the targeted pathologic condition or disorder as described herein.
  • treating may include directly affecting or curing, suppressing, inhibiting, preventing, reducing the severity of, delaying the onset of, reducing symptoms associated with the disease, disorder or condition, or a combination thereof.
  • "treating" may encompass inter alia delaying progression, expediting remission, inducing remission, augmenting remission, speeding recovery, increasing efficacy of or decreasing resistance to alternative therapeutics, or a combination thereof.
  • preventing or “impeding” may encompass, inter alia, delaying the onset of symptoms, preventing relapse to a disease, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, or a combination thereof.
  • “suppressing” or “inhibiting” may encompass, inter alia, reducing the severity of symptoms, reducing the severity of an acute episode, reducing the number of symptoms, reducing the incidence of disease-related symptoms, reducing the latency of symptoms, ameliorating symptoms, reducing secondary symptoms, reducing secondary infections, prolonging patient survival, or a combination thereof.
  • symptoms are primary, while in another embodiment, symptoms are secondary.
  • primary refers to a symptom that is a direct result of a particular disease or disorder
  • secondary refers to a symptom that is derived from or consequent to a primary cause.
  • the compounds for use in the present invention treat primary or secondary symptoms or secondary complications.
  • symptoms may be any manifestation of a disease or pathological condition.
  • a tumor or cancer being treated is a survivin-expressing tumor or survivin-expressing cancer.
  • the cancer being treated is breast cancer, a central nervous system (CNS) cancer, a head and neck cancer, an osteosarcoma (OSA), a canine osteosarcoma (OSA), or Ewing's sarcoma (ES).
  • the cancer is pancreatic cancer.
  • the cancer is ovarian cancer.
  • the cancer is gastric cancer.
  • the cancer is a carcinomatous lesion of the pancreas.
  • the cancer is pulmonary adenocarcinoma.
  • the cancer is colorectal adenocarcinoma.
  • 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 a CNS 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 mesothelioma. In another embodiment, the cancer is malignant mesothelioma (MM).
  • MM malignant mesothelioma
  • the cancer is a melanoma. In another embodiment, the cancer is a glioma. In another embodiment, the cancer is a germ cell tumor. In another embodiment, the cancer is a choriocarcinoma. In another embodiment, the cancer is a lymphoma, leukemia, myeloma or any survivin-expressing cancer known in the art.
  • the cancer is a pancreatic carcinoma. In another embodiment, the cancer is pancreatic ductal carcinoma. In another embodiment, the cancer is acinar cell carcinoma of the pancreas, or cystadenocarcinoma. In another embodiment, the cancer is pancreatic neuroendocrine tumor. In another embodiment, the cancer is insulinoma or gastrinoma. In another embodiment, the cancer is any prostate carcinoma known in the art.
  • the cancer is refractory. In another embodiment, the cancer is advanced. In another embodiment, the cancer is a metastasis. In another embodiment, the cancer has metastasized. In another embodiment, the metastasis of pancreartic cancer, which in one embodiment, comprises metastasis to bone, and in another embodiment, comprises metastasis to other organs. In another embodiment, the cancer is any survivin-expressing pancreatic cancer. In another embodiment, a cancer or solid tumor is a result of relapse or metastatic disease. [00283] In another embodiment, cells of a tumor that is targeted by the methods and compositions disclosed herein express surviving or a fragment thereof. In another embodiment, the tumor is associated with survivin.
  • survivin expressed by a tumor cell is the target of the immune responses induced by methods and compositions of the present invention.
  • cells of the tumor that is targeted by methods and compositions disclosed herein express low levels of MHC.
  • cells of the tumor that is targeted by methods and compositions disclosed herein express high levels of PD-L1.
  • PD-L1 suppresses or attenuates the T effector cell response to survivin.
  • the PD-L1 mediated immunosuppression is neutralized by methods and compositions disclosed herein.
  • cancer or tumors may be prevented in specific populations known to be susceptible to a particular cancer or tumor.
  • susceptibilty may be due to environmental factors, such as smoking, which in one embodiment, may cause a population to be subject to lung cancer, while in another embodiment, such susceptbility may be due to genetic factors, for example a population with BRCAl/2 mutations may be susceptible, in one embodiment, to breast cancer, and in another embodiment, to ovarian cancer.
  • one or more mutations on chromosome 8q24, chromosome 17ql2, and chromosome 17q24.3 may increase susceptibility to pancreatic cancer, as is known in the art.
  • Other genetic and environmental factors contributing to cancer susceptibility are known in the art.
  • the methods disclosed herein induce the expansion of T effector cells in peripheral lymphoid organs leading to an enhanced presence of T effector cells at the tumor site.
  • the methods disclosed herein induce the expansion of T effector cells in peripheral lymphoid organs leading to an enhanced presence of T effector cells at the periphery.
  • Such expansion of T effector cells leads to an increased ratio of T effector cells to regulatory T cells in the periphery and at the tumor site without affecting the number of Tregs.
  • peripheral lymphoid organs include, but are not limited to, the spleen, peyer's patches, the lymph nodes, the adenoids, etc.
  • the increased ratio of T effector cells to regulatory T cells occurs in the periphery without affecting the number of Tregs. In another embodiment, the increased ratio of T effector cells to regulatory T cells occurs in the periphery, the lymphoid organs and at the tumor site without affecting the number of Tregs at these sites. In another embodiment, the increased ratio of T effector cells decreases the frequency of Tregs, but not the total number of Tregs at these sites. [00286]
  • an administration of an immunogenic compositions disclosed herein stimulates the anti-tumor activity of T effector cells. In another embodiment, the administration of an immunogenic compositions disclosed herein negates tumor-mediated immunosuppression of anti-survivin T effector cell response. In yet another embodiment, the administration of an immunogenic composition disclosed herein neutralizes tumor-mediated immunosuppressive signaling. In another embodiment, the administration of an immunogenic compositions disclosed herein desensitizes T effector cells to tumor-mediated immunosuppression.
  • a method for treating a survivin-expressing cancer in a subject comprising administering to the subject a combination therapy which comprises a PD-1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or a B7-1 antagonist and further comprising a live-attenuated Listeria strain that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells.
  • a method of treating a survivin-expressing cancer in a subject comprising the step of administering to the subject a combination therapy which comprises a PD- 1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or a B7-1 antagonist and further comprising a recombinant Listeria comprising a nucleic acid encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to a PEST-containing peptide or polypeptide.
  • a method for treating a survivin-expressing cancer in a subject comprising administering to the subject a combination therapy which comprises a PD-1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or a B7-1 antagonist and further comprises a live-attenuated Listeria monocytogenes strain comprising an expression vector encoding a survivin antigen or immunogenic fragment thereof fused to a tLLO.
  • a method for treating a cancer in an individual comprising administering to the individual a combination therapy which comprises a PD-1 antagonist, a PD-L1 antagonist, a PD- L2 antagonist or a B7-1 antagonist and further comprising an ADXS-31-265 (10403S actA ⁇ ' ⁇ pADV265) strain.
  • a method of preventing a tumor or cancer in a subject comprising the step of administering to the subject a combination therapy provided herein.
  • the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer.
  • a method of preventing a survivin-expressing tumor growth in a subject the method comprising the step of administering to the subject a combination therapy.
  • the administration of a survivin- expressing Listeria in the context of a fusion protein with a truncated LLO elicits an immune response against other tumor-associated antigens as a result of epitope spreading.
  • disclosed herein is a method of treating a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy disclosed herein.
  • a method of delaying the progression of a survivin- expressing cancer in a subject comprising the step of administering to the subject a combination therapy provided herein.
  • disclosed herein is a method of delaying the progression of a survivin- expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination disclosed herein.
  • disclosed herein is a method of prolonging the survival of a subject having a survivin-expressing cancer in a subject, the method comprising the step of administering to the subject a combination disclosed herein.
  • disclosed herein is a method of prolonging the survival of a subject having a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy disclosed herein.
  • disclosed herein is a method of inhibiting, impeding, or delaying metastatic disease in a subject having a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy disclosed herein.
  • disclosed herein is a method of inducing an anti-survivin immune response in a subject, the method comprising the step of administering to the subject a combination therapy provided herein.
  • a method of augmenting an anti-survivin immune response in a subject the method comprising the step of administering to the subject a combination therapy provided herein.
  • a method of preventing an escape mutation in the treatment of survivin over-expressing cancers the method comprising the step of administering to the subject a combination therapy provided herein.
  • a method of inducing regression of a survivin antigen-expressing cancer in a subject comprising the step of administering to the subject a combination therapy provided herein.
  • disclosed herein is a method of decreasing the frequency of intra- tumoral T regulatory cells, the method comprising the step of administering to the subject a combination therapy provided herein.
  • a method of treating a metastatic disease coming from a survivin-expressing tumor in a subject the method comprising the step of administering to the subject a combination therapy provided herein.
  • a method of breaking tolerance in a subject to a survivin-expressing tumor or cancer in the subject comprising the step of administering to the subject a combination therapy provided herein.
  • a method of impeding growth of a survivin- expressing cancer in a subject comprising the step of administering to the subject a combination therapy provided herein.
  • the methods disclosed herein of inducing an anti-survivin immune response allow treating a survivin-expressing tumor in a subject.
  • the methods provided herein, of inducing an anti-survivin immune response allow prolonging survival of a subject suffering from a survivin-expressing cancer.
  • the methods disclosed herein of inducing an anti-survivin immune response induce regression of a tumor or cancer in a subject suffering from a survivin-expressing cancer.
  • the methods disclosed herein of inducing an anti-survivin immune response break tolerance to survivin in a subject suffering from a survivin-expressing tumor.
  • a method of treating further comprises administering said combination therapy following a relapse or metastasis in the subject.
  • any composition comprising an immunosuppressive molecule antagonist and a Listeria strain described herein may be used in the methods of this invention.
  • Compositions comprising an immunosuppressive molecule antagonist Listeria strains have been described in detail above.
  • the present invention provides a method for "epitope spreading" of an anti-tumor response.
  • the immunization using the compositions and methods disclosed herein induce epitope spreading onto other tumors bearing antigens other than the antigen carried in the vaccine of the present invention.
  • 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.
  • a method of treating, suppressing, or inhibiting a cancer or a tumor growth in a subject by epitope spreading wherein and in another embodiment, the cancer is associated with expression of an antigen or fragment thereof comprised in the composition of the present invention.
  • the method comprises administering to the subject a combination therapy of the present invention.
  • the subject mounts an immune response against the antigen-expressing cancer or the antigen-expressing tumor, thereby treating, suppressing, or inhibiting a cancer or a tumor growth in a subject.
  • a method of increasing a ratio of T effector cells to regulatory T cells (Tregs) in the spleen and tumor microenvironments of a subject comprising administering the immunogenic composition provided herein.
  • increasing a ratio of T effector cells to regulatory T cells (Tregs) in the spleen and tumor microenvironments in a subject allows for a more profound anti-tumor response in the subject.
  • the T effector cells comprise CD4+FoxP3- T cells. In another embodiment, the T effector cells are CD4+FoxP3- T cells. In another embodiment, the T effector cells comprise CD4+FoxP3- T cells and CD8+ T cells. In another embodiment, the T effector cells are CD4+FoxP3- T cells and CD8+ T cells. In another embodiment, the regulatory T cells is a CD4+FoxP3+ T cell.
  • the present invention provides methods of treating, protecting against, and inducing an immune response against a tumor or a cancer, comprising the step of administering to a subject the combination therapy provided herein.
  • the present invention provides a method of preventing or treating a tumor or cancer in a human subject, comprising the step of administering to the subject the combination therapy strain provided herein, the recombinant Listeria strain comprising a recombinant polypeptide comprising an N-terminal fragment of an LLO protein and tumor- associated antigen, whereby the recombinant Listeria strain induces an immune response against the tumor-associated antigen, thereby treating a tumor or cancer in a human subject.
  • the immune response is a T-cell response.
  • the T-cell response is a CD4+FoxP3- T cell response.
  • the T-cell response is a CD8+ T cell response.
  • the T-cell response is a CD4+FoxP3- and CD8+ T cell response.
  • the present invention provides a method of protecting a subject against a tumor or cancer, comprising the step of administering to the subject the combination therapy provided herein.
  • the present invention provides a method of inducing regression of a tumor in a subject, comprising the step of administering to the subject the combination therapy provided herein.
  • the present invention provides a method of reducing the incidence or relapse of a tumor or cancer, comprising the step of administering to the subject the combination therapy provided herein.
  • the present invention provides a method of suppressing the formation of a tumor in a subject, comprising the step of administering to the subject the combination therapy provided herein.
  • the present invention provides a method of inducing a remission of a cancer in a subject, comprising the step of administering to the subject the combination therapy provided herein.
  • the present invention provides a method of extending a remission of a tumor or cancer in a subject, comprising the step of administering to the subject the combination therapy provided herein.
  • the present invention provides a method of reducing the size of a tumor a subject, comprising the step of administering to the subject the combination therapy provided herein.
  • the nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide is integrated into the Listeria genome.
  • the nucleic acid is in a plasmid in the recombinant Listeria vaccine strain.
  • the nucleic acid molecule is in a bacterial artificial chromosome in the recombinant Listeria vaccine strain.
  • a method of treating reduces tumor size. Reduction of tumor size may be partial or by 100%. In another embodiment, methods disclosed herein reduce tumor size by 90%. In another embodiment, methods disclosed herein reduce tumor size by 80%. In another embodiment, methods reduce tumor size by 70%. In another embodiment, methods reduce tumor size by 60%. In another embodiment, methods reduce tumor size by 50%.
  • a method of treating increases the time to disease progression.
  • the time to disease progression was increased by at least 2 moths as compared to untreated subject.
  • the time to disease progression was increased by at least 4 moths as compared to untreated subject.
  • the time to disease progression was increased by at least 6 moths as compared to untreated subject.
  • the time to disease progression was increased by at least 1 year as compared to untreated subject.
  • the time to disease progression was increased by at least 2 years as compared to untreated subject.
  • the time to disease progression was increased by at least 3 years as compared to untreated subject.
  • the time to disease progression was increased by at least 4 years as compared to untreated subject.
  • the time to disease progression was increased by at least 5 years as compared to untreated subject.
  • the present invention provides a method of impeding a growth of a survivin-expressing cancer in a subject, comprising administering to the subject a combination therapy provided herein, wherein the recombinant polypeptide comprising an N- terminal fragment of a LLO protein fused to a survivin antigen, and wherein the antigen has one or more subdominant CD8+ T cell epitopes.
  • the antigen does not contain any of the dominant CD8+ T cell epitopes.
  • 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 or 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 CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 45% of the antigen- specific CD8+ 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 refers to an epitope recognized by over 60% of the antigen- specific CD8+ 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 term 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.
  • the term refers to an epitope recognized by over 80% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 85% of the antigen- specific CD 8+ T cells. In another embodiment, the term refers to an epitope recognized by over 90% of the antigen- specific CD8+ 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 CD8+ T cells.
  • Subdominant CD8+ T cell epitope refers to an epitope recognized by fewer than 30% of the antigen- specific CD8+ 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 CD8+ 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.
  • 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 over 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 over 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.
  • 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 term refers to an epitope recognized by fewer than 6% of the antigen- specific CD8+ 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 CD8+ T cells.
  • the term refers to an epitope recognized by fewer than 2% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 1% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 0.5% of the antigen- specific CD8+ T cells.
  • vaccination with recombinant Listeria expressing the LLO-antigen fusions disclosed herein induces epitope spreading.
  • vaccination with LLO- antigen fusions, even outside the context of survivin, induces epitope spreading as well.
  • 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.
  • the antigen in methods and compositions disclosed herein is, in one embodiment, expressed at a detectable level on a non-tumor 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.
  • the non-tumor cell is a germ cell.
  • Detectable level refers, in one embodiment, to a level that is detectable when using a standard assay.
  • the assay is an immunological assay.
  • the assay is enzyme-linked immunoassay (ELISA).
  • the assay is Western blot.
  • the assay is FACS.
  • the assay is Western blot.
  • the assay is PCR. It is to be understood by a skilled artisan that other assays available in the art can be used in the methods provided herein.
  • 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.
  • the present invention provides a method for inducing formation of cytotoxic T cells in a host having cancer, comprising administering to the host a combination therapy of the present invention, thereby inducing formation of cytotoxic T cells in a host having cancer.
  • disclosed herein is a method of administering a composition of the present invention. In another embodiment, disclosed herein is a method of administering a vaccine of the present invention. In another embodiment, disclosed herein is a method of administering a recombinant polypeptide or recombinant nucleotide of the present invention. In another embodiment, the step of administering a composition, recombinant polypeptide or recombinant nucleotide disclosed herein is performed with an attenuated recombinant form of Listeria comprising a recombinant nucleotide or expressing a recombinant polypeptide, each in its own discrete embodiment.
  • the administering is performed with a different attenuated bacterial vector.
  • the administering is performed with a DNA vaccine (e.g. a naked DNA vaccine).
  • administration of a recombinant polypeptide disclosed herein is performed by producing the protein recombinantly, then administering the recombinant protein to a subject.
  • a 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, and then is transferred to the subject.
  • Each therapeutic agent in a combination therapy of the invention may be administered simultaneously (i.e., in the same medicament), concurrently (i.e., in separate medicaments administered one right after the other in any order) or sequentially in any order.
  • Sequential administration is particularly useful when the therapeutic agents in the combination therapy are in different dosage forms (one agent is a tablet or capsule and another agent is a sterile liquid) and/or are administered on different dosing schedules, e.g., a chemotherapeutic that is administered at least daily and a biotherapeutic that is administered less frequently, such as once weekly, once every two weeks, or once every three weeks.
  • the term "simultaneous administration" in relation to the administration of medicaments may encompass the administration of medicaments such that the individual medicaments are delivered to a subject at the same time, through a same or different routes of administration.
  • Dosage units for an immunosuppressive molecule antagonist may be expressed as a flat dose, i.e., 100 mg, 200 mg, 300 mg, or as a patient-specific dose, i.e., mg/kg (mg therapeutic agent/kg of body weight) or mg/m (quantity in milligrams per square meter of body surface area).
  • a flat dose i.e., 100 mg, 200 mg, 300 mg
  • a patient-specific dose i.e., mg/kg (mg therapeutic agent/kg of body weight) or mg/m (quantity in milligrams per square meter of body surface area).
  • the dose of the attenuated Listeria strain comprised by the immunogenic composition disclosed herein is administered to a subject at a dose of 1 x 10 - 3.31 x 10 10 CFU. In another embodiment, the dose is 1 x 10 8 - 3.31 x 10 10 CFU. In another embodiment, the dose is 1 x 10 9 - 3.31 x 10 10 CFU. In another embodiment, the dose is 5-500 x
  • the dose is 7-500 x 108 CFU. In another embodiment, the dose is 10-500 x 10 8 CFU. In another embodiment, the dose is 20-500 x 10 8 CFU. In another embodiment, the dose is 30-500 x 10 8 CFU. In another embodiment, the dose is 50-500 x 108 CFU. In another embodiment, the dose is 70-500 x 10 CFU. In another embodiment, the dose is 100-500 x 10 8 CFU. In another embodiment, the dose is 150-500 x 10 8 CFU. In another embodiment, the dose is 5-300 x 10 8 CFU. In another embodiment, the dose is 5-200 x 108 CFU. In another embodiment, the dose is 5-150 x 10 CFU. In another embodiment, the dose is 5-100 x
  • the dose is 5-70 x 108 CFU. In another embodiment, the dose is 5-50 x 10 8 CFU. In another embodiment, the dose is 5-30 x 108 CFU. In another embodiment, the dose is 5-20 x 10 8 CFU. In another embodiment, the dose is 1-30 x 109 CFU. In another embodiment, the dose is 1-20 x 10 9 CFU. In another embodiment, the dose is 2-30 x 10 9 CFU. In another embodiment, the dose is 1-10 x 10 9 CFU. In another embodiment, the dose is 2-10 x 10 9 CFU. In another embodiment, the dose is 3-10 x 10 9 CFU. In another embodiment, the dose is 2- 7 x 10 9 CFU.
  • the dose is 2-5 x 10 9 CFU. In another embodiment, the dose is 3-5 x 10 9 CFU. [00329] In another embodiment, the dose is 1 x 10 organisms. In another embodiment, the dose is 1 x 10 8 organisms. In another embodiment, the dose is 1 x 109 organisms. In another embodiment, the dose is 1.5 x 10 9 organisms. In another embodiment, the dose is 2 x 10 9 organisms. In another embodiment, the dose is 3 x 10 9 organisms. In another embodiment, the dose is 4 x 10 9 organisms. In another embodiment, the dose is 5 x 10 9 organisms. In another embodiment, the dose is 6 x 10 9 organisms. In another embodiment, the dose is 7 x 10 9 organisms.
  • the dose is 8 x 10 9 organisms. In another embodiment, the dose is 10 x 10 9 organisms. In another embodiment, the dose is 1.5 x 10 10 organisms. In another embodiment, the dose is 2 x 10 10 organisms. In another embodiment, the dose is 2.5 x 10 10 organisms. In another embodiment, the dose is 3 x 10 10 organisms. In another embodiment, the dose is 3.3 x 10 10 organisms. In another embodiment, the dose is 4 x 10 10 organisms. In another embodiment, the dose is 5 x 10 10 organisms. Each dose and range of doses represents a separate embodiment of the present invention.
  • repeat administrations (doses) of compositions disclosed herein may be undertaken immediately following the first course of treatment or after an interval of days, weeks or months to achieve tumor regression.
  • repeat doses may be undertaken immediately following the first course of treatment or after an interval of days, weeks or months to achieve suppression of tumor growth.
  • Assessment may be determined by any of the techniques known in the art, including diagnostic methods such as imaging techniques, analysis of serum tumor markers, biopsy, or the presence, absence or amelioration of tumor associated symptoms.
  • Boosting may encompass administering an additional strain or immunogenic composition or recombinant Listeria strain dose or immune checkpoint inhibitor alone or in combination to a subject.
  • 2 boosts or a total of 3 inoculations
  • 3 boosts are administered.
  • 4 boosts are administered.
  • 5 boosts are administered.
  • 6 boosts are administered.
  • more than 6 boosts are administered.
  • a method of present invention further comprises the step of boosting the subject with a recombinant Listeria strain or immune checkpoint inhibitor as provided herein.
  • the recombinant Listeria strain used in the booster inoculation is the same as the strain used in the initial "priming" inoculation.
  • the booster strain is different from the priming strain.
  • the recombinant immune checkpoint inhibitor used in the booster inoculation is the same as the inhibitor used in the initial "priming" inoculation.
  • the booster inhibitor is different from the priming inhibitor.
  • the same doses are used in the priming and boosting inoculations.
  • a larger dose is used in the booster.
  • the methods disclosed herein further comprise the step of administering to the subject a booster vaccination.
  • the booster vaccination follows a single priming vaccination.
  • a single booster vaccination is administered after the priming vaccinations.
  • two booster vaccinations are administered after the priming vaccinations.
  • three booster vaccinations are administered after the priming vaccinations.
  • the period between a prime and a boost strain is experimentally determined by the skilled artisan.
  • the period between a prime and a boost strain is 1 week, in another embodiment it is 2 weeks, in another embodiment, it is 3 weeks, in another embodiment, it is 4 weeks, in another embodiment, it is 5 weeks, in another embodiment it is 6-8 weeks, in yet another embodiment, the boost strain is administered 8-10 weeks after the prime strain.
  • a method disclosed herein further comprises boosting the subject with a immunogenic composition comprising an attenuated Listeria strain provided herein.
  • a method disclosed herein comprises the step of administering a booster dose of the immunogenic composition comprising the attenuated Listeria strain provided herein.
  • the booster dose is an alternate form of the immunogenic composition.
  • the methods disclosed herein further comprise the step of administering to the subject a booster immunogenic composition.
  • the booster dose follows a single priming dose of the immunogenic composition.
  • a single booster dose is administered after the priming dose.
  • two booster doses are administered after the priming dose.
  • three booster doses are administered after the priming dose.
  • the period between a prime and a boost dose of an immunogenic composition comprising the attenuated Listeria disclosed herein is experimentally determined by the skilled artisan.
  • the dose is experimentally determined by a skilled artisan.
  • the period between a prime and a boost dose is 1 week, in another embodiment it is 2 weeks, in another embodiment, it is 3 weeks, in another embodiment, it is 4 weeks, in another embodiment, it is 5 weeks, in another embodiment it is 6-8 weeks, in yet another embodiment, the boost dose is administered 8-10 weeks after the prime dose of the immunogenic composition.
  • DNA strain priming followed by boosting with protein in adjuvant or by viral vector delivery of DNA encoding antigen appears to be the most effective way of improving antigen specific antibody and CD4+ T-cell responses or CD8+ T-cell responses respectively.
  • US 2002/0165172 Al describes simultaneous administration of a vector construct encoding an immunogenic portion of an antigen and a protein comprising the immunogenic portion of an antigen such that an immune response is generated.
  • the document is limited to hepatitis B antigens and HIV antigens.
  • U.S. Pat. No. 6,500,432 is directed to methods of enhancing an immune response of nucleic acid vaccination by simultaneous administration of a polynucleotide and polypeptide of interest.
  • simultaneous administration means administration of the polynucleotide and the polypeptide during the same immune response, preferably within 0-10 or 3-7 days of each other.
  • the antigens contemplated by the patent include, among others, those of Hepatitis (all forms), HSV, HIV, CMV, EBV, RSV, VZV, HPV, polio, influenza, parasites (e.g., from the genus Plasmodium), and pathogenic bacteria (including but not limited to M. tuberculosis, M. leprae, Chlamydia, Shigella, B. burgdorferi, enterotoxigenic E. coli, S. typhosa, H. pylori, V. cholerae, B. pertussis, etc.). All of the above references are herein incorporated by reference in their entireties.
  • the recombinant polypeptide of methods disclosed herein is expressed by the recombinant Listeria strain.
  • the expression is mediated by a nucleotide molecule carried by the recombinant Listeria strain.
  • the immunosuppressive molecule antagonist and the live- attenuated Listeria strain that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells are combined in a single dosage form.
  • the immunosuppressive molecule antagonist and the live-attenuated Listeria monocytogenes strain bioengineered, by transforming it with an expression vector to express a survivin antigen fused to a tLLO are combined in a single dosage form.
  • pADV265) strain are combined in a single dosage form.
  • administering administration of an immunosuppressive molecule antagonist and a live-attenuated Listeria strain disclosed herein that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells is maintained throughout a period of treatment or prevention.
  • anti-cancer activity is achieved by subsequent administration of either component in isolation, i.e.- the immunosuppressive molecule antagonist or the live-attenuated Listeria strain (or a composition comprising either component).
  • the live-attenuated Listeria strain that is used to stimulate is used to stimulate
  • APCs capable of driving a cellular immune response to survivin expressing cells e.g. ADXS31- 265 (10403S pADV265) strain
  • ADXS31- 265 10403S pADV265 strain
  • one of the live-attenuated Listeria strains disclosed herein is administered after administration of the immunosuppressive molecule antagonist.
  • Each possibility represents a separate embodiment of the methods and compositions as provided herein.
  • an administration regimen comprises administering an immunosuppressive molecule antagonist disclosed herein followed by administration of a recombinant Listeria vaccine strain provided herein.
  • the order of administration of components of combination therapy is reversed.
  • administration of one component is immediately followed by administration of the other component.
  • the interval is at least 1-2 hours.
  • the interval is at least 2-3 hours.
  • the interval is at least 3-4 hours.
  • the interval is at least 4-5 hours.
  • the interval is at least 5-6 hours.
  • the interval is at least 6-8 hours.
  • the interval at least is 8-10 hours. In another embodiment, the interval is at least 10-12 hours. In another embodiment, the interval is at least one day. In another embodiment, the interval is at least two days. In another embodiment, the interval is at least three days. In another embodiment, the interval is at least four days. In another embodiment, the interval is at least five days. In another embodiment, the interval is at least six days. In another embodiment, the interval is at least seven days. In yet another embodiment, the interval is more than seven days.
  • At least one of the therapeutic agents in the combination therapy is administered using the same dosage regimen (dose, frequency and duration of treatment) that is typically employed when the agent is used as monotherapy for treating the same cancer.
  • the patient receives a lower total amount of at least one of the therapeutic agents in the combination therapy than when the agent is used as monotherapy, e.g., smaller doses, less frequent doses, and/or shorter treatment duration.
  • the method comprises the step of co-administering a composition comprising a recombinant Listeria with an additional therapy.
  • the additional therapy is surgery, chemotherapy, an immunotherapy, a radiation therapy, an antibody based immunotherapy provided herein, or a combination thereof.
  • the additional therapy precedes administration of a composition comprising a recombinant Listeria.
  • the additional therapy follows administration of the composition comprising a recombinant Listeria.
  • the additional therapy is an antibody therapy.
  • a composition comprising a recombinant Listeria is administered in increasing doses in order to increase the T-effector cell to regulatory T cell ration and generate a more potent anti-tumor immune response.
  • the anti-tumor immune response can be further strengthened by providing the subject having a tumor with cytokines including, but not limited to IFN- ⁇ , TNF-a, and other cytokines known in the art to enhance cellular immune response, some of which can be found in US Patent Serial No. 6,991,785, incorporated by reference herein.
  • an immunosuppressive molecule, a recombinant Listeria and an additional therapy are administered separately.
  • an administration regimen comprises administering a combination therapy disclosed herein via any regimen described above, followed by administration of one of additional therapies described above.
  • the order of administration of combination therapy and one of the additional therapies is reversed.
  • administration of the combination therapy is immediately followed or preceded by administration of one of the additional therapies.
  • the interval is at least 1-2 hours.
  • the interval is 2-3 hours.
  • the interval is 3-4 hours.
  • the interval is 4-5 hours.
  • the interval is 5-6 hours.
  • the interval is 6-8 hours.
  • the interval is 7-10 hours.
  • the interval is 9-12 hours. In another embodiment, the interval is at least one day. In another embodiment, the interval is two to three days. In another embodiment, the interval is three to five days. In another embodiment, the interval is five to seven days. In another embodiment, the interval is seven to ten days. In yet another embodiment, the interval is ten to fifteen days.
  • a method of increasing antigen- specific T cells in a subject suffering from cancer or having a tumor comprising the step of administering to the subject an immunogenic composition comprising a recombinant Listeria vaccine strain comprising a nucleic acid molecule, the nucleic acid molecule comprising a first open reading frame encoding fusion polypeptide, wherein the fusion polypeptide comprises a PEST sequence-containing polypeptide or PEST-sequence containing peptide fused to survivin antigen or fragment thereof, as described herein.
  • a combination therapy of the invention is administered to a patient who has not been previously treated with a biotherapeutic or chemotherapeutic agent, i.e., is treatment-naive.
  • the combination therapy is administered to a patient who failed to achieve a sustained response after prior therapy with a biotherapeutic or chemotherapeutic agent, i.e., is treatment-experienced.
  • sustained response may encompass a sustained therapeutic effect after cessation of treatment with a therapeutic agent, or a combination therapy described herein.
  • the sustained response has a duration that is at least the same as the treatment duration, or at least 1.5, 2.0, 2.5 or 3 times longer than the treatment duration.
  • a Bliss value of zero (0) is considered additive.
  • additive means that the result of the combination of the two targeted agents is the sum of each agent individually.
  • a combination therapy of the invention is typically used to treat a tumor that is large enough to be found by palpation or by imaging techniques well known in the art, such as MRI, ultrasound, or CAT scan.
  • a combination therapy of the invention is used to treat an advanced stage tumor having dimensions of at least about 200 mm 3 ' 300 mm 3 , 400 mm 3 , 500 mm 3 , 750 mm 3 , or up to 1000 mm 3.
  • a combination therapy of the invention is administered to a patient diagnosed with a pancreatic cancer that tests positive for PD-L1 expression.
  • PD-L1 expression is detected using a diagnostic anti-human PD-L1 antibody, or antigen binding fragment thereof, in an IHC assay on an FFPE or frozen tissue section of a tumor sample removed from the patient.
  • the patient's physician would order a diagnostic test to determine PD-L1 expression in a tumor tissue sample removed from the patient prior to initiation of treatment with the PD-1 antagonist or PD-L1 antagonist and the live-attenuated Listeria strains provided for herein, but it is envisioned that the physician could order the first or subsequent diagnostic tests at any time after initiation of treatment, such as for example after completion of a treatment cycle.
  • selecting a dosage regimen for a combination therapy of the invention depends on several factors, including the serum or tissue turnover rate of the entity, the level of symptoms, the immunogenicity of the entity, and the accessibility of the target cells, tissue or organ in the individual being treated.
  • a dosage regimen maximizes the amount of each therapeutic agent delivered to the patient consistent with an acceptable level of side effects.
  • the dose amount and dosing frequency of each biotherapeutic and chemotherapeutic agent in the combination depends in part on the particular therapeutic agent, the severity of the cancer being treated, and patient characteristics. Guidance in selecting appropriate doses of antibodies, cytokines, and small molecules are available.
  • Determination of the appropriate dosage regimen may be made by the clinician, e.g., using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment, and will depend, for example, the patient's clinical history (e.g., previous therapy), the type and stage of the cancer to be treated and biomarkers of response to one or more of the therapeutic agents in the combination therapy.
  • Biotherapeutic agents in a combination therapy of the invention may be administered by continuous infusion, or by doses at intervals of, e.g., daily, every other day, three times per week, or one time each week, two weeks, three weeks, monthly, bimonthly, etc.
  • a total weekly dose is generally at least 0.05 ⁇ g/kg, 0.2 ⁇ g/kg, 0.5 ⁇ g/kg, 1 ⁇ g/kg, 10 ⁇ g/kg, 100 ⁇ g/kg, 0.2 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg body weight or more. See, e.g., Yang et al. (2003) New Engl. J. Med.
  • the dosing regimen will comprise administering the anti- human PD-1 mAb at a flat dose of 100 to 500 mg or a weight-based dose of 1 to 10 mg/kg at intervals of about 14 days (+ 2 days) or about 21 days (+ 2 days) or about 30 days (+ 2 days) throughout the course of treatment.
  • the dosing regimen will comprise administering the anti- human PD-1 mAb at a dose of from about 0.005 mg/kg to about 10 mg/kg, with intra-patient dose escalation.
  • the interval between doses will be progressively shortened, e.g., about 30 days (+ 2 days) between the first and second dose, about 14 days (+ 2 days) between the second and third doses.
  • the dosing interval will be about 14 days (+ 2 days), for doses subsequent to the second dose.
  • treatment regimen In one embodiment, the terms "treatment regimen”, “dosing protocol” and “dosing regimen” are used interchangeably herein and encompass the dose and timing of administration of each therapeutic agent in a combination of the invention.
  • a subject will be administered an intravenous (IV) infusion of a medicament comprising any of the immunosuppressive molecules antagonists described herein.
  • the PD-1 antagonist in the combination therapy is nivolumab, which is administered intravenously at a dose selected from the group consisting of: 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W.
  • the PD-1 antagonist in the combination therapy PD-1 antagonist is administered in a liquid medicament at a dose selected from the group consisting of 200 mg Q3W, 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W or equivalents of any of these doses (e.g., a PK model of a PD-1 antagonist estimates that the fixed dose of 200 mg Q3W provides exposures that are consistent with those obtained with 2 mg/kg Q3W).
  • a PD-1 antagonist is administered as a liquid medicament which comprises 25 mg/ml the PD-1 antagonist, 7% (w/v) sucrose, 0.02% (w/v) polysorbate 80 in 10 mM histidine buffer pH 5.5, and the selected dose of the medicament is administered by IV infusion over a time period of 30 minutes +/-10 min.
  • Listeria in the combination therapy is a live-attenuated Listeria strain disclosed herein (e.g. ADXS31-265), which is administered in a liquid medicament at a dose selected from the group consisting of 1 x 10 9 , 5 x 10 9 and 1 x 10 10 CFU.
  • a live-attenuated Listeria strain disclosed herein e.g. ADXS31-265
  • a liquid medicament at a dose selected from the group consisting of 1 x 10 9 , 5 x 10 9 and 1 x 10 10 CFU.
  • a dose ranges from about 1 x 10 9 CFU up to 3.31 x 10 10 CFU, from about 5 x 10 8 CFU up to 5 x 10 10 CFU, from about 7 x 10 8 CFU up to 5 x 10 10 CFU, from about 1 x 10 9 CFU up to 5 x 10 10 CFU, from about 2 x 10 9 CFU up to 5 x 10 10 CFU, from about 3 x 10 9 CFU up to 5 x 10 10 CFU, from about 5 x 10 9 CFU up to 5 x 10 10 CFU, from about 7 x 10 9 CFU up to 5 x 10 10 CFU, from about 1 x 10 10 CFU up to 5 x 10 10 CFU, from about 1.5 x 10 9 CFU up to 5 x 10 10 CFU, from about 5 x 10° CFU - up to 3 x 10 10 CFU, from about 5 x 10° CFU up to 2 x 10 10 CFU, from about 5 x 10° CFU up to 1.5 x
  • a dose of recombinant Listeria ranges from about 1 x 10 organisms to aboutl.5 x 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 10 8 organisms to about 1.5 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 10 9 organisms to about 2 x 10 9 organisms. -, In another embodiment, a dose of recombinant Listeria ranges from about 2 x 10 9 organisms to about 5 x 10 9 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 2 x 10 9 organisms to about 1 x 10 10 organisms.
  • a dose of recombinant Listeria ranges from about 3 x 10 9 organisms to about 1 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 4 x 10 9 organisms to about 1 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 1 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 6 x 10 9 organisms to about 1 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 7 x 10 9 organisms to about 1 x 10 10 organisms.
  • a dose of recombinant Listeria ranges from about 1 x 10 9 organisms to about 5 x 10 9 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 10 9 organisms to about 4 x 10 9 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 10 9 organisms to about 3 x 10 9 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 8 x 10 9 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 1.5 x 10 10 organisms.
  • a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 2 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 2.5 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 3 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 3.5 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 4 x 10 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 10 9 organisms to about 5 x 10 10 organisms. In another embodiment, the dose ranges
  • compositions containing strains and compositions disclosed herein are 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, subcutaneously, intra-peritonealy, intra- ventricularly, intra-cranially, intra-vaginally or intra-tumorally.
  • enteral route of administration may encompass the administration via any part of the gastrointestinal tract.
  • enteral routes include oral, mucosal, buccal, and rectal route, or intragastric route.
  • Parenteral route of administration may encompass a route of administration other than enteral route.
  • parenteral routes of administration examples include intravenous, intramuscular, intradermal, intraperitoneal, intratumor, intravesical, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, transtracheal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal, subcutaneous, or topical administration.
  • the antibodies and compositions of the disclosure can be administered using any suitable method, such as by oral ingestion, nasogastric tube, gastrostomy tube, injection, infusion, implantable infusion pump, and osmotic pump.
  • the suitable route and method of administration may vary depending on a number of factors such as the specific antibody being used, the rate of absorption desired, specific formulation or dosage form used, type or severity of the disorder being treated, the specific site of action, and conditions of the patient, and can be readily selected by a person skilled in the art.
  • the strains 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 formulations include solutions, suspensions, dispersions, emulsions, oils and the like.
  • the active ingredient is formulated in a capsule.
  • the compositions disclosed herein comprise, in addition to the active compound and the inert carrier or diluent, a hard gelating capsule.
  • a composition comprising a Listeria strain is 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.
  • a vaccine or composition disclosed herein is 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.
  • a vaccine of the methods and compositions disclosed herein may be administered to a host vertebrate animal, preferably a mammal, and more preferably a human, either alone or in combination with a pharmaceutically acceptable carrier.
  • a vaccine is administered in an amount effective to induce an immune response to the Listeria strain itself or to a heterologous antigen which the Listeria species has been modified to express.
  • the amount of vaccine or immunogenic composition to be administered may be routinely determined by one of skill in the art when in possession of the present disclosure.
  • a pharmaceutically acceptable carrier may include, but is not limited to, sterile distilled water, saline, phosphate buffered solutions or bicarbonate buffered solutions.
  • the pharmaceutically acceptable carrier selected and the amount of carrier to be used will depend upon several factors including the mode of administration, the strain of Listeria and the age and disease state of the vaccinee.
  • administration of the vaccine may be by an oral route, or it may be parenteral, intranasal, intramuscular, intravascular, intrarectal, intraperitoneal, or any one of a variety of well-known routes of administration.
  • the route of administration may be selected in accordance with the type of infectious agent or tumor to be treated.
  • the present invention provides a method of treating, suppressing, or inhibiting at least one tumor in a subject comprising administering the immunogenic composition provided herein.
  • an attenuated bacteria or attenuated Listeria, or ADXS31-
  • the optimal dose for a combination therapy comprising an anti-PD-1 antibody disclosed herein in combination with a live-attenuated Listeria strain disclosed herein is identified by dose escalation of one or both of these agents.
  • the optimal dose for a composition comprising either the anti-PD-1 antibody disclosed herein or the live- attenuated Listeria strain disclosed herein is identified by dose escalation of one or both of these agents.
  • a patient is treated with the combination therapy disclosed herein on day 1 of weeks 1, 4 and 7 in a 12 week cycle, starting with anti-PD-1 antibody is administered at a starting dose of 50, 100, 150, or 200 mg, and a live-attenuated Listeria strain disclosed herein at a starting dose of ranging from about 1 x 10 7 CFU to about 3.5 x 1010 CFU.
  • the anti-PD-1 antibody infusion is administered first, followed by a NSAIDS, e.g., naproxen or ibuprofen, and oral antiemetic medication within a predetermined amount of time prior to administration of a live-attenuated Listeria strain provided herein.
  • NSAIDS e.g., naproxen or ibuprofen
  • the predetermined amount of time is 5-10 min, 11-20 min, 21-40 min, 41-60 min. In another embodiment, the predetermined amount of time is at least one hour. In another embodiment, the predetermined amount of time is 1-2 hours, 2-4 hours, 4-6 hours, 6- 10 hours. In another embodiment, administrations of a NSAIDS, e.g., naproxen or ibuprofen, and oral antiemetic medication is repeated on a need basis to the subject, prior to administration of a live-attenuated Listeria strain provided herein.
  • a NSAIDS e.g., naproxen or ibuprofen
  • oral antiemetic medication is repeated on a need basis to the subject, prior to administration of a live-attenuated Listeria strain provided herein.
  • anti-PD-1 antibody is administered at a starting dose of
  • Q3W 50, 100, 150 or 200 mg Q3W and a live-attenuated Listeria strain disclosed herein is administered Q3W at a starting dose of between 1 x 10 7 and 3.5 x 1010 CFU.
  • a live-attenuated Listeria strain disclosed herein is administered at a starting dose of 5 x 10 9 Q3W and an anti-PD-1 antibody is administered at a starting dose of 200 mg Q3W, and if the starting dose of the combination is not tolerated by the patient, then the dose of the live-attenuated Listeria strain disclosed herein is reduced to 1 x 10 9 cfu Q3W or the dose of the anti-PD-1 antibody is reduced to 150 mg Q3W. It is to be understood by a skilled artisan that the doses of any of the components of a combination therapy disclosed herein may be incrementally adjusted to a lower or higher dose, as further provided herein, based on a subjects response to the combination therapy.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed, as determined by those skilled in the art.
  • a treatment cycle begins with the first day of combination treatment and lasts for at least 12 weeks, 24 weeks or 48 weeks.
  • the timing between the separate IV infusions of an anti-PDl antibody and a live-attenuated Listeria strain disclosed herein is between about 15 minutes to about 45 minutes.
  • the invention contemplates that an anti-PD-1 antibody and a live-attenuated Listeria strain disclosed herein may be administered in either order or by simultaneous IV infusion.
  • the combination therapy is administered for at least 2 to 4 weeks after the patient achieves a CR.
  • a patient selected for treatment with the combination therapy of the invention has been diagnosed with a metastatic prostate cancer and the patient has progressed or become resistant to no more than 2 prior systemic treatment regimens. In some embodiments, a patient selected for treatment with the combination therapy of the invention has been diagnosed with a metastatic prostate cancer and the patient has progressed or become resistant to no more than 3 prior systemic treatment regimens.
  • the present invention also provides a medicament which comprises a PD-1 antagonist disclosed herein and a pharmaceutically acceptable excipient.
  • the PD-1 antagonist is a biotherapeutic agent, e.g., a mAb
  • the antagonist may be produced in a producing cell line known in the art, such as, but not limited to CHO cells using conventional cell culture and recovery/purification technologies.
  • a medicament comprising an anti-PD-1 antibody disclosed herein may be provided as a liquid formulation or prepared by reconstituting a lyophilized powder with sterile water for injection prior to use.
  • WO 2012/135408 describes the preparation of liquid and lyophilized medicaments comprising an anti-PD-1 antibody that are suitable for use in the present invention.
  • a medicament comprising an anti-PD-1 antibody is provided in a glass vial which contains about 50 mg of anti-PD-1 antibody.
  • the present invention also provides a medicament which comprises a live- attenuated Listeria strain disclosed herein and a pharmaceutically acceptable excipient.
  • the PD-1 antagonist medicament and the live-attenuated Listeria strain disclosed herein medicament may be provided as a kit which comprises a first container and a second container and a package insert.
  • the first container contains at least one dose of a medicament comprising an anti-PD-1 antibody
  • the second container contains at least one dose of a medicament comprising a live-attenuated Listeria strain provided herein
  • the package insert, or label which comprises instructions for treating a patient for a prostate cancer using the medicaments.
  • the first and second containers may be comprised of the same or different shape (e.g., vials, syringes and bottles) and/or material (e.g., plastic or glass).
  • the kit may further comprise other materials that may be useful in administering the medicaments, such as diluents, filters, IV bags and lines, needles and syringes.
  • the anti-PD-1 antagonist is an anti-PD-1 antibody and the instructions state that the medicaments are intended for use in treating a patient having a prostate cancer that tests positive for PD-L1 expression by an IHC assay.
  • the term “comprise” or grammatical forms thereof may encompass the inclusion of the indicated active agent, such as the Lm strains of this invention, as well as inclusion of other active agents, such as an antibody or functional fragment thereof, and pharmaceutically acceptable carriers, excipients, emollients, stabilizers, etc., as are known in the pharmaceutical industry.
  • the term “consisting essentially of may encompass a composition, whose only active ingredient is the indicated active ingredient, however, other compounds may be included which are for stabilizing, preserving, etc. the formulation, but are not involved directly in the therapeutic effect of the indicated active ingredient.
  • the term “consisting essentially of may encompass components, which exert a therapeutic effect via a mechanism distinct from that of the indicated active ingredient. In some embodiments, the term “consisting essentially of may encompass components, which exert a therapeutic effect and belong to a class of compounds distinct from that of the indicated active ingredient. In some embodiments, the term “consisting essentially of may encompass components, which exert a therapeutic effect and may be distinct from that of the indicated active ingredient, by acting via a different mechanism of action. In some embodiments, the term “consisting essentially of may encompass components which facilitate the release of the active ingredient. In some embodiments, the term “consisting” may encompass a composition, which contains the active ingredient and a pharmaceutically acceptable carrier or excipient.
  • the term "About" when used to modify a numerically defined parameter may encompass variation of the parameter in quantitative terms plus or minus 5%, or in another embodiment plus or minus 10%, or in another embodiment plus or minus 15%, or in another embodiment plus or minus 20% of stated numerical value for that parameter.
  • a dose of about 200 mg of the PD-1 antagonist may vary between 180 mg and 220 mg.
  • the term “subject” can encompass a mammal including an adult human or a human child, teenager or adolescent in need of therapy for, or susceptible to, a condition or its sequelae, and also may include non-human mammals such as dogs, cats, pigs, cows, sheep, goats, horses, rats, and mice. It will also be appreciated that the term may encompass livestock. The term “subject” does not exclude an individual that is normal in all respects.
  • mammal for purposes of treatment refers to any animal classified as a mammal, including, but not limited to, humans, domestic and farm animals, and zoo, sports, or pet animals, such as canines, including dogs, and horses, cats, cattle, pigs, sheep, etc.
  • DNA sequence of plasmid pAdv265.5 comprises
  • Tumor pieces were harvested from primary pancreatic tumors arising spontaneously in KPC mice. Tumors were minced to lxl mm pieces and incubated with collagenase (1 mg/mL) and DNAse I (75ug/mL,Roche #10104159001) for thirty minutes at 37°C with 5% C0 2. The digested tumor was homogenized and passed through a 70 ⁇ filter and washed with Dulbecco's Modified Eagle's medium (DMEM).
  • DMEM Dulbecco's Modified Eagle's medium
  • Peri-tumoral LN were defined as lymph nodes that were physically attached to pancreatic tumors in KPC mice. Peri-tumoral LN from individual mice were removed from tumors and pooled for homogenization by gentle mashing on a 70 ⁇ cell strainer. Mesenteric lymph nodes were excised and homogenized by gentle mashing on a 70 ⁇ cell strainer. Single cell suspensions were washed and resuspended in DMEM supplemented with 10% FCS.
  • Lm Listeria monocytogenes
  • LmddA-265 encoding human Survivin protein
  • ADXS 11-001 encoding the HPV-E7 protein
  • Mice were injected i.p. with 1x10 8 CFU of Lm and 2x108 CFU of Lm for the second immunization.
  • KPC mice received 1 Lm immunization of 1x10 CFU.
  • PD-1 blockade 100 ⁇ g of anti-mouse PD-1 (clone RMP1-14; BioXCell BE0146) or control 2A3 antibody (BioXCell BE0089) were diluted in sterile PBS and administered i.p.
  • Tissues were flash frozen in Tissue-Tek O.C.T. Compound (V.W.R. 25608-930) and stored at -80°C. Frozen tissue sections were prepared by the University of Pennsylvania Cancer Histology Core. For PD-L1 and Her2 staining, tissue samples were thawed and immediately fixed with 3% buffered formaldehyde for 5 minutes at room temperature. Slides were then washed 2 times with PBST (PBS with 0.1% Tween-20) for 2 minutes each wash. Slides were then blocked with 10% goat serum in PBS for 45 minutes at room temperature, followed by a wash in PBST.
  • PBST PBS with 0.1% Tween-20
  • Tissues were flash frozen in Tissue-Tek O.C.T. Compound (V.W.R. 25608-930) and stored at -80C°. Frozen tissue sections were prepared by the University of Pennsylvania Cancer Histology Core and the University of Pennsylvania Molecular Pathology Imaging Core. Tissue samples were thawed and immediately fixed with either 3% buffered formaldehyde for 15 minutes at room temperature (for CD45 and mSurvivin staining) or with methanol at -20°C for 10 minutes (for CD3, CD4, CD8 and Foxp3 staining). Slides were then washed 2 times with PBST (PBS with 0.1% Tween-20) for 2 minutes each wash.
  • PBST PBS with 0.1% Tween-20
  • Endogenous peroxidase activity was blocked by incubating slides in a solution of 0.3% hydrogen peroxide (diluted in distilled H 2 0) for 10 minutes at room temperature. Slides were washed as described above. Slides were then blocked with 10% goat serum in PBS for 45 minutes at room temperature.
  • rat anti-mouse CD45 (clone 30-F11; BD 550539), rat anti-mouse CD3 antibody (clone KT3; Serotec/Bio-Rad MCA500GA) at a dilution of 1:50
  • rat anti-mouse CD4 antibody (clone GK1.5; BioXCell) at a dilution of 15ug/mL
  • rat anti-mouse CD8 antibody (clone 2.43; BioXCell) at a dilution of 15ug/mL
  • rat anti-mouse Foxp3 antibody (clone FJK-16s;eBioscience 14-5773-82) at a dilution of 1: 100
  • rat anti-mouse Survivin (CST 71G4B7).
  • Detection was performed with DAB (3, 3 '-diaminobenzidine) HRP substrate that was prepared according to manufacturer instructions (Vector Laboratories SK- 4100). Slides were washed in distilled H 2 0 and counterstained with Hematoxylin (Hematoxylin Stain-Harris; Sigma- Aldrich HHS 16) diluted 1: 1 in distilled H 2 0 for forty seconds. Slides were rinsed with H 2 0 and dehydrated by sequential washes of 95% ethanol, 100% ethanol and xylenes. After drying, slides were mounted using Cytoseal 60 (Fisher 23-244257). Images were acquired on an Olympus BX43 microscope using CellSens Digital Imaging Software (Olympus). Quantification was performed by averaging the number of positive events counted in 4 high powered fields (40X magnification).
  • Tumor pieces were harvested from primary PDA tumors that spontaneously arose in KPC mice backcrossed onto the C57BL/6 genetic background. Tumor was minced to lxl mm pieces and incubated with collagenase (lmg/mL) for thirty minutes at 37°C with 5% C0 2. The digested tumor was homogenized and passed through a 70 ⁇ filter and washed with DMEM. Cells were spun at 1600 RPM for 5 minutes and washed 2 times with DMEM. Cells were resuspended in serum-free media and plated at a density of 1-3 xlO 6 cells/well of a 6-well plate.
  • cells were cultured in DMEM supplemented with 5% fetal calf serum, 1% L-glutamine, and gentamycin. Cells were used for in vitro studies after at least 5 passages.
  • IFN- ⁇ treatment of cell lines cells were plated in 12 well plates 24 hours prior to IFN- ⁇ addition.
  • Mouse recombinant IFN- ⁇ (R&D 485 Ml/CF) was added at 100 ng/niL and incubated for 12-72 hours.
  • Flow cytometric analysis PDA tumor cell lines was performed by dissociating adherent cells with 0.05% Trypsin/EDTA (Gibco Life Technologies), followed by washes in DMEM and PBS. Single cell suspensions from tissue samples were prepared as describe above. lxlO 6 cells were added to a round bottom 96-well plate for antibody incubations. Cells were first stained with Live/Dead Fixable Aqua Dead Cell Stain (Life Technologies L34957) according to manufacturer instructions.
  • MHC1 FITC H-2Db clone KH95; BD 553573
  • MHCII PE I-A/I-E clone M5/115.15.2; BD 557000
  • PD-L1 PE clone MIH5; BD 558091
  • CD3 DPerCP/Cy5.5 clone 145-2C11; eBioscience 561008
  • CD44 APC e-Fluor 780 clone IM7; eBioscience 47-0441-80
  • PD1 PE/Cy7 clone RMPl-30; Biolegend 109110
  • CD8b PE clone H35-17.2; eBio 53-0083-82
  • mice were treated with Lm-LLO and Lm-LLO-cHER2 prior to PDA tumor implantation (Fig. 3A) and the growth of subsequently implanted PDA tumors was assessed. The preventive treatment has no observable effect on growth of 7861. PDA cell line-derived tumors introduced in mice (Fig. 3B).
  • STIMULATES T CELL INFILTRATION PDA tumor cells were examined for expression of survivin and found to express high levels of survivin (Fig. 4A). The effect of, Lm-LLO-Survivin treatment on tumor growth and T cell infiltration was examined. Surprisingly, the treatment had no effect on growth of 69.PDA cell line- derived tumors established in mice (Fig. 4B) despite clear increase of T-cell infiltration into these tumors (Figs. 4C and D).
  • PD-Ll/PD-1 signaling pathway The expression of the components of PD-Ll/PD-1 signaling pathway was investigated. While mouse tumors express high levels of PD-Ll (Fig. 5B), the T cells isolated from mesenteric versus peritumoral lymph nodes show high level of PD-1 expression (Fig. 5C).
  • EXAMPLE 7 COMBINATION LM-LLO-SURVIVIN-ANTI PD1 TREATMENT OF PDA

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Environmental Sciences (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Endocrinology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Husbandry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to combination therapies useful for the treatment of cancer. In particular, the invention relates to the treatment of pancreatic cancer using an antagonist of a Programmed Death 1 protein (PD-1) in combination with a live attenuated recombinant Listeria strain comprising a fusion protein of a PEST sequence-containing polypeptide fused to a tumor-associated antigen.

Description

COMBINATION OF A PD-1 ANTAGONIST AND A LISTERIA BASED VACCINE FOR TREATING PANCREATIC CANCER
GOVERNMENT INTEREST STATEMENT
[0001] This invention was made with government support under Grant Number W81XWH-12-1- 0411 awarded by the Department of Defense and NIH K08CA138907, awarded by the National Institutes of Health. The United States government has certain rights in the invention.
FIELD OF INVENTION
[0002] The present invention relates to combination therapies useful for the treatment of cancer. In particular, the invention relates to the treatment of pancreatic cancer using an antagonist of a Programmed Death 1 protein (PD-1) in combination with a live attenuated recombinant Listeria strain comprising a fusion protein of a PEST sequence-containing polypeptide fused to a tumor-associated antigen.
BACKGROUND OF THE INVENTION
[0003] Pancreatic cancer arises when cells in the pancreas begin to multiply out of control and form a mass. These cancer cells have the ability to invade other parts of the body. There are a number of types of pancreatic cancer, and the most common type, pancreatic adenocarcinoma, accounts for about 85% of cases. In 2012 pancreatic cancers of all types were the seventh most common cause of cancer deaths, resulting in 330,000 deaths globally. In the United States, pancreatic cancer is the fourth most common cause of deaths due to cancer. The disease occurs most often in the developed world, where about 70% of the new cases in 2012 originated. Pancreatic adenocarcinoma typically has a very poor prognosis: after diagnosis, 25% of people survive one year and 5% live for five years. For cancers diagnosed early the five-year survival rate rises to about 20%. Risk factors for pancreatic cancer include tobacco smoking, obesity, diabetes, and certain rare genetic conditions. Pancreatic cancer can be treated with surgery, radiotherapy, chemotherapy, palliative care, or a combination of these. Surgery is the only treatment that can cure the disease. [0004] PD-1 is recognized as an important player in immune regulation and the maintenance of peripheral tolerance. PD-1 is moderately expressed on naive T, B and NKT cells and up-regulated by T/B cell receptor signaling on lymphocytes, monocytes and myeloid cells.
[0005] Two known ligands for PD-1, PD-Ll (B7-H1) and PD-L2 (B7-DC), are expressed in human cancers arising in various tissues. In large sample sets of ovarian, renal, colorectal, pancreatic, liver cancers and melanoma, it was shown that PD-Ll expression correlated with poor prognosis and reduced overall survival irrespective of subsequent treatment. Similarly, PD-1 expression on tumor infiltrating lymphocytes was found to mark dysfunctional T cells in breast cancer and melanoma and to correlate with poor prognosis in renal cancer. Thus, it has been proposed that PD-Ll expressing tumor cells interact with PD-1 expressing T cells to attenuate T cell activation and evasion of immune surveillance, thereby contributing to an impaired immune response against the tumor. There are several monoclonal antibodies that inhibit the interaction between PD-1 and one or both of its ligands PD-Ll and PD-L2 are in clinical development for treating cancer.
[0006] Survivin, an inhibitor of apoptosis protein, is encoded by the BIRC5 gene in humans. This protein is highly conserved across species (human and canine survivin proteins share 91.5% sequence identity). Survivin is a member of the inhibitor of apoptosis (IAP) family. The survivin protein functions to inhibit caspase activation, thereby leading to negative regulation of apoptosis or programmed cell death. Survivin is detectable in a wide range of adult tissues in humans, dogs and mice.
[0007] Survivin is highly expressed in most human cancers and associated with chemotherapy resistance, increased tumor recurrence, and shorter patient survival. Due to its presence in many types of cancer tumors, e.g. breast and colon cancer, lymphoma, leukemia, and melanoma, it is believed that survivin could serve as a universal target antigen for anticancer immunotherapy. Expression of survivin has also been reported in the vast majority of canine lymphomas, as well as in canine mast cell tumor, hemangiosarcoma, and transitional cell carcinoma. High levels of survivin expression in canine B cell lymphoma correlate with reduced disease-free interval (DFI) and median survival time (MST). Immunization of human patients using survivin peptides has shown some promise in the human clinics with only mild grade Ι/Π adverse effects that include anemia, lethargy and fever. Serious adverse effects have not been reported.
[0008] Listeria monocytogenes (L. monocytogenes or Lm) 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. Since L. monocytogenes has access to both phagosomal and cytosolic compartments, antigens delivered by Lm can be presented in the context of both MHC I and Π molecules, resulting in strong but preferentially cellular immune responses.
[0009] The low survival rates of pancreatic cancer patients create a clear need for a more robust treatment of this type of cancer. The present invention addresses the needs of subjects suffering from pancreatic cancer by providing a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen or immunogenic fragment thereof and an immunosuppressive molecule antagonist.
SUMMARY OF THE INVENTION
[0010] In one aspect, disclosed herein a method of eliciting an anti-tumor or anti-cancer immune response against a tumor or cancer in a subject, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist, thereby eliciting an anti-tumor or anti-cancer immune response in said subject.
[0011] In another aspect, disclosed herein a method of preventing a tumor or cancer in a subject, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration treats a tumor or a cancer in said subject. In another embodiment, the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer.
[0012] In one aspect, disclosed herein a method of treating a tumor or cancer in a subject, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration treats a tumor or a cancer in said subject. In another embodiment, the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer. [0013] In another aspect, disclosed herein a method of delaying metastatic disease in a subject having tumor or cancer, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration delays metastatic disease in said subject.
[0014] In another aspect, disclosed herein a method of breaking tolerance to survivin in a subject having tumor or cancer, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration breaks tolerance to survivin in said subject.
[0015] In another aspect, disclosed herein a method of prolonging survival of a subject having tumor or cancer, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration prolongs survival in said subject.
[0016] In another aspect, disclosed herein a method of stimulating an anti-tumor immune response in a subject having tumor or cancer, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration stimulates an anti-tumor immune response in said subject.
[0017] In another aspect, said antagonist is an antibody or functional fragment thereof. In another aspect, said immunosuppressive molecule is PD-1 B7-1, PD-L1, and PD-L2. In another aspect, said recombinant Listeria strain comprises a nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide, wherein the said fusion polypeptide comprises a PEST sequence- containing polypeptide fused to survivin antigen or immunogenic fragment thereof. In another aspect, said PEST-containing peptide or polypeptide is selected from the group consisting of an N-terminal non-hemolytic Listeriolysin (LLO) fragment, an N-terminal ActA fragment, and a PEST-containing amino acid sequence. In another aspect, said nucleic acid molecule comprises a second open reading frame encoding a metabolic enzyme, and wherein said metabolic enzyme complements a mutation in the chromosome of said recombinant Listeria strain. In another aspect, the metabolic enzyme is an alanine racemase enzyme or a D-amino acid transferase enzyme. In another aspect, said recombinant Listeria comprises a mutation in the actA virulence gene. In another aspect, said recombinant attenuated Listeria is a Listeria monocytogenes.
[0018] In another aspect, said subject is a human. In another aspect, administering a composition comprising said recombinant attenuated Listeria prevents escape mutations within a tumor or cancer, results in progression free survival, inhibiting tumor growth, inducing cancer regression, extension of progression free survival (PFS), increasing time to disease progression, or any combination thereof. In another aspect, said tumor or cancer comprises a pancreatic tumor or pancreatic cancer. In another aspect, said cancer is a refractory cancer. In another embodiment, said tumor is a survivin-expressing tumor. In another embodiment, said cancer is a survivin- expressing cancer. [0019] Other features and advantages disclosed herein will become apparent from the following detailed description examples and figures. It should be understood, however, that the detailed description and the specific examples while indicating preferred embodiments of the invention are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
[0020] The subject matter regarded as the invention is particularly pointed out and distinctly claimed in the concluding portion of the specification. The invention, however, both as to organization and method of operation, together with objects, features, and advantages thereof, may best be understood by reference to the following detailed description when read with the accompanying drawings in which:
[0021] Figures 1A-D. Presents a representative image of Her2 staining by immunofluorescence (IF) on spontaneously arising tumor in the KPC (KrasG12D/+; Trp53R172H/+; Pdx-lCre) model (Figure 1A). Tumor cell lines derived from KPC mice showing Her2 expression by flow cytometry (Figure IB). Western blot analysis of tumor cell lines from KPC mice show Her2 expression (Figure 1C). Pancreatic ductal carcinoma (PDA) cell lines implanted s.c. with anti- CD40 treatment inducing T cells specific for Her2-specific peptides predicted by syfpethi algorithm (Figure ID). [0022] Figures 2A-C. Shows two tumor cell lines selected for studies: 7681. PDA, which shows minimal stromal compartment and 69. PDA with stromal compartment (Figure 2A). Presents 14 day old established tumors (7681) that were treated weekly beginning on day 0 with Listeria vaccine constructs and wherein tumor volume was assessed at day +14 (Figure 2B). Mice were challenged with 69. PDA tumors and treated with a single dose of vaccine on day 4 and tumor growths were monitored (Figure 2C). [0023] Figures 3A-B. Presents a treatment model schematic for prophylaxis therapy with
Listeria vaccine (Figure 3A). Presents tumor growth curves showing that Lm-LLO-E7 and Lm- LLO-Her2 treatment does not affect tumor growth (Figure 3B).
[0024] Figures 4A-D. Presents flow cytometry analysis of tumor cell line for expression of surviving (intracellular analysis) (Figure 4A). Presents tumor growth curves for mice challenged with 69. PDA and treated beginning on day 10 with single dose of Listeria vaccine (Figure 4B). Presents LSL-KrasG12D/+ ;LSL-Trp53R172H/+ ;Pdx-l-Cre (KPC) mice with established tumors that were treated with single dose of Listeria vaccines and wherein 14 days later tumor tissue was evaluated for T cell infiltration (Figure 4C). Presents a representative IHC images of T cell infiltration 14 days after Listeria vaccine therapy (Figure 4D). [0025] Figures 5A-C. Presents Ex vivo flow cytometric analysis of CD45+ Epcam(neg) leukocytes vs CD45(neg) Epcam+ tumor cells isolated from treatment naive KPC mice. Shown is MFI for MHC Class I molecules (H-2Db and H-2Kb), MHC Class II (I-A/I-E) and co- stimulatory molecules CD86 and CD40. ND = not detectable compared to FMO (Figure 5A). Shows a representative immunofluorescence image showing expression of PD-L1 in tumor from KPC mouse (Figure 5B). Shows expression of PD-1 on T cells isolated from mesenteric versus peritumoral lymph nodes from a KPC mouse (Figure 5C).
[0026] Figures 6A-D. Presents the graph showing the impact of IFN-g treatment on percentage of tumor cells expressing MHC class I (Figure 6A). Presents the graph showing the impact of IFN-g treatment on the level of MHC expression (Figure 6B). Presents the graph showing the impact of IFN-g treatment on percentage of tumor cells expressing PD-L1 (Figure 6C). Presents the graph showing the impact of IFN-g treatment on the level of PD-L1 expression (Figure 6D).
[0027] Figures 7A-D. Presents the treatment model schematic for Listeria vaccine therapy in combination with PD-1 blockade (Figure 7A). Presents the individual mouse growth curves (Figure 7B). Presents the average growth curves (Figure 7C). Presents the overall survival (Figure 7D). DETAILED DESCRIPTION OF THE PRESENT INVENTION
[0028] In the following detailed description, numerous specific details are set forth in order to provide a thorough understanding of the invention. However, it will be understood by those skilled in the art that the present invention may be practiced without these specific details. In other instances, well-known methods, procedures, and components have not been described in detail so as not to obscure the present invention.
Abbreviations. Throughout the detailed description and examples of the invention the following abbreviations will be used:
[0029] ADXS31-265 Listeria monocytogenes (10403S dal dat actA'
[0030] APC antigen presenting cell
[0031] BID One dose twice daily
[0032] CBD Cholesterol Binding Domain
[0033] CDR Complementarity determining region
[0034] CFU Colony-forming units
[0035] CHO Chinese hamster ovary
[0036] DFS Disease free survival
[0037] DTR Dose limiting toxicity
[0038] FFPE formalin-fixed, paraffin-embedded
[0039] FR Framework region
[0040] IgG Immunoglobulin G
[0041] IHC Immunohistochemistry or immunohistochemical
[0042] LLO Listeriolysin O polypeptide
[0043] tLLO truncated Listeriolysin O
[0044] Lm Listeria monocytogenes
[0045] MTD Maximum tolerated dose
[0046] NCBI National Center for Biotechnology Information
[0047] NCI National Cancer Institute
[0048] OR Overall response
[0049] ORF Open reading frame
[0050] OS Overall survival
[0051] PCR Polymerase chain reaction [0052] PD Progressive disease
[0053] PFS Progression free survival
[0054] PR Partial response
[0055] Q2W One dose every two weeks
[0056] Q3W One dose every three weeks
[0057] Q4W One dose every four weeks
[0058] QD One dose per day
[0059] RECIST Response Evaluation Criteria in Solid Tumors
[0060] SD Stable disease
[0061] SDS-PAGE Sodium dodecyl sulfate- Polyacrylamide gel electrophoresis
[0062] TILs Tumor infiltrating lymphocytes
[0063] VH Immunoglobulin heavy chain variable region
[0064] VK Immunoglobulin kappa light chain variable region I. RECOMBINANT LISTERIA STRAINS
[0065] In one embodiment, disclosed herein is a recombinant attenuated Listeria strain comprising a nucleic acid molecule encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof or an immunogenic fragment thereof fused to a PEST-containing peptide or polypeptide. In another embodiment, the nucleic acid molecule comprises a first open reading frame encoding the fusion polypeptide. In another embodiment, a PEST-containing peptide or polypeptide is a truncated Listeriolysin O (LLO) protein (tLLO). In another embodiment, the PEST-containing peptide or polypeptide is a fragment of LLO. In another embodiment, the PEST-containing peptide or polypeptide is an N- terminal fragment of LLO. In another embodiment, a PEST-containing polypeptide or polypeptide is a truncated ActA protein. In another embodiment, a PEST-containing peptide or polypeptide is fragment of an ActA protein. In another embodiment, the PEST-containing peptide or polypeptide is an N-terminal fragment of ActA. In another embodiment, the PEST-containing peptide or polypeptide is PEST-containing amino acid sequence. In another embodiment, the nucleic acid molecule further comprises a second open reading frame encoding a metabolic enzyme, wherein the metabolic enzyme complements a mutation in the chromosome of the recombinant Listeria strain.
[0066] It will be appreciated by a skilled artisan that the term "nucleic acids" or "nucleotide" may encompass a string of at least two base-sugar-phosphate 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 et 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 as provided herein.
[0067] The term "amino acid" or "amino acids" is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phospho threonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine. Furthermore, the term "amino acid" may include both D- and L-amino acids.
[0068] It will be appreciated by a skilled artisan that the term "open reading frame" or "ORF" may encompass a portion of an organism's genome which contains a sequence of bases that could potentially encode a protein. In another embodiment, the start and stop ends of the ORF are not equivalent to the ends of the mRNA, but they are usually contained within the mRNA. In one embodiment, ORFs are located between the start-code sequence (initiation codon) and the stop- codon sequence (termination codon) of a gene. Thus, in one embodiment, a nucleic acid molecule operably integrated into a genome as an open reading frame with an endogenous polypeptide is a nucleic acid molecule that has integrated into a genome in the same open reading frame as an endogenous polypeptide.
[0069] It will be appreciated by a skilled artisan that the term "endogenous" may encompass an item that has developed or originated within the reference organism or arisen from causes within the reference organism. For example, endogenous refers to native.
[0070] It will be appreciated by a skilled artisan that the term "fragment" may encompass a protein or polypeptide that is shorter or comprises fewer amino acids than the full length protein or polypeptide. In one embodiment, a fragment is an N-terminal fragment. In another embodiment, a fragment is a C-terminal fragment. In yet another embodiment, a fragment is an intrasequential section of the protein or peptide. In one embodiment, a fragment is a functional fragment. In another embodiment, a fragment is an immunogenic fragment. In one embodiment, a fragment has 10-20 amino acids, while in another embodiment, a fragment has more than 5 amino acids, while in another embodiment, a fragment has 100-200 amino acids, while in another embodiment, a fragment has 100-500 amino acids, while in another embodiment, a fragment has 50-200 amino acids, while in another embodiment, a fragment has 10-250 amino acids.
[0071] In an alternate embodiment, the term "fragment" refers to a nucleic acid that is shorter or comprises fewer nucleotides than the full length nucleic acid. In one embodiment, a fragment is a 5 '-terminal fragment. In another embodiment, a fragment is a 3 '-terminal fragment.
In yet another embodiment, a fragment encodes an intrasequential section of the protein. In one embodiment, a fragment has 10-20 nucleotides, while in another embodiment, a fragment has more than 5 nucleotides, while in another embodiment, a fragment has 100-200 nucleotides, while in another embodiment, a fragment has 100-500 nucleotides, while in another embodiment, a fragment has 50-200 nucleotides, while in another embodiment, a fragment has 10-250 nucleotides.
[0072] It will be appreciated by a skilled artisan that the term "functional" within the meaning of the invention, may encompass the innate ability of a protein, peptide, nucleic acid, fragment or a variant thereof to exhibit a biological activity or function. In one embodiment, such a biological function is its binding property to an interaction partner, e.g., a membrane-associated receptor, and in another embodiment, its trimerization property. In the case of functional fragments and the functional variants of the invention, these biological functions may in fact be changed, e.g., with respect to their specificity or selectivity, but with retention of the basic biological function.
[0073] It will be appreciated by a skilled artisan that the term "functional fragment" may encompass an immunogenic fragment and elicits an immune response when administered to a subject alone or in a strain composition provided herein. In another embodiment, a functional fragment has biological activity as will be understood by a skilled artisan and as further provided herein.
[0074] It will be appreciated by a skilled artisan that the term "fused" may encompass an operable linkage by covalent bonding. In one embodiment, the term encompasses recombinant fusion (of nucleic acid sequences or open reading frames thereof). In another embodiment, the term encompasses chemical conjugation.
[0075] In one embodiment, an LLO sequence comprises PEST amino acid (AA) sequence. In another embodiment, the PEST amino acid sequence is a putative PEST sequence present in LLO. In another embodiment, a truncated LLO comprises a PEST amino acid (AA) sequence. In another embodiment, a PEST AA sequence comprises a truncated LLO sequence. In another embodiment, a PEST AA sequence comprises an LLO fragment sequence. In another embodiment, a PEST AA sequence comprises a truncated ActA sequence. In another embodiment, a truncated ActA sequence comprises a PEST sequence. In another embodiment, PEST AA sequence comprises an ActA fragment sequence. For example, the PEST amino acid sequence may be KENS IS S M APP AS PP AS PKTPIEKKH ADEID K (SEQ ID NO: 1). Furthermore, fusion of an antigen to other Listeria monocytogenes (Lm) PEST AA sequences from Listeria may enhance immunogenicity of the antigen.
[0076] In one embodiment, "truncated LLO", an "N-terminal LLO fragment" or "ALLO" are used interchangeably herein and refer to a non-hemolytic fragment of LLO that comprises a putative PEST sequence. In another embodiment, the terms refer to an LLO fragment that comprises a PEST domain. In another embodiment, the LLO fragment is at least 492 amino acids (AA) long. In another embodiment, the LLO fragment is 492-528 AA long.
[0077] In another embodiment, the LLO fragment consists of about the first 441 AA of the LLO protein. In another embodiment, the LLO fragment consists of about the first 420 AA of LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
[0078] 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 1175. 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.
[0079] In another embodiment, the LLO fragment contains residues of a homologous LLO protein that correspond to one of the above AA ranges. The residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous LLO protein has an insertion or deletion, relative to an LLO protein utilized herein, then the residue numbers can be adjusted accordingly. In another embodiment, the LLO fragment is any other LLO fragment known in the art.
[0080] In one embodiment, an N-terminal LLO protein fragment of the methods and compositions disclosed herein comprises, in one embodiment, SEQ ID No: 3. In another embodiment, the LLO fragment comprises an LLO signal peptide. In another embodiment, the LLO fragment comprises SEQ ID No: 4. In another embodiment, the LLO fragment consists of SEQ ID No: 4. In another embodiment, the LLO fragment consists essentially of SEQ ID No: 4. In another embodiment, the LLO fragment corresponds to SEQ ID No: 4. In another embodiment, the LLO fragment is homologous to SEQ ID No: 4. In another embodiment, the LLO fragment is homologous to a fragment of SEQ ID No: 4. The LLO fragments (ALLO) used in some of the Examples was 416 AA long (exclusive of the signal sequence). In this truncated fragment, 88 residues from the carboxy terminus (which contain the activation domain, including cysteine 484) were removed. It will be understood by those skilled in the art that any ALLO without the activation domain, and in particular without cysteine 484, are suitable for methods and compositions of the present invention. In another embodiment, fusion of a heterologous antigen, such as survivin antigen or fragment thereof to any ALLO enhances cell mediated and anti-tumor immunity of the antigen. In another embodiment, fusion of a heterologous antigen, such as survivin antigen or fragment thereof to a PEST amino acid sequence, such as that provided in SEQ ID NO: 1, enhances cell mediated and anti-tumor immunity of the antigen. In another embodiment, the LLO fragment lacks the cholesterol-binding domain (CBD). LLO mutants lacking the CBD are disclosed in US Patent Serial No. 8,771,702, which is incorporated by reference herein in its entirety.
[0081] It will be appreciated by the skilled artisan that the terms "PEST amino acid sequence," "PEST AA sequence," "PEST sequence-containing polypeptide," "PEST sequence-containing protein," or "PEST-containing peptide or polypeptide" are used interchangeably herein and may encompass a PEST sequence peptide or a fragment of an LLO protein or a fragment of an ActA protein. PEST sequence peptides are known in the art and are described in US Patent Serial No. 7,635,479, and in US Patent Publication Serial No. 2014/0186387, both of which are hereby incorporated in their entirety herein.
[0082] In one embodiment, fusion of an antigen to the PEST sequence of Listeria monocytogenes enhances cell mediated and anti-tumor immunity of the antigen. Thus, fusion of an antigen to PEST- amino acid sequences from other prokaryotic organisms (including but not limited to, other Listeria species) would be expected to have similar effect. In another embodiment, the PEST sequence is embedded within the antigenic protein. Thus, "fusion" refers to an antigenic protein comprising both the antigen and the PEST amino acid sequence either linked at one end of the antigen or embedded within the antigen. PEST sequences derived from other prokaryotic organisms will also enhance immunogenicity of the antigen. In another embodiment, a PEST sequence of prokaryotic organisms can be identified routinely in accordance with methods such as described by Rechsteiner and Roberts (TBS 21:267-271, 1996) for L. monocytogenes. Alternatively, PEST amino acid sequences from other prokaryotic organisms can also be identified based by this method. Other prokaryotic organisms wherein PEST amino acid sequences would be expected include, but are not limited to, other Listeria species. For example, the L. monocytogenes protein ActA contains four such sequences. These are KTEEQPS E VNTGPR (SEQ ID NO: 5), KASVTDTSEGDLDSSMQSADESTPQPLK (SEQ ID NO: 6), KNEE VN AS DFPPPPTDEELR (SEQ ID NO: 7), and RGGIPTS EEFS SLNS GDFTDDENS ETTEEEIDR (SEQ ID NO: 8). Also Streptolysin O from Streptococcus sp. contains a PEST sequence. For example, Streptococcus pyogenes Streptolysin O comprises the PEST sequence KQNTASTETTTTNEQPK (SEQ ID NO: 9) at amino acids 35-51 and Streptococcus equisimilis Streptolysin O comprises the PEST sequence KQNTANTETTTTNEQPK (SEQ ID NO: 10) at amino acids 38-54. Further, it is believed that the PEST sequence can be embedded within the antigenic protein. Thus, for purposes of the present invention, by "fusion" when in relation to PEST sequence fusions, it is meant that the antigenic protein comprises both the antigen and the PEST amino acid sequence either linked at one end of the antigen or embedded within the antigen.
[0083] It will be appreciated by a skilled artisan that the term "immunogenicity" or "immunogenic" may encompass the innate ability of a protein, peptide, nucleic acid, antigen or organism to elicit an immune response in an animal when the protein, peptide, nucleic acid, antigen or organism is administered to the animal. Thus, "enhancing the immunogenicity" in one embodiment, refers to increasing the ability of a protein, peptide, nucleic acid, antigen or organism to elicit an immune response in an animal when the protein, peptide, nucleic acid, antigen or organism is administered to an animal. The increased ability of a protein, peptide, nucleic acid, antigen or organism to elicit an immune response can be measured by, in one embodiment, a greater number of antibodies to a protein, peptide, nucleic acid, antigen or organism, a greater diversity of antibodies to an antigen or organism, a greater number of T-cells specific for a protein, peptide, nucleic acid, antigen or organism, a greater cytotoxic or helper T- cell response to a protein, peptide, nucleic acid, antigen or organism, and the like.
[0084] In another embodiment, the construct or nucleic acid molecule is expressed from an episomal or plasmid vector, with a nucleic acid sequence encoding a PEST sequence-containing polypeptide or a PEST-sequence peptide. In another embodiment, the plasmid is stably maintained in the recombinant Listeria vaccine strain in the absence of antibiotic selection. In another embodiment, the plasmid does not confer antibiotic resistance upon the recombinant Listeria. In another embodiment, the fragment is a functional fragment. In another embodiment, the fragment is an immunogenic fragment.
[0085] It will be appreciated by a skilled artisan that the term "vector" may encompass a plasmid. In another embodiment, the term may encompass an integration vector capable of being transformed into the Listeria host and being incorporated in the Listeria's, chromosome in a manner that allows expression of the genes comprised by the vector. In another embodiment, the term may encompass a non-integration vector that does not integrate in the Listeria's, chromosome but instead is present in the cytoplasm of the Listeria. In another embodiment, the term may encompass a plasmid comprising an integration vector. In another embodiment, the integration vector is a site- specific integration vector.
[0086] The skilled artisan, when equipped with the present disclosure, will readily understand that different transcriptional promoters, terminators, carrier vectors or specific gene sequences (e.g. those in commercially available cloning vectors) can be used successfully in the methods and compositions provided herein. As is contemplated in the present invention, these functionalities are provided in, for example, the commercially available vectors known as the pUC series. In another embodiment, non-essential DNA sequences (e.g. antibiotic resistance genes) are removed. In another embodiment, a commercially available plasmid is used in the present invention. Such plasmids are available from a variety of sources, for example, Invitrogen (La JoUa, CA), Stratagene (La Jolla, CA), Clontech (Palo Alto, CA), or can be constructed using methods well known in the art. Another embodiment is a plasmid such as pCR2.1 (Invitrogen, La Jolla, CA), which is a prokaryotic expression vector with a prokaryotic origin of replication and promoter/regulatory elements to facilitate expression in a prokaryotic organism. In another embodiment, extraneous nucleotide sequences are removed to decrease the size of the plasmid and increase the size of the cassette that can be placed therein. Such methods are well known in the art, and are described in, for example, Sambrook et al. (1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York) and Ausubei et al. (1997, Current Protocols in Molecular Biology, Green & Wiley, New York).
[0087] The LLO protein utilized to construct vaccines disclosed herein has, in another embodiment, the sequence:
[0088] MKKIMLVFITLILVSLPIAQQTEAKDASAFNKENSISSMAPPASPPASPKTPIEKKH ADEIDKYIQGLDYNKNNVLVYHGDAVTNVPPRKGYKDGNEYIVVEKKKKSINQNNADIQ VVNAISSLTYPGALVKANSELVENQPDVLPVKRDSLTLSIDLPGMTNQDNKIVVKNATKSN VNNAVNTLVERWNEKYAQAYPNVSAKIDYDDEMAYSESQLIAKFGTAFKAVNNSLNVNF GAISEGKMQEEVISFKQIYYNVNVNEPTRPSRFFGKAVTKEQLQALGVNAENPPAYISSVA YGRQVYLKLSTNSHSTKVKAAFDAAVSGKSVSGDVELTNIIKNSSFKAVIYGGSAKDEVQI IDGNLGDLRDILKKG ATFNRETPGVPIA YTTNFLKDNELA VIKNNS E YIETTS KA YTDGKINI DHS GG YV AQFNIS WDE VNYDPEGNErVQHKNWS ENNKS KLAHFTS SIYLPGNARNINV YA KECTGLA WEWWRT VIDDRNLPLVKNRNIS rWGTTLYPKYS NKVDNPIE (GenBank Accession No. P13128; SEQ ID NO: 2; nucleic acid sequence is set forth in GenBank Accession No. X15127). The first 25 AA of the proprotein corresponding to this sequence are the signal sequence and are cleaved from LLO when it is secreted by the bacterium. Thus, in this embodiment, the full length active LLO protein is 504 residues long. In another embodiment, the above LLO fragment is used as the source of the LLO fragment incorporated in a vaccine of the present invention.
[0089] In another embodiment, the N-terminal fragment of an LLO protein utilized in compositions and methods disclosed herein has the sequence:
[0090] MKKIMLVFITLILVSLPIAQQTEAKDASAFNKENSISSVAPPASPPASPKTPIEKKHA DEIDKYIQGLDYNKNNVLVYHGDAVTNVPPRKGYKDGNEYrVVEKKKKSINQNNADIQV VNAIS S LT YPGALVKANS ELVENQPD VLP VKRDS LTLS IDLPGMTNQDNKrVVKN ATKSN V NNAVNTLVERWNEKYAQAYSNVSAKIDYDDEMAYSESQLIAKFGTAFKAVNNSLNVNFG AIS EGKMQEE VIS FKQIYYNVNVNEPTRPS RFFGKA VTKEQLQ ALG VN AENPPA YIS S V A Y GRQV YLKLS TNS HS TKVKA AFD AA VS GKS VS GD VELTNIIKNS S FKA VIYGGS AKDE VQII DGNLGDLRDILKKG ATFNRETPGVPIA YTTNFLKDNELA VIKNNS E YIETTS KA YTDGKINI DHS GG YV AQFNIS WDE VNYD (SEQ ID NO: 3).
[0091] In another embodiment, the LLO fragment corresponds to about AA 20-442 of an LLO protein utilized herein.
[0092] In another embodiment, the LLO fragment has the sequence:
[0093] MKKIMLVFITLILVSLPIAQQTEAKDASAFNKENSISSVAPPASPPASPKTPIEKKHA DEIDKYIQGLDYNKNNVLVYHGDAVTNVPPRKGYKDGNEYIVVEKKKKSINQNNADIQV VNAIS S LT YPGALVKANS ELVENQPD VLP VKRDS LTLS IDLPGMTNQDNKrVVKN ATKSN V NNAVNTLVERWNEKYAQAYSNVSAKIDYDDEMAYSESQLIAKFGTAFKAVNNSLNVNFG AIS EGKMQEE VIS FKQIYYNVNVNEPTRPS RFFGKA VTKEQLQ ALG VN AENPPA YIS SV AY GRQV YLKLS TNS HS TKVKA AFD AAVS GKS VS GD VELTNIIKNS S FKA VIYGGS AKDE VQII DGNLGDLRDILKKG ATFNRETPGVPIA YTTNFLKDNELA VIKNNS E YIETTS KA YTD (SEQ ID NO: 4).
[0094] As used herein, "truncated LLO", "tLLO", or "ALLO" refers to a fragment of LLO that comprises the PEST-amino acid sequence. In another embodiment, the term refers to an LLO fragment that comprises a PEST sequence. In another embodiment, "ALLO" refers to 416AA LLO fragment as defined above.
[0095] In another embodiment, the terms truncated LLO, tLLO, or ALLO refer to an LLO fragment that does not contain the activation domain at the amino terminus and does not include cysteine 484. In another embodiment, the terms refer to an LLO fragment that is not hemolytic. In another embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation of the activation domain. In another embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation of cysteine 484. In another embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation at another location. In another embodiment, the LLO is rendered nonhemolytic by a deletion or mutation of the cholesterol binding domain (CBD) as detailed in US Patent No. 8,771,702, which is incorporated by reference herein. [0096] In another embodiment, the recombinant nucleotide encoding a truncated LLO protein comprises the sequence set forth in SEQ ID NO: 11: aaaaaataatgctagtttttattacacttatattag ttagtctaccaattgcgcaacaaactgaagcaaaggatgcatctgcattcaataaagaaaattcaatttcatccatggcaccaccagcatctccgcc tgcaagtcctaagacgccaatcgaaaagaaacacgcggatgaaatcgataagtatatacaaggattggattacaataaaaacaatgtattagtata ccacggagatgcagtgacaaatgtgccgccaagaaaaggttacaaagatggaaatgaatatattgttgtggagaaaaagaagaaatccatcaat caaaataatgcagacattcaagttgtgaatgcaatttcgagcctaacctatccaggtgctctcgtaaaagcgaattcggaattagtagaaaatcaac cagatgttctccctgtaaaacgtgattcattaacactcagcattgatttgccaggtatgactaatcaagacaataaaatagttgtaaaaaatgccacta aatcaaacgttaacaacgcagtaaatacattagtggaaagatggaatgaaaaatatgctcaagcttatccaaatgtaagtgcaaaaattgattatgat gacgaaatggcttacagtgaatcacaattaattgcgaaatttggtacagcatttaaagctgtaaataatagcttgaatgtaaacttcggcgcaatcagt gaagggaaaatgcaagaagaagtcattagttttaaacaaatttactataacgtgaatgttaatgaacctacaagaccttccagatttttcggcaaagc tgttactaaagagcagttgcaagcgcttggagtgaatgcagaaaatcctcctgcatatatctcaagtgtggcgtatggccgtcaagtttatttgaaatt atcaactaattcccatagtactaaagtaaaagctgcttttgatgctgccgtaagcggaaaatctgtctcaggtgatgtagaactaacaaatatcatcaa aaattcttatgaccttcaaagccgtaatttacggaggttccgcaaaagatgaagttcaaatcatcgacggcaacctcggagacttacgcgatattttg aaaaaaggcgctacttttaatcgagaaacaccaggagttcccattgcttatacaacaaacttcctaaaagacaatgaattagctgttattaaaaacaa ctcagaatatattgaaacaacttcaaaagcttatacagatggaaaaattaacatcgatcactctggaggatacgttgctcaattcaacatttcttggga tgaagtaaattatgatctcgag .
[0097] In another embodiment, the recombinant nucleotide has the sequence set forth in SEQ ID NO: 11. In another embodiment, the recombinant nucleotide comprises a sequence that encodes a fragment of an LLO protein.
[0098] In another embodiment, the LLO fragment consists of about the first 441 AA of the LLO protein. In another embodiment, the LLO fragment consists of about the first 420 AA of LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
[0099] 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.
[00100] In another embodiment, an LLO fragment disclosed herein contains residues of a homologous LLO protein that correspond to one of the above AA ranges. The residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous LLO protein has an insertion or deletion, relative to an LLO protein utilized herein, then the residue numbers can be adjusted accordingly. Other LLO fragments are known in the art.
[00101] In another embodiment, the LLO fragment consists of about the first 441 AA of the LLO protein. In another embodiment, the LLO fragment consists of about the first 420 AA of LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the LLO protein.
[00102] In another embodiment, a homologous LLO refers to identity to an LLO sequence (e.g. to one of SEQ ID No: 2-4) of greater than 70%. In another embodiment, a homologous LLO refers to identity to one of SEQ ID No: 2-4 of greater than 72%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 75%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 78%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 80%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 82%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 83%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 85%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 87%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 88%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 90%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 92%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 93%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 95%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 96%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 97%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 98%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of greater than 99%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 2-4 of 100%.
[00103] In one embodiment, an ActA protein comprises the sequence set forth in SEQ ID NO: 12: [00104] MGLNRFMRAMMVVFITANCrriNPDIIFAATDSEDSSLNTDEWEEEKTEEQPSEVN TGPRYETAREVSSRDIKELEKSNKVRNTNKADLIAMLKEKAEKGPNINNNNSEQTENAAIN EEASGADRPAIQVERRHPGLPSDSAAEIKKRRKAIASSDSELESLTYPDKPTKVNKKKVAK ESVADASESDLDSSMQSADESSPQPLKANQQPFFPKVFKKIKDAGKWVRDKIDENPEVKK
ArVDKSAGLIDQLLTKKKSEEVNASDFPPPPTDEELRLALPETPMLLGFNAPATSEPSSFEFP PPPTDEELRLALPETPMLLGFN AP ATS EPS SFEFPPPPTEDELEIIRET AS SLDS S FTRGDLAS L RNAINRHSQNFSDFPPIPTEEELNGRGGRPTSEEFSSLNSGDFTDDENSETTEEEIDRLADLR DRGTGKHSRNAGFLPLNPFAS S PVPS LSPKVS KISDRALIS DITKKTPFKNPS QPLN VFNKKT TTKT VTKKPTP VKT APKLAELPATKPQET VLRENKTPFIEKQ AETNKQS INMPS LPVIQKE A TESDKEEMKPQTEEKMVEESESANNANGKNRSAGIEEGKLIAKSAEDEKAKEEPGNHTTLI LAMLAIG VFS LGAFIKIIQLRKNN . The first 29 AA of the proprotein corresponding to this sequence are the signal sequence and are cleaved from ActA protein when it is secreted by the bacterium. In one embodiment, an ActA polypeptide or peptide comprises the signal sequence, AA 1-29 of SEQ ID NO: 12 above. In another embodiment, an ActA polypeptide or peptide does not include the signal sequence, AA 1-29 of SEQ ID NO: 12 above.
[00105] In one embodiment, a truncated ActA protein comprises an N-terminal fragment of an ActA protein. In another embodiment, a truncated ActA protein is an N-terminal fragment of an ActA protein. In one embodiment, a truncated ActA protein comprises the sequence set forth in SEQ ID NO: 13:
[00106] MRAMMVVFITANCmNPDIIFAATDSEDSSLNTDEWEEEKTEEQPSEVNTGPRYE TAREVSSRDIKELEKSNKVRNTNKADLIAMLKEKAEKGPNINNNNSEQTENAAINEEASGA DRPAIQVERRHPGLPSDS AAEIKKRRKAIAS S DS ELES LT YPDKPTKVNKKKVAKES VAD A SESDLDSSMQSADESSPQPLKANQQPFFPKVFKKIKDAGKWVRDKIDENPEVKKAIVDKSA GLIDQLLTKKKSEEVNASDFPPPPTDEELRLALPETPMLLGFNAPATSEPSSFEFPPPPTDEEL RLALPETPMLLGFN AP ATSEPS S FEFPPPPTEDELEIIRET AS SLDS S FTRGDLAS LRN AINRHS QNFSDFPPIPTEEELNGRGGRP. In another embodiment, the ActA fragment comprises the sequence set forth in SEQ ID NO: 13.
[00107] In another embodiment, a truncated ActA protein comprises the sequence set forth in SEQ ID NO: 14: MGLNRFMRAMMVVFITANCITINPDIIFAATDSEDSSLNTDEWEEEKTEEQPSE VNTGPRYETAREVSSRDIKELEKSNKVRNTNKADLIAMLKEKAEKG.
[00108] In another embodiment, the ActA fragment is any other ActA fragment known in the art.
[00109] In another embodiment, the recombinant nucleotide encoding a truncated ActA protein comprises the sequence set forth in SEQ ID NO: 15:atgcgtgcgatgatggtggttttcattac tgccaattgcattacgattaaccccgacataatatttgcagcgacagatagcgaagattctagtctaaacacagatgaatgggaagaagaaaaaac agaagagcaaccaagcgaggtaaatacgggaccaagatacgaaactgcacgtgaagtaagttcacgtgatattaaagaactagaaaaatcgaa taaagtgagaaatacgaacaaagcagacctaatagcaatgttgaaagaaaaagcagaaaaaggtccaaatatcaataataacaacagtgaacaa actgagaatgcggctataaatgaagaggcttcaggagccgaccgaccagctatacaagtggagcgtcgtcatccaggattgccatcggatagc gcagcggaaattaaaaaaagaaggaaagccatagcatcatcggatagtgagcttgaaagccttacttatccggataaaccaacaaaagtaaata agaaaaaagtggcgaaagagtcagttgcggatgcttctgaaagtgacttagattctagcatgcagtcagcagatgagtcttcaccacaacctttaa aagcaaaccaacaaccatttttccctaaagtatttaaaaaaataaaagatgcggggaaatgggtacgtgataaaatcgacgaaaatcctgaagtaa agaaagcgattgttgataaaagtgcagggttaattgaccaattattaaccaaaaagaaaagtgaagaggtaaatgcttcggacttcccgccaccac ctacggatgaagagttaagacttgctttgccagagacaccaatgcttcttggttttaatgctcctgctacatcagaaccgagctcattcgaatttccac caccacctacggatgaagagttaagacttgctttgccagagacgccaatgcttcttggttttaatgctcctgctacatcggaaccgagctcgttcga atttccaccgcctccaacagaagatgaactagaaatcatccgggaaacagcatcctcgctagattctagttttacaagaggggatttagctagtttg agaaatgctattaatcgccatagtcaaaatttctctgatttcccaccaatcccaacagaagaagagttgaacgggagaggcggtagacca.
[00110] In another embodiment, the recombinant nucleotide has the sequence set forth in SEQ ID NO: 15. In another embodiment, the recombinant nucleotide comprises other sequences that encode a fragment of an ActA protein.
[00111] In another embodiment, "truncated ActA" or "AActA" refers to a fragment of ActA that comprises at least one PEST sequence. In another embodiment, the terms refer to an ActA fragment that comprises more than one PEST sequence.
[00112] The N-terminal ActA protein fragment of methods and compositions disclosed herein comprises, in one embodiment, SEQ ID No: 13. In another embodiment, the ActA fragment comprises an ActA signal peptide. In another embodiment, the ActA fragment comprises SEQ ID No: 14. In another embodiment, the ActA fragment consists approximately of SEQ ID No: 13 or SEQ ID No: 14. In another embodiment, the ActA fragment consists essentially of SEQ ID No: 13 or SEQ ID No: 14. In another embodiment, the ActA fragment corresponds to SEQ ID No: 13 or SEQ ID No: 14. In another embodiment, the ActA fragment is homologous to SEQ ID No: 13 or SEQ ID No: 14. In another embodiment, the ActA fragment is homologous to a fragment of SEQ ID No: 13 or SEQ ID No: 14.
[00113] In another embodiment, a PEST- sequence is another PEST- AA sequence derived from a prokaryotic organism. The PEST- sequence may be other PEST- sequences known in the art.
[00114] In another embodiment, the ActA fragment consists of about the first 100 AA of the ActA protein.
[00115] In another embodiment, the ActA fragment consists of about residues 1-25. In another embodiment, the ActA fragment consists of about residues 1-50. In another embodiment, the ActA fragment consists of about residues 1-75. In another embodiment, the ActA fragment consists of about residues 1-100. In another embodiment, the ActA fragment consists of about residues 1-125. In another embodiment, the ActA fragment consists of about residues 1-150. In another embodiment, the ActA fragment consists of about residues 1-175. In another embodiment, the ActA fragment consists of about residues 1-200. In another embodiment, the ActA fragment consists of about residues 1-225. In another embodiment, the ActA fragment consists of about residues 1-250. In another embodiment, the ActA fragment consists of about residues 1-275. In another embodiment, the ActA fragment consists of about residues 1-300. In another embodiment, the ActA fragment consists of about residues 1-325. In another embodiment, the ActA fragment consists of about residues 1-338. In another embodiment, the ActA fragment consists of about residues 1-350. In another embodiment, the ActA fragment consists of about residues 1-375. In another embodiment, the ActA fragment consists of about residues 1-400. In another embodiment, the ActA fragment consists of about residues 1-450. In another embodiment, the ActA fragment consists of about residues 1-500. In another embodiment, the ActA fragment consists of about residues 1-550. In another embodiment, the ActA fragment consists of about residues 1-600. In another embodiment, the ActA fragment consists of about residues 1-639. In another embodiment, the ActA fragment consists of about residues 30-100. In another embodiment, the ActA fragment consists of about residues 30-125. In another embodiment, the ActA fragment consists of about residues 30- 150. In another embodiment, the ActA fragment consists of about residues 30-175. In another embodiment, the ActA fragment consists of about residues 30-200. In another embodiment, the ActA fragment consists of about residues 30-225. In another embodiment, the ActA fragment consists of about residues 30-250. In another embodiment, the ActA fragment consists of about residues 30-275. In another embodiment, the ActA fragment consists of about residues 30-300. In another embodiment, the ActA fragment consists of about residues 30-325. In another embodiment, the ActA fragment consists of about residues 30-338. In another embodiment, the ActA fragment consists of about residues 30-350. In another embodiment, the ActA fragment consists of about residues 30-375. In another embodiment, the ActA fragment consists of about residues 30-400. In another embodiment, the ActA fragment consists of about residues 30-450. In another embodiment, the ActA fragment consists of about residues 30-500. In another embodiment, the ActA fragment consists of about residues 30-550. In another embodiment, the ActA fragment consists of about residues 1-600. In another embodiment, the ActA fragment consists of about residues 30-604.
[00116] In another embodiment, an ActA fragment disclosed herein contains residues of a homologous ActA protein that correspond to one of the above AA ranges. The residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous ActA protein has an insertion or deletion, relative to an ActA protein utilized herein, then the residue numbers can be adjusted accordingly. Other ActA fragments are known in the art.
[00117] In another embodiment, a homologous ActA refers to identity of an ActA sequence (e.g. to one of SEQ ID No: 12) of greater than 70%. In another embodiment, a homologous ActA refers to identity to one of SEQ ID No: 12 of greater than 72%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 75%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 78%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 80%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 82%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 83%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 85%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 87%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 88%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 greater than 90%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12of greater than 92%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 93%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 95%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 96%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 97%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 98%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of greater than 99%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 12 of 100%.
[00118] It will be appreciated by a skilled artisan that the term "homology," when in reference to any nucleic acid sequence disclosed herein may encompass a percentage of nucleotides in a candidate sequence that are identical with the nucleotides of a corresponding native nucleic acid sequence .
[00119] Homology may be determined by a 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.
[00120] In another embodiment, "homology" refers to identity to a sequence selected from the sequences disclosed herein of greater than 68%. In another embodiment, "homology" refers to identity to a sequence selected from the sequences disclosed herein of greater than 70%. In another embodiment, "homology" refers to identity to a sequence selected from the sequences disclosed herein 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%.
[00121] In another embodiment, LLO protein, Act A protein, or a fragment thereof that are provided for in the present invention need not be that which is set forth exactly in the sequences set forth herein, but rather that other alterations, modifications, or changes can be made that retain the functional characteristics of an LLO or ActA protein fused to an antigen as set forth elsewhere herein. In another embodiment, the present invention utilizes an analog of an LLO protein, ActA protein, or fragment thereof. Analogs differ, in another embodiment, from naturally occurring proteins or peptides by conservative AA sequence differences or by modifications which do not affect sequence, or by both. [00122] It will be appreciated by a skilled artisan that the term "Conservatively modified variants" or "conservative substitution" may encompass substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity or other desired property of the protein, such as antigen affinity and/or specificity. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity.
[00123] In another embodiment, homology is determined 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. J., 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 °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 μg/mL denatured, sheared salmon sperm DNA .
[00124] In one embodiment, a recombinant Listeria strain disclosed herein lacks antibiotic resistance genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises a plasmid comprising a nucleic acid encoding an antibiotic resistance gene. In another embodiment, the recombinant Listeria strain disclosed herein comprises a plasmid that does not encode an antibiotic resistance gene.
[00125] In one embodiment, a recombinant Listeria disclosed herein is capable of escaping the phagolysosome.
[00126] In one embodiment, a polypeptide disclosed herein is a fusion protein comprising an additional polypeptide selected from the group consisting of: non-hemolytic LLO protein or an N- terminal fragment thereof, a PEST sequence, or an ActA protein or a fragment thereof. In another embodiment, an additional polypeptide is fused to the survivin antigen. In another embodiment, an additional polypeptide is functional. In another embodiment, a fragment of an additional polypeptide is immunogenic. In another embodiment, an additional polypeptide is immunogenic.
[00127] In one embodiment, a nucleic acid sequence encoding survivin or an antigenic portion thereof is integrated in frame with LLO in the Listeria chromosome. In another embodiment, an integrated nucleic acid sequence encoding survivin or an antigenic portion thereof is integrated in frame with ActA at the actA locus. In another embodiment, the chromosomal nucleic acid encoding ActA is replaced by a nucleic acid molecule comprising a sequence encoding an antigen. In another embodiment, the antigen is survivin or a fragment thereof. In another embodiment, the survivin is human survivin.
[00128] It will be appreciated by a skilled artisan that the terms "antigenic portion thereof, "a fragment thereof and "immunogenic portion thereof in regard to a protein, peptide or polypeptide are used interchangeably herein and may encompass a protein, polypeptide, peptide, including recombinant forms thereof comprising a domain or segment that leads to the mounting of an immune response when present in, or, in some embodiments, detected by, a host, either alone, or in the context of a fusion protein, as described herein.
[00129] In one embodiment, a nucleic acid molecule disclosed herein comprises a first open reading frame encoding a recombinant polypeptide comprising a survivin or an antigenic fragment thereof. In another embodiment, the recombinant polypeptide further comprises a truncated LLO protein, a truncated ActA protein or PEST sequence peptide fused to a survivin or an antigenic portion thereof. In another embodiment, the truncated LLO protein is an N-terminal LLO or fragment thereof. In another embodiment, the truncated ActA protein is an N-terminal ActA protein or fragment thereof.
[00130] In one embodiment, a nucleic acid molecule disclosed herein further comprises a second open reading frame encoding a metabolic enzyme. In another embodiment, the metabolic enzyme complements a mutation in the chromosome of the recombinant Listeria strain. In another embodiment, the metabolic enzyme encoded by the second open reading frame is an alanine racemase enzyme {dal). In another embodiment, the metabolic enzyme encoded by the second open reading frame is a D-amino acid transferase enzyme (dat). In another embodiment, the Listeria strains disclosed herein comprise a mutation in the endogenous dal/ dat genes. In another embodiment, the Listeria lacks the dal/dat genes. In another embodiment, the Listeria lacks the dal/dat and actA genes.
[00131] In another embodiment, a nucleic acid molecule of the methods and compositions disclosed herein is operably linked to a promoter/regulatory sequence. In another embodiment, the first open reading frame of methods and compositions disclosed herein is operably linked to a promoter/regulatory sequence. In another embodiment, the second open reading frame of methods and compositions disclosed herein is operably linked to a promoter/regulatory sequence. In another embodiment, each of the open reading frames is operably linked to a promoter/regulatory sequence.
[00132] It will be appreciated by a skilled artisan that the term "operably linked" may encompass a transcriptional and translational regulatory nucleic acid that is positioned relative to any coding sequences in such a manner that transcription is initiated. Generally, this will mean that the promoter and transcriptional initiation or start sequences are positioned 5' to the coding region. In another embodiment, the term "operably linked" refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
[00133] It will be appreciated by a skilled artisan that the term "metabolic enzyme" may encompass an enzyme involved in synthesis of a nutrient required by the host bacteria. In one embodiment, the term refers to an enzyme required for synthesis of a nutrient required by the host bacteria. In another embodiment, the term refers to an enzyme involved in synthesis of a nutrient utilized by the host bacteria. In another embodiment, the term refers to an enzyme involved in synthesis of a nutrient required for sustained growth of the host bacteria. In another embodiment, the enzyme is required for synthesis of the nutrient.
[00134] In another embodiment, a recombinant Listeria is an attenuated auxotrophic strain. In another embodiment, a recombinant auxotrophic strain comprises strains described in US Patent No. 8,114,414, which is incorporated by reference herein in its entirety. In one embodiment, the attenuated strain is Lm dal{-)dat{-) (Lmdd). In another embodiment, the attenuated strains is Lm dal(-)dat(-)AactA (LmddA). LmddA is based on a Listeria vaccine vector which is attenuated due to the deletion of virulence gene actA and retains the plasmid for a survivin antigen fusion polypeptide in vivo and in vitro by complementation of dal gene.
[00135] In another embodiment, an attenuated strain disclosed herein is ADXS31-265, which comprises a pAdv 265.5 plasmid (SEQ ID NO: 16). In another embodiment, the attenuated strain is LmAactA. In another embodiment, the attenuated strain is LmAprfA. In another embodiment, the attenuated strain is LmAplcB. In another embodiment, the attenuated strain is LmAplcA. In another embodiment, the attenuated strain is LmAinlA. In another embodiment, the attenuated strain is LmAinlB. In another embodiment, the attenuated strain is LmAinlC. In another embodiment, the strain is the double mutant or triple mutant of any of the above-mentioned strains. In another embodiment, this strain exerts a strong adjuvant effect which is a property of Listeria-based vaccines. In another embodiment, this strain is constructed from the EGD Listeria backbone. In another embodiment, the strain used in the invention is a Listeria strain that expresses a nonhemolytic LLO or a fragment thereof.
[00136] In another embodiment, a Listeria strain disclosed herein is an auxotrophic mutant. In another embodiment, the Listeria strain is deficient in a gene encoding a vitamin synthesis gene. In another embodiment, the Listeria strain is deficient in a gene encoding pantothenic acid synthase.
[00137] In one embodiment, a Listeria strain disclosed herein is deficient in an amino acid (AA) metabolism enzyme. In another embodiment, the generation of auxotrophic strains of Listeria deficient in D-alanine, for example, may be accomplished in a number of ways that are well known to those of skill in the art, including deletion mutagenesis, insertion mutagenesis, and mutagenesis which results in the generation of frameshift mutations, mutations which cause premature termination of a protein, or mutation of regulatory sequences which affect gene expression. In another embodiment, mutagenesis can be accomplished using recombinant DNA techniques or using traditional mutagenesis technology using mutagenic chemicals or radiation and subsequent selection of mutants. In another embodiment, deletion mutants are preferred because of the accompanying low probability of reversion of the auxotrophic phenotype. In another embodiment, mutants of D-alanine which are generated according to the protocols presented herein may be tested for the ability to grow in the absence of D-alanine in a simple laboratory culture assay. In another embodiment, those mutants which are unable to grow in the absence of this compound are selected for further study.
[00138] In another embodiment, in addition to the aforementioned D-alanine associated genes, other genes involved in synthesis of a metabolic enzyme, as provided herein, may be used as targets for mutagenesis of Listeria.
[00139] In one embodiment, a metabolic enzyme complements a mutation in the chromosome of the recombinant bacterial strain. In another embodiment, an endogenous metabolic gene is mutated in the chromosome. In another embodiment, an endogenous metabolic gene is deleted from the chromosome. In another embodiment, a metabolic enzyme is an amino acid metabolism enzyme. In another embodiment, a metabolic enzyme catalyzes a formation of an amino acid used for a cell wall synthesis in the recombinant Listeria strain. In another embodiment, a metabolic enzyme is an alanine racemase enzyme. In another embodiment, a metabolic enzyme is a D-amino acid transferase enzyme.
[00140] It will be appreciated by a skilled artisan that the term "episomal expression vector" may encompass a nucleic acid vector which may be linear or circular, and which is usually double- stranded in form. In one embodiment, an episomal expression vector comprises a gene of interest. In another embodiment, the inserted gene of interest is not interrupted or subjected to regulatory constraints which often occur from integration into cellular DNA. In another embodiment, the presence of the inserted heterologous gene does not lead to rearrangement or interruption of the cell's own important regions. In another embodiment, episomal vectors persist in multiple copies in the bacterial cytoplasm, resulting in amplification of the gene of interest, and, in another embodiment, viral trans-acting factors are supplied when necessary. In another embodiment, in stable transfection procedures, the use of episomal vectors often results in higher transfection efficiency than the use of chromosome-integrating plasmids (Belt, P.B.G.M., et al (1991) Efficient cDNA cloning by direct phenotypic correction of a mutant human cell line (HPRT2) using an Epstein-Barr virus-derived cDNA expression vector. Nucleic Acids Res. 19, 4861-4866; Mazda, O., et al. (1997) Extremely efficient gene transfection into lympho- hematopoietic cell lines by Epstein-Barr virus-based vectors. J. Immunol. Methods 204, 143- 151). In one embodiment, the episomal expression vectors of the methods and compositions as disclosed herein may be delivered to cells in vivo, ex vivo, or in vitro by any of a variety of the methods employed to deliver DNA molecules to cells. The vectors may also be delivered alone or in the form of a pharmaceutical composition that enhances delivery to cells of a subject.
[00141] In one embodiment, an auxotrophic Listeria strain disclosed herein comprises an episomal expression vector comprising a metabolic enzyme that complements the auxotrophy of the auxotrophic Listeria strain. 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. In another embodiment, the episomal expression vector lacks an antibiotic resistance marker. In one embodiment, an antigen, for example, a survivin antigen, of the methods and compositions as disclosed herein is fused to a polypeptide comprising a PEST sequence. In another embodiment, the polypeptide comprising a PEST sequence is a truncated LLO. In another embodiment, the polypeptide comprising a PEST sequence is ActA. [00142] In another embodiment, a Listeria strain disclosed herein is deficient in an AA metabolism enzyme. In another embodiment, the Listeria strain is deficient in a D-glutamic acid synthase gene. In another embodiment, the Listeria strain is deficient in the dat gene. In another embodiment, the Listeria strain is deficient in the dal gene. In another embodiment, the Listeria strain is deficient in the dga gene. In another embodiment, the Listeria strain is deficient in a gene involved in the synthesis of diaminopimelic acid (DAP). In another embodiment, the Listeria strain is deficient in a gene involved in the synthesis of Cysteine synthase A (CysK). In another embodiment, the gene is vitamin-B 12 independent methionine synthase. In another embodiment, the gene is trpA. In another embodiment, the gene is trpB. In another embodiment, the gene is trpE. In another embodiment, the gene is asnB. In another embodiment, the gene is gltD. In another embodiment, the gene is gltB. In another embodiment, the gene is leuA. In another embodiment, the gene is argG. In another embodiment, the gene is thrC. In another embodiment, the Listeria strain is deficient in one or more of the genes described hereinabove.
[00143] In another embodiment, a Listeria strain disclosed herein is deficient in a synthase gene. In another embodiment, the gene is an AA synthesis gene. In another embodiment, the gene is folP. In another embodiment, the gene is dihydrouridine synthase family protein. In another embodiment, the gene is ispD. In another embodiment, the gene is ispF. In another embodiment, the gene is phosphoenolpyruvate synthase. In another embodiment, the gene is hisF. In another embodiment, the gene is hisH. In another embodiment, the gene is flil. In another embodiment, the gene is ribosomal large subunit pseudouridine synthase. In another embodiment, the gene is ispD. In another embodiment, the gene is bifunctional GMP synthase/glutamine amidotransferase protein. In another embodiment, the gene is cobS. In another embodiment, the gene is cobB. In another embodiment, the gene is cbiD. In another embodiment, the gene is uroporphyrin- III C- methyltransferase/ uroporphyrinogen-III synthase. In another embodiment, the gene is cobQ. In another embodiment, the gene is uppS. In another embodiment, the gene is truB. In another embodiment, the gene is dxs. In another embodiment, the gene is mvaS. In another embodiment, the gene is dapA. In another embodiment, the gene is ispG. In another embodiment, the gene is folC. In another embodiment, the gene is citrate synthase. In another embodiment, the gene is argj. In another embodiment, the gene is 3-deoxy-7-phosphoheptulonate synthase. In another embodiment, the gene is indole-3-glycerol-phosphate synthase. In another embodiment, the gene is anthranilate synthase/ glutamine amidotransferase component. In another embodiment, the gene is metiB. In another embodiment, the gene is menaquinone- specific isochorismate synthase. In another embodiment, the gene is phosphoribosylformylglycinamidine synthase I or II. In another embodiment, the gene is phosphoribosylaminoimidazole-succinocarboxamide synthase. In another embodiment, the gene is carB. In another embodiment, the gene is car A. In another embodiment, the gene is thy A. In another embodiment, the gene is mgsA. In another embodiment, the gene is aroB. In another embodiment, the gene is hepB. In another embodiment, the gene is rluB. In another embodiment, the gene is ilvB. In another embodiment, the gene is ilvN. In another embodiment, the gene is alsS. In another embodiment, the gene is fabF. In another embodiment, the gene is fabH. In another embodiment, the gene is pseudouridine synthase. In another embodiment, the gene is pyrG. In another embodiment, the gene is truA. In another embodiment, the gene is pabB. In another embodiment, the gene is an atp synthase gene (e.g. atpC, atpD-2, aptG, atpA-2, etc).
[00144] In another embodiment, the gene is phoP. In another embodiment, the gene is aroA. In another embodiment, the gene is aroC. In another embodiment, the gene is aroD. In another embodiment, the gene is plcB.
[00145] In another embodiment, a Listeria strain disclosed herein is deficient in a peptide transporter. In another embodiment, the gene is ABC transporter/ ATP-binding/permease protein. In another embodiment, the gene is oligopeptide ABC transporter/ oligopeptide-binding protein. In another embodiment, the gene is oligopeptide ABC transporter/ permease protein. In another embodiment, the gene is zinc ABC transporter/ zinc-binding protein. In another embodiment, the gene is sugar ABC transporter. In another embodiment, the gene is phosphate transporter. In another embodiment, the gene is ZIP zinc transporter. In another embodiment, the gene is drug resistance transporter of the EmrB/QacA family. In another embodiment, the gene is sulfate transporter. In another embodiment, the gene is proton-dependent oligopeptide transporter. In another embodiment, the gene is magnesium transporter. In another embodiment, the gene is formate/nitrite transporter. In another embodiment, the gene is spermidine/putrescine ABC transporter. In another embodiment, the gene is Na/Pi-cotransporter. In another embodiment, the gene is sugar phosphate transporter. In another embodiment, the gene is glutamine ABC transporter. In another embodiment, the gene is major facilitator family transporter. In another embodiment, the gene is glycine betaine/L-proline ABC transporter. In another embodiment, the gene is molybdenum ABC transporter. In another embodiment, the gene is techoic acid ABC transporter. In another embodiment, the gene is cobalt ABC transporter. In another embodiment, the gene is ammonium transporter. In another embodiment, the gene is amino acid ABC transporter. In another embodiment, the gene is cell division ABC transporter. In another embodiment, the gene is manganese ABC transporter. In another embodiment, the gene is iron compound ABC transporter. In another embodiment, the gene is maltose/maltodextrin ABC transporter. In another embodiment, the gene is drug resistance transporter of the BcrlCflA family. In another embodiment, the gene is a subunit of one of the above proteins.
[00146] In one embodiment, disclosed herein is a nucleic acid molecule that is used to transform the Listeria in order to arrive at a recombinant Listeria. In another embodiment, the nucleic acid disclosed herein used to transform Listeria lacks a virulence gene. In another embodiment, the nucleic acid molecule is integrated into the Listeria genome and carries a non- functional virulence gene. In another embodiment, the virulence gene is mutated in the recombinant Listeria. In yet another embodiment, the nucleic acid molecule is used to inactivate the endogenous gene present in the Listeria genome. In yet another embodiment, the virulence gene is an actA gene, an inlA gene, and MB gene, an inlC gene, inlJ gene, a plbC gene, a bs/z gene, or a /?r ¾ gene. It is to be understood by a skilled artisan, that the virulence gene can be any gene known in the art to be associated with virulence in the recombinant Listeria.
[00147] In one embodiment, a live attenuated Listeria disclosed herein is a recombinant Listeria. In another embodiment, a recombinant Listeria disclosed herein comprises a mutation of a genomic internalin C (MB) gene. In another embodiment, the recombinant Listeria comprises a mutation or a deletion of a genomic actA gene and a genomic internalin B gene. In one embodiment, translocation of Listeria to adjacent cells is inhibited by the deletion of the actA gene and/or the inlB gene, which are involved in the process, thereby resulting in unexpectedly high levels of attenuation with increased immunogenicity and utility as a strain backbone.
[00148] It will be appreciated by a skilled artisan that the term "attenuation," may encompass a diminution in the ability of the bacterium to cause disease in an animal. In other words, for example the pathogenic characteristics of the attenuated Listeria strain have been lessened compared with wild-type Listeria, although the attenuated Listeria is capable of growth and maintenance in culture. Using as an example the intravenous inoculation of Balb/c mice with an attenuated Listeria, the lethal dose at which 50% of inoculated animals survive (LD50) is preferably increased above the LD50 of wild-type Listeria by at least about 10-fold, more preferably by at least about 100-fold, more preferably at least about 1,000 fold, even more preferably at least about 10,000 fold, and most preferably at least about 100,000-fold. An attenuated strain of Listeria is thus one which does not kill an animal to which it is administered, or is one which kills the animal only when the number of bacteria administered is vastly greater than the number of wild type non-attenuated bacteria which would be required to kill the same animal. An attenuated bacterium should also be construed to mean one which is incapable of replication in the general environment because the nutrient required for its growth is not present therein. Thus, the bacterium is limited to replication in a controlled environment wherein the required nutrient is provided. The attenuated strains disclosed herein are therefore environmentally safe in that they are incapable of uncontrolled replication.
[00149] In yet another embodiment, a Listeria strain disclosed herein is an inlA mutant, an inlB mutant, an inlC mutant, an inlJ mutant, prfA mutant, actA mutant, a dal/dat mutant, a prfA mutant, a plcB deletion mutant, or a double mutant lacking both plcA and plcB. In another embodiment, the Listeria comprises a deletion or mutation of these genes individually or in combination. In another embodiment, the Listeria disclosed herein lack each one of genes. In another embodiment, the Listeria disclosed herein lack at least one and up to ten of any gene provided herein, including the act A, prfA, and dal/dat genes. In another embodiment, the prfA mutant is a D133V prfA mutant.
[00150] In one embodiment, the metabolic gene, the virulence gene, etc. is lacking in a chromosome of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is lacking in the chromosome and in any episomal genetic element of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is lacking in the genome of the virulence strain. In one embodiment, the virulence gene is mutated in the chromosome. In another embodiment, the virulence gene is deleted from the chromosome. In another embodiment, the metabolic gene, the virulence gene, etc. is mutated in a chromosome of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is mutated in the chromosome and in any episomal genetic element of the Listeria strain. In another embodiment, the metabolic gene, virulence gene, etc. is mutated in the genome of the virulence strain. In another embodiment, the virulence gene is deleted from the chromosome.
[00151] In one embodiment, a recombinant Listeria strain disclosed herein is attenuated. In another embodiment, the recombinant Listeria lacks the actA virulence gene. In another embodiment, the recombinant Listeria lacks the prfA virulence gene. In another embodiment, the recombinant Listeria lacks the inlB gene. In another embodiment, the recombinant Listeria lacks both, the actA and inlB genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA gene. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous inlB gene. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous inlC gene. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA and inlB genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA, MB, and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA, inlB, and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation of the endogenous actA, MB, and inlC genes. In another embodiment, the recombinant Listeria strain disclosed herein comprises an inactivating mutation in any single gene or combination of the following genes: actA, dal, dat, MB, inlC, prfA, plcA, plcB.
[00152] It will be appreciated by a skilled artisan that the term "mutation" and grammatical equivalents thereof, include any type of mutation or modification to the sequence (nucleic acid or amino acid sequence), and includes a deletion mutation, a truncation, an inactivation, a disruption, or a translocation. These types of mutations are readily known in the art.
[00153] In one embodiment, in order to select for auxotrophic bacteria comprising a plasmid encoding a metabolic enzyme or a complementing gene provided herein, transformed auxotrophic bacteria are grown on a media that will select for expression of the amino acid metabolism gene or the complementing gene. In another embodiment, a bacteria auxotrophic for D-glutamic acid synthesis is transformed with a plasmid comprising a gene for D-glutamic acid synthesis, and the auxotrophic bacteria will grow in the absence of D-glutamic acid, whereas auxotrophic bacteria that have not been transformed with the plasmid, or are not expressing the plasmid encoding a protein for D-glutamic acid synthesis, will not grow. In another embodiment, a bacterium auxotrophic for D- alanine synthesis will grow in the absence of D-alanine when transformed and expressing the plasmid disclosed herein if the plasmid comprises an isolated nucleic acid encoding an amino acid metabolism enzyme for D-alanine synthesis. Such methods for making appropriate media comprising or lacking necessary growth factors, supplements, amino acids, vitamins, antibiotics, and the like are well known in the art, and are available commercially (Becton-Dickinson, Franklin Lakes, NJ).
[00154] In another embodiment, once the auxotrophic bacteria comprising the plasmid disclosed herein have been selected on appropriate media, the bacteria are propagated in the presence of a selective pressure. Such propagation comprises growing the bacteria in media without the auxotrophic factor. The presence of the plasmid expressing an amino acid metabolism enzyme in the auxotrophic bacteria ensures that the plasmid will replicate along with the bacteria, thus continually selecting for bacteria harboring the plasmid. The skilled artisan, when equipped with the present disclosure and methods herein will be readily able to scale-up the production of the Listeria vaccine vector by adjusting the volume of the media in which the auxotrophic bacteria comprising the plasmid are growing.
[00155] The skilled artisan will appreciate that, in another embodiment, other auxotroph strains and complementation systems are adopted for the use with the methods and compositions disclosed herein.
[00156] In one embodiment, a recombinant Listeria strain disclosed herein expresses a recombinant polypeptide. In another embodiment, a recombinant Listeria strain comprises a plasmid that encodes a recombinant polypeptide. In another embodiment, a recombinant nucleic acid disclosed herein is in a plasmid in the recombinant Listeria strain provided herein. In another embodiment, the plasmid is an episomal plasmid that does not integrate into the recombinant Listeria strain's chromosome. In another embodiment, the plasmid is an integrative plasmid that integrates into the Listeria strain's chromosome. In another embodiment, the plasmid is a multicopy plasmid.
[00157] In one embodiment, a recombinant Listeria strain disclosed herein comprises a nucleic acid molecule encoding a fusion protein comprising a tumor associated antigen. In one embodiment, a tumor associated antigen comprises a survivin polypeptide or a fragment thereof. In another embodiment, a survivin polypeptide comprises a human survivin. In another embodiment, a survivin polypeptide is a human survivin. In another embodiment, a survivin polypeptide comprises a mouse survivin. In one embodiment, the recombinant Listeria strain disclosed herein comprises a nucleic acid molecule encoding a fusion protein comprising a survivin protein or a fragment thereof. In another embodiment, survivin or an antigenic portion thereof comprises a tumor specific antigen.
[00158] In one embodiment, the human survivin protein comprises the amino sequence as set forth in SEQ ID NO: 17 GAPTLPPAWQPFLKDHRISTFKNWPF
LEGC AC APERM AE AGFIHCPTENEPDLAQCFFCFKELEGWEPDDDPIEEHKKHS S GC AFLS VKKQFEELTLGEFLKLDRERAKNKIAKETNNKKKEFEETAKKVRRAIEQLAAMD. In another embodiment, the survivin protein is a homologue of SEQ ID No: 17. In another embodiment, the survivin protein is a variant of SEQ ID No: 17. In another embodiment, the survivin protein is a fragment of SEQ ID No: 17.
[00159] In one embodiment, the mouse survivin protein comprises the amino sequence as set forth in SEQ ID NO: 18 MGAPALPQIWQLYLKNYRIATFKNWPF
LEDCACAPERMAEAGFIHCPTENEPDLAQCFFCFKELEGWEPDDNPIEEHRKHSPGCAFLT VKKQMEELTVSEFLKLDRQRAKNKIAKETNNKQKEFEETAKTTRQSIEQLAA. In another embodiment, the survivin protein is a homologue of SEQ ID No: 18. In another embodiment, the survivin protein is a variant of SEQ ID No: 17. In another embodiment, the survivin protein is a fragment of SEQ ID No: 18.
[00160] In another embodiment, a survivin fragment disclosed herein consists of about residues 1- 25. In another embodiment, the survivin fragment consists of about residues 1-50. In another embodiment, the survivin fragment consists of about residues 1-75. In another embodiment, the survivin fragment consists of about residues 1-100. In another embodiment, the survivin fragment consists of about residues 1-125. In another embodiment, the survivin fragment consists of full length survivin. In another embodiment, the survivin fragment is lacking a signal sequence.
[00161] In another embodiment, a survivin disclosed herein fragment contains residues of a homologous survivin protein that correspond to one of the above AA ranges. The residue numbers need not, in another embodiment, correspond exactly with the residue numbers enumerated above; e.g. if the homologous survivin protein has an insertion or deletion, relative to an survivin protein utilized herein, then the residue numbers can be adjusted accordingly. Other survivin fragments are known in the art.
[00162] In another embodiment, a homologous survivin has identity to a survivin sequence (e.g. to one of SEQ ID No: 17 or SEQ ID No: 18) of greater than 70%. In another embodiment, a homologous survivin refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 72%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 75%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 78%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 80%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 82%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 83%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 85%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 87%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 88%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 90%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 92%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 93%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 95%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 96%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 97%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 98%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of greater than 99%. In another embodiment, a homologous refers to identity to one of SEQ ID No: 17 or SEQ ID No: 18 of 100%.
[00163] In another embodiment, the human survivin protein is encoded by a nucleotide molecule having the sequence SEQ ID NO: 19: ggtgccccgacgttgccccctgcctggcagccctttctc aaggaccaccgcatctctacattcaagaactggcccttcttggagggctgcgcctgcgccccggagcggatggccgaggctggcttcatccact gccccactgagaacgagccagacttggcccagtgtttcttctgcttcaaggagctggaaggctgggagccagatgacgaccccatagaggaac ataaaaagcattcgtccggttgcgctttcctttctgtcaagaagcagtttgaagaattaacccttggtgaatttttgaaactggacagagaaagagcc aagaacaaaattgcaaaggaaaccaacaataagaagaaagaatttgaggaaactgcgaagaaagtgcgccgtgccatcgagcagctggctgc catggattga (SEQ ID No: 19; GenBank Accession No. CR541740). In another embodiment, the survivin protein is encoded by a homologue of SEQ ID No: 19. In another embodiment, the survivin protein is encoded by a variant of SEQ ID No: 19. In another embodiment, the survivin protein is encoded by an isomer of SEQ ID No: 19. In another embodiment, the survivin protein is encoded by a fragment of SEQ ID No : 19.
[00164] In another embodiment, the survivin protein is a mouse survivin protein is encoded by a nucleotide molecule having the sequence SEQ ID NO: 20: tcgagggagctccggcg ctgccccagatctggcagctgtacctcaagaactaccgcatcgccaccttcaagaactggcccttcctggaggactgcgcctgcgccccagagc gaatggcggaggctggcttcatccactgccctaccgagaacgagcctgatttggcccagtgttttttctgctttaaggaattggaaggctgggaac ccgatgacaacccgatagaggagcatagaaagcactcccctggctgcgccttcctcactgtcaagaagcagatggaagaactaaccgtcagtg aattcttgaaactggacagacagagagccaagaacaaaattgcaaaggagaccaacaacaagcaaaaagagtttgaagagactgcaaagact acccgtcagtcaattgagcagctggctgcctaa. (SEQ ID No: 20; GenBank Accession No. NM_009689). In another embodiment, the survivin protein is encoded by a homologue of SEQ ID No: 20. In another embodiment, the survivin protein is encoded by a variant of SEQ ID No: 20. In another embodiment, the survivin protein is encoded by an isomer of SEQ ID No: 20. In another embodiment, the survivin protein is encoded by a fragment of SEQ ID No: 20.
[00165] In another embodiment, the survivin protein of the methods and compositions disclosed herein is a human survivin protein (SEQ ID NO: 19). In another embodiment, the survivin protein is a mouse survivin protein (SEQ ID NO: 20). In another embodiment, the survivin protein is a rat survivin protein. In another embodiment, the survivin protein is a primate survivin protein. In another embodiment, the survivin protein is a canine survivin protein. In another embodiment, the survivin protein is a survivin protein of human or other animal species or combinations thereof known in the art. In another embodiment, a nucleic acid sequence encoding a canine survivin protein is set forth in SEQ ID NO: 21:
[00166]
1 attaaccgcc agatttgaat cgcgggaccc gttggcagag gtggcggcgg cggcatgggt
61 gccccgacgt tgccccctgc ctggcagccc tttctcaagg accaccgcat ctctacattc 121 aagaactggc ccttcttgga gggctgcgcc tgcaccccgg accggatggc agaggccggc
181 ttcatccact gtcccactga gaacgagcca gacttggccc agtgtttctt ctgcttcaag
241 gagctggaag gctgggagcc agatgatgac cctatagagg agcataaaaa acattcatct
301 ggttgtgctt tcctttctgt caagaagcag tttgaagaat taacccttgg tgaatttttg
361 aaactggaca gagaaagagc caagaacaaa attgcaaagg aaaccaacaa taagaagaaa
421 gaatttgagg aaactgcgaa gaaagtgcgc cgtgccatcg agcagctggc tgccatggat
481 tgaggcctct ggccggagct gcctggtccc agagtggctg caccacttcc agggtttatt
541 ccctggtgcc accagccttc ctgtgggccc cttagcaatg tcttaggaaa ggagatcaac
601 attttcaaat tagatgtttc aactgtgctc ttgttttgtc ttgaaagtgg caccagaggt
661 gcttctgcct gtgcagcggg tgctgctggt aacagtggct gcttctctct ctctctctct
721 ttttgggggc tcatttttgc tgttttgatt cccgggctta ccaggtgaga agtgagggag
781 gaagaaggca gtgtcccttt tgctagagct gacagctttg ttcgcgtggg cagagccttc
841 cacagtgaat gtgtctggac ctcatgttgt tgaggctgtc acagtcctga gtgtggactt
901 ggcaggtgcc tgttgaatct gagctgcagg ttccttatct gtcacacctg tgcctcctca
961 gaggacagtt tttttgttgt tgtgtttttt tgtttttttt tttttggtag atgcatgact
1021 tgtgtgtgat gagagaatgg agacagagtc cctggctcct ctactgttta acaacatggc
1081 tttcttattt tgtttgaatt gttaattcac agaatagcac aaactacaat tcaaactaag
1141 cacaaagcca ttctaagtca ttggggaaac ggggtgaact tcaggtggat gaggagacag
1201 aatagagtga taggaagcgt tggcagatac tccttttgcc actgctgtgt gattagacag
1261 gcccagtgag ccgcggggca catgctggcc gctcctccct cagaaaaagg cagtggccta
1321 aatccttttt aaatgacttg gctcgatgct gtgggggact ggctgggctg ctgcaggccg
1381 tgtgtctgtc agcccaacct tcacatctgt cacgttctcc acacggggga gagacgcagt
1441 ccgcccaggt ccccgctttc tttggaggca gcagctcccg cagagctgaa gtctggcgta
1501 agatgatgga tttgattccg ccctcctgcc ctgtcataga gctgcagggt ggattgttac
1561 agcttcgctg gaaacctctg gaggtcatac tcggctgttc ctgagaaata aaaagcctgt
1621 catttcaaac (SEQ ID NO: 21). In another embodiment, a nucleic acid encoding a canine survivin protein is set forth in GenBank Accession No. AB095108.1. In another embodiment, a nucleic acid encoding a survivin protein is a homolog of SEQ ID NO: 21. In another embodiment, the nucleic acid sequence is a variant of SEQ ID NO: 21.
[00167] In one embodiment, an amino acid sequence encoding a canine survivin protein is set forth in SEQ ID NO: 22:
[00168] MGAPTLPPAWQPFLKDHRI STFKNWPFLEGCACTPDRMAEAGF IHCPTENEPDLAQCFFCFKELEGWEPDDD PIEEHKKHSSGCAFLSVKKQFEELTLGEFLKLDRERAKNKIAKETNNKKKEFEETAKKVRRAIEQLAAMD
[00169] In another embodiment, a nucleic acid encoding a canine survivin protein is set forth in GenBank Accession No. BAC22748.2. In another embodiment, an amino acid sequence encoding a survivin protein is a homolog of SEQ ID NO: 22. In another embodiment, the amino acid sequence is a variant of SEQ ID NO: 22.
[00170] In one embodiment, an amino acid sequence of survivin disclosed herein has 99% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 95% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 90% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 85% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 80% identity with that of human survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has at least 75% identity with that of human survivin amino acid sequence.
[00171] In one embodiment, an amino acid sequence of survivin disclosed herein has 99% identity with that of mouse survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 95% identity with that of mouse survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 90% identity with that of mouse survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 85% identity with that of mouse survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 80% identity with that of mouse survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 75% identity with that of mouse survivin amino acid sequence.
[00172] In one embodiment, an amino acid sequence of survivin disclosed herein has 99% identity with that of a canine survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 95% identity with that of canine survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 90% identity with that of canine survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 85% identity with that of canine survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 80% identity with that of canine survivin amino acid sequence. In another embodiment, the amino acid sequence of survivin has 75% identity with that of canine survivin amino acid sequence.
[00173] In another embodiment, a survivin protein that is the source of a survivin antigen for use in the methods and compositions as disclosed herein is a Baculoviral IAP Repeat Containing 5 Protein. In another embodiment, the survivin protein is API4 protein. In another embodiment, the survivin protein is EPR-1 protein. In another embodiment, the survivin protein is Apoptosis Inhibitor 4 protein. In another embodiment, the survivin protein is IAP4 protein. In another embodiment, the survivin protein is Survivin Variant 3 Alpha.
[00174] In one embodiment, a survivin protein or fragment thereof of the methods and compositions disclosed herein does not include a signal sequence thereof. In another embodiment, omission of the signal sequence enables the survivin fragment to be successfully secreted in Listeria.
[00175] In one embodiment, nucleic acids encoding the recombinant polypeptides disclosed herein also encode a signal peptide or sequence. In another embodiment, the fusion protein of methods and compositions disclosed herein comprises an LLO signal sequence. In one embodiment, a heterologous antigen may be expressed in Listeria through the use of a signal sequence, such as a Listerial signal sequence, for example, the hemolysin signal sequence or the ActA signal sequence. Alternatively, for example, foreign genes can be expressed downstream from a L. monocytogenes promoter without creating a fusion protein. In another embodiment, the signal peptide is bacterial (Listerial or non-Listerial). In one embodiment, the signal peptide is native to the bacterium. In another embodiment, the signal peptide is foreign to the bacterium. In another embodiment, the signal peptide is a signal peptide from Listeria monocytogenes, such as a secAl signal peptide. In another embodiment, the signal peptide is an Usp45 signal peptide from Lactococcus lactis, or a Protective Antigen signal peptide from Bacillus anthracis. In another embodiment, the signal peptide is a secA2 signal peptide, such the p60 signal peptide from Listeria monocytogenes. In addition, the recombinant nucleic acid molecule optionally comprises a third polynucleotide sequence encoding p60, or a fragment thereof. In another embodiment, the signal peptide is a Tat signal peptide, such as a B. subtilis Tat signal peptide (e.g., PhoD). In one embodiment, the signal peptide is in the same translational reading frame encoding the recombinant polypeptide.
[00176] In another embodiment, the survivin protein is a splice variant 1 survivin protein. In another embodiment, the survivin protein is a splice variant 2 survivin protein. In another embodiment, the survivin protein is a splice variant 3 survivin protein. In another embodiment, the survivin protein is a transcript variant 1 survivin protein. In another embodiment, the survivin protein is a transcript variant 2 survivin protein. In another embodiment, the survivin protein is a transcript variant 3 survivin protein. In another embodiment, the survivin protein is a transcript variant 4 survivin protein. In another embodiment, the survivin protein is a transcript variant 5 survivin protein. In another embodiment, the survivin protein is a transcript variant 6 survivin protein. In another embodiment, the survivin protein is a transcript variant 7 survivin protein.
[00177] In another embodiment, the survivin protein that is the source of a survivin peptide of methods and compositions as disclosed herein is a human survivin protein. In another embodiment, the survivin protein is a primate survivin protein. In another embodiment, the survivin protein is a canine survivin protein. Survivin proteins of other species are known in the art.
[00178] It will be appreciated by a skilled artisan that the term "homologue" may encompass a nucleic acid or amino acid sequence which shares a certain percentage of sequence identity with a particular nucleic acid or amino acid sequence. In one embodiment, a sequence useful in the composition and methods as disclosed herein may be a homologue of a particular LLO sequence or N-terminal fragment thereof. In another embodiment, a sequence useful in the composition and methods as disclosed herein may be a homologue of an antigenic polypeptide, which in one embodiment, is survivin or a functional fragment thereof. In one embodiment, a homolog of a polypeptide and, in one embodiment, the nucleic acid encoding such a homolog, disclosed herein maintains the functional characteristics of the parent polypeptide. For example, in one embodiment, a homolog of an antigenic polypeptide disclosed herein maintains the antigenic characteristic of the parent polypeptide. In another embodiment, a sequence useful in the composition and methods as disclosed herein may be a homologue of any sequence described herein. In one embodiment, a homologue shares at least 70% identity with a particular sequence.
In another embodiment, a homologue shares at least 72% identity with i a . particular sequence. In another embodiment, a homologue shares at least 75% identity with a particular sequence. In another embodiment, a homologue shares at least 78% identity with a particular sequence. In another embodiment, a homologue shares at least 80% identity with a particular sequence. In another embodiment, a homologue shares at least 82% identity with a particular sequence. In another embodiment, a homologue shares at least 83% identity with a particular sequence. In another embodiment, a homologue shares at least 85% identity with a particular sequence. In another embodiment, a homologue shares at least 87% identity with a particular sequence. In another embodiment, a homologue shares at least 88% identity with a particular sequence. In another embodiment, a homologue shares at least 90% identity with a particular sequence. In another embodiment, a homologue shares at : least 90.5% ) identity with a particular sequence. In another embodiment, a homologue shares at least 91% identity with a particular sequence. In another embodiment, a homologue shares at least 92% identity with a particular sequence. In another embodiment, a homologue shares at least 93% identity with a particular sequence. In another embodiment, a homologue shares at least 95% identity with a particular sequence. In another embodiment, a homologue shares at least 96% identity with a particular sequence. In another embodiment, a homologue shares at least 97% identity with a particular sequence. In another embodiment, a homologue shares at least 98% identity with a particular sequence. In another embodiment, a homologue shares at least 99% identity with a particular sequence. In another embodiment, a homologue shares 100% identity with a particular sequence.
[00179] It will be appreciated by a skilled artisan that the term "antigenic polypeptide" may encompass a polypeptide, peptide or recombinant peptide as described hereinabove that is foreign to a host and leads to the mounting of an immune response when present in, or, in some embodiments, detected by, a host.
[00180] In one embodiment, an "antigenic polypeptide" may encompass a polypeptide, a peptide, a recombinant polypeptide, or a recombinant peptide as described herein that is processed and presented on MHC class I and/or class II molecules present in a subject's cells leading to the mounting of an immune response when present in, or, in another embodiment, detected by, the host. In one embodiment, the antigen may be foreign to the host. In another embodiment, the antigen might be present in the host but the host does not elicit an immune response against it because of immunologic tolerance.
[00181] In one embodiment, it is to be understood that a homolog of any of the sequences disclosed herein and/or described herein are considered to be encompassed by the present invention.
[00182] In another embodiment, a recombinant Listeria strain of the methods and compositions as disclosed herein comprise a nucleic acid molecule operably integrated into the Listeria genome as an open reading frame with an endogenous ActA sequence. In another embodiment, a recombinant Listeria strain of the methods and compositions disclosed herein comprise an episomal expression vector comprising a nucleic acid molecule encoding fusion protein comprising an antigen fused to an ActA or a truncated ActA. In one embodiment, the expression and secretion of the antigen is under the control of an ActA promoter and ActA signal sequence and it is expressed as fusion to 1- 233 amino acids of ActA (truncated ActA or tActA). In another embodiment, the truncated ActA consists of the first 390 amino acids of the wild type ActA protein as described in US Patent Serial No. 7,655,238, which is incorporated by reference herein in its entirety. In another embodiment, the truncated ActA is an ActA-NlOO or a modified version thereof (referred to as ActA-NlOO*) in which a PEST motif has been deleted and containing the nonconservative QDNKR substitution as described in US Patent Publication Serial No. 2014/0186387.
[00183] In one embodiment, an attenuated auxotrophic Listeria vaccine strain is based on a Listeria vaccine vector which is attenuated due to the deletion of virulence gene actA and retains the plasmid for survivin expression in vivo and in vitro by complementation of dal gene. In one embodiment, the Listeria strain expresses and secretes a survivin protein fused to the first 441 amino acids of listeriolysin O (LLO). In another embodiment, the Listeria is a dal/dat/actA Listeria having a mutation in the dal, dat and actA endogenous genes. In another embodiment, the mutation is a deletion, a truncation or an inactivation of the mutated genes. In another embodiment, the expression and secretion of the fusion protein LLO-survivin from the attenuated auxotrophic strain disclosed herein that expresses a survivin antigen/LLO fusion protein after an in vitro passage is comparable to that of the Lm-LLO- survivin in TCA precipitated cell culture supernatants after 8 hours of in vitro growth.
[00184] In another embodiment, the dal/dat/actA mutant Listeria strain is highly attenuated and has a better safety profile than previous Listeria vaccine generation, as it is more rapidly cleared from the spleens of the immunized mice. In another embodiment, the dal/dat/actA mutant Listeria strain results in a longer delay of tumor onset in transgenic animals than Listeria vaccines based on more virulent antibiotic resistant strains (see US Publication No. 2011/0142791, which is incorporated by reference herein in its entirety). In another embodiment, the dal/dat/actA mutant Listeria strain causes a significant decrease in intra-tumoral T regulatory cells (Tregs). In another embodiment, the lower frequency of Tregs in tumors treated with LmddA vaccines result in an increased intratumoral CD8/Tregs ratio, suggesting that a more favorable tumor microenvironment can be obtained after immunization with LmddA vaccines.
[00185] In another embodiment, the terms "LmddA", "LmAddA" or dal/dat/actA mutant Listeria are used interchangeably herein.
[00186] In one embodiment, the present invention provides a recombinant polypeptide comprising an N-terminal fragment of an LLO protein fused to a survivin protein or to a fragment thereof. In one embodiment, the present invention provides a recombinant polypeptide consisting of an N- terminal fragment of an LLO protein fused to a survivin protein or fused to a fragment thereof. [00187] In one embodiment, the present invention provides a recombinant polypeptide comprising an N-terminal fragment of an ActA protein fused to a survivin protein or to a fragment thereof. In another embodiment, the present invention provides a recombinant polypeptide consisting of an N- terminal fragment of an ActA protein fused to a survivin protein or fused to a fragment thereof.
[00188] In one embodiment, a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are fused directly to one another. In another embodiment, the genes encoding a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are fused directly to one another. In another embodiment, a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are operably attached via a linker peptide. In another embodiment, a PEST-containing polypeptide or peptide and survivin antigen or immunogenic fragment thereof are attached via a heterologous peptide. In another embodiment, a PEST-containing polypeptide or peptide is N-terminal to the survivin antigen or immunogenic fragment thereof. In another embodiment, a PEST-containing polypeptide or peptide is expressed and used alone, i.e., in unfused form. In another embodiment, a PEST-containing polypeptide or peptide is the N-terminal-most portion of the fusion protein. In another embodiment, a PEST- containing polypeptide or peptide is an N-terminal LLO protein fragment. In another embodiment, a truncated LLO is truncated at the C-terminal to arrive at an N-terminal LLO. In another embodiment, a PEST-containing polypeptide or peptide is the tLLO. In another embodiment, a PEST-containing polypeptide or peptide is an ActA polypeptide or a fragment thereof. In another embodiment, a PEST-containing polypeptide or peptide is an N-terminal fragment of ActA. In another embodiment, a PEST-containing amino acid sequence is a PEST peptide.
[00189] In one embodiment, the present invention provides a recombinant polypeptide comprising survivin or a fragment thereof fused to a PEST amino acid sequence. In another embodiment, a recombinant polypeptide comprises survivin or a fragment thereof fused to 1-2 PEST amino acid sequences. In another embodiment, a recombinant polypeptide comprises survivin or a fragment thereof fused to 2-3 PEST amino acid sequences. In another embodiment, a recombinant polypeptide comprises survivin or a fragment thereof fused to 3-4 PEST amino acid sequences.
[00190] It will be appreciated by a skilled artisan that the terms "polypeptide," "peptide" and "recombinant peptide" may encompass, in another embodiment, a peptide or polypeptide of any length. In another embodiment, a peptide or recombinant peptide as disclosed herein has one of the lengths enumerated above for an HMW-MAA fragment.
[00191] In one embodiment, the term "peptide" refers to native peptides (either degradation products, synthetically synthesized peptides or recombinant peptides) and/or peptidomimetics (typically, synthetically synthesized peptides), such as peptoids and semipeptoids which are peptide analogs, which may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells. Such modifications include, but are not limited to N terminus modification, C terminus modification, peptide bond modification, including, but not limited to, CH2-NH, CH2-S, CH2-S=0, 0=C-NH, CH2-0, CH2-CH2, S=C- NH, CH=CH or CF=CH, backbone modifications, and residue modification. Methods for preparing peptidomimetic compounds are well known in the art and are specified, for example, in Quantitative Drug Design, C.A. Ramsden Gd., Chapter 17.2, F. Choplin Pergamon Press (1992), which is incorporated by reference as if fully set forth herein. Further details in this respect are provided hereinunder.
[00192] Peptide bonds (-CO-NH-) within the peptide may be substituted, for example, by N- methylated bonds (-N(CH3)-CO-), ester bonds (-C(R)H-C-0-0-C(R)-N-), ketomethylen bonds (- CO-CH2-), *-aza bonds (-NH-N(R)-CO-), wherein R is any alkyl, e.g., methyl, carba bonds (-CH2- NH-), hydroxyethylene bonds (-CH(OH)-CH2-), thioamide bonds (-CS-NH-), olefinic double bonds (-CH=CH-), retro amide bonds (-NH-CO-), peptide derivatives (-N(R)-CH2-CO-), wherein R is the "normal" side chain, naturally presented on the carbon atom.
[00193] These modifications can occur at any of the bonds along the peptide chain and even at several (2-3) at the same time. Natural aromatic amino acids, Trp, Tyr and Phe, may be substituted for synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
[00194] In addition to the above, the peptides disclosed herein may also include one or more modified amino acids or one or more non-amino acid monomers (e.g. fatty acids, complex carbohydrates etc).
[00195] 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.
[00196] In another embodiment, a construct or nucleic acid molecule disclosed herein 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 virus recombinants expressing either Listeria monocytogenes partial listeriolysin or Brucella abortus ribosomal L7/L12 protein. Vet Microbiol 2005, 109(1-2): 11-7); and Jiang 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 another embodiment, a temperature sensitive plasmid is used to select the recombinants.
[00197] It will be appreciated by a skilled artisan that the term "recombination site" or "site- specific recombination site" may encompass a sequence of bases in a nucleic acid molecule that is recognized by a recombinase (along with associated proteins, in some cases) that mediates exchange or excision of the nucleic acid segments flanking the recombination sites. The recombinases and associated proteins are collectively referred to as "recombination proteins" see, e.g., Landy, A., (Current Opinion in Genetics & Development) 3:699-707; 1993). [00198] In another embodiment, a construct or nucleic acid molecule disclosed herein is integrated into the Listerial chromosome using transposon insertion. Techniques for transposon 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.
[00199] In another embodiment, the construct or nucleic acid molecule 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 Bacterid 2002; 184(15): 4177-86). In certain embodiments of this method, an integrase gene and attachment site of a bacteriophage (e.g. U153 or PSA listeriophage) 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. In another embodiment, the present invention further comprises a phage based chromosomal integration system for clinical applications, where a host strain that is auxotrophic for essential enzymes, including, but not limited to, d-alanine racemase can be used, for example Lm dal{-)dat{-). In another embodiment, in order to avoid a "phage curing step," a phage integration system based on PSA is used (Lauer, et al., 2002 J Bacterid, 184:4177-4186). This requires, in another embodiment, continuous selection by antibiotics to maintain the integrated gene. Thus, in another embodiment, the current invention enables the establishment of a phage based chromosomal integration system that does not require selection with antibiotics. Instead, an auxotrophic host strain can be complemented.
[00200] It will be appreciated by a skilled artisan that the term "phage expression vector" or "phagemid" may encompass any phage-based recombinant expression system for the purpose of expressing a nucleic acid sequence of the methods and compositions as disclosed herein in vitro or in vivo, constitutively or inducibly, in any cell, including prokaryotic, yeast, fungal, plant, insect or mammalian cell. A phage expression vector typically can both reproduce in a bacterial cell and, under proper conditions, produce phage particles. The term includes linear or circular expression systems and encompasses both phage-based expression vectors that remain episomal or integrate into the host cell genome. [00201] In another embodiment, conjugation is used to introduce genetic material and/or plasmids into bacteria. Methods for conjugation are well known in the art, and are described, for example, in Nikodinovic J et al (A second generation snp-derived Escherichia coli-Streptomyces shuttle expression vector that is generally transferable by conjugation. Plasmid. 2006 Nov;56(3):223-7) and Auchtung JM et al (Regulation of a Bacillus subtilis mobile genetic element by intercellular signaling and the global DNA damage response. Proc Natl Acad Sci U S A. 2005 Aug 30; 102(35): 12554-9).
[00202] Antibiotic resistance genes are used in the conventional selection and cloning processes commonly employed in molecular biology and vaccine preparation. Antibiotic resistance genes contemplated in the present invention include, but are not limited to, gene products that confer resistance to ampicillin, penicillin, methicillin, streptomycin, erythromycin, kanamycin, tetracycline, cloramphenicol (CAT), neomycin, hygromycin, gentamicin and others well known in the art.
[00203] Plasmids and other expression vectors useful in the present invention are described elsewhere herein, and can include such features as a promoter/regulatory sequence, an origin of replication for gram negative and gram positive bacteria, an isolated nucleic acid encoding a fusion protein and an isolated nucleic acid encoding an amino acid metabolism gene. Further, an isolated nucleic acid encoding a fusion protein and an amino acid metabolism gene will have a promoter suitable for driving expression of such an isolated nucleic acid. Promoters useful for driving expression in a bacterial system are well known in the art, and include bacteriophage lambda, the bla promoter of the beta-lactamase gene of pBR322, and the CAT promoter of the chloramphenicol acetyl transferase gene of pBR325. Further examples of prokaryotic promoters include the major right and left promoters of 5 bacteriophage lambda (PL and PR), the trp, recA, lacZ, lad, and gal promoters of E. coli, the alpha-amylase (Ulmanen et al, 1985. J. Bacterid. 162:176-182) and the S28-specific promoters of B. subtilis (Gilman et al, 1984 Gene 32: 11- 20), the promoters of the bacteriophages of Bacillus (Gryczan, 1982, In: The Molecular Biology of the Bacilli, Academic Press, Inc., New York), and Streptomyces promoters (Ward et al, 1986, Mol. Gen. Genet. 203:468- 478). Additional prokaryotic promoters contemplated in the present invention are reviewed in, for example, Glick (1987, J. Ind. Microbiol. 1:277-282); Cenatiempo, (1986, Biochimie, 68:505-516); and Gottesman, (1984, Ann. Rev. Genet. 18:415-442). Further examples of promoter/regulatory elements contemplated in the present invention include, but are not limited to the Listerial prfA promoter, the Listerial hly promoter, the Listerial p60 promoter and the Listerial actA promoter (GenBank Acc. No. NC_003210) or fragments thereof.
[00204] In another embodiment, a plasmid of methods and compositions disclosed herein comprises a gene encoding a fusion protein. In another embodiment, subsequences are cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments are then, in another embodiment, ligated to produce the desired DNA sequence. In another embodiment, DNA encoding the antigen is produced using DNA amplification methods, for example polymerase chain reaction (PCR), as discussed below
[00205] In another embodiment, DNA encoding the fusion protein or the recombinant protein disclosed herein is cloned using DNA amplification methods such as polymerase chain reaction (PCR). Thus, for example, the gene for non-hemolytic LLO is PCR amplified, using a sense primer comprising a suitable restriction site and an antisense primer comprising another restriction site, e.g. a non-identical restriction site to facilitate cloning. The same is repeated for the isolated nucleic acid encoding an antigen. Ligation of the non-hemolytic LLO and antigen sequences and insertion into a plasmid or vector produces a vector encoding non-hemolytic LLO joined to a terminus of the antigen. The two molecules are joined either directly or by a short spacer introduced by the restriction site.
[00206] Fusion proteins comprising the survivin antigen or immunogenic fragment thereof may be prepared by any suitable method, including, for example, cloning and restriction of appropriate sequences or direct chemical synthesis. Alternatively, subsequences may be cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments may then be ligated to produce the desired DNA sequence. In one embodiment, DNA encoding the antigen can be produced using DNA amplification methods, for example polymerase chain reaction (PCR). First, the segments of the native DNA on either side of the new terminus are amplified separately. The 5' end of the one amplified sequence encodes the peptide linker, while the 3' end of the other amplified sequence also encodes the peptide linker. Since the 5' end of the first fragment is complementary to the 3' end of the second fragment, the two fragments (after partial purification, e.g. on LMP agarose) can be used as an overlapping template in a third PCR reaction. The amplified sequence will contain codons, the segment on the carboxy side of the opening site (now forming the amino sequence), the linker, and the sequence on the amino side of the opening site (now forming the carboxyl sequence). The antigen is ligated into a plasmid.
[00207] In another embodiment, a nucleic acid sequences encoding the fusion or recombinant proteins are transformed into a variety of host cells, including E. coli, other bacterial hosts, such as Listeria, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cells lines and myeloma cell lines. The recombinant fusion protein gene will be operably linked to appropriate expression control sequences for each host. Promoter/ regulatory sequences are described in detail elsewhere herein. In another embodiment, the plasmid further comprises additional promoter regulatory elements, as well as a ribosome binding site and a transcription termination signal. For eukaryotic cells, the control sequences will include a promoter and an enhancer derived from e.g. immunoglobulin genes, SV40, cytomegalovirus, etc., and a polyadenylation sequence. In another embodiment, the sequences include splice donor and acceptor sequences.
[00208] "Stably maintained" refers to maintenance of a nucleic acid molecule or plasmid in the absence of selection (e.g. antibiotic selection) for 10 generations, without detectable loss. In another embodiment, the period is 15 generations. In another embodiment, the period is 20 generations. In another embodiment, the period is 25 generations. In another embodiment, the period is 30 generations. In another embodiment, the period is 40 generations. In another embodiment, the period is 50 generations. In another embodiment, the period is 60 generations. In another embodiment, the period is 80 generations. In another embodiment, the period is 100 generations. In another embodiment, the period is 150 generations. In another embodiment, the period is 200 generations. In another embodiment, the period is 300 generations. In another embodiment, the period is 500 generations. In another embodiment, the period is more than 500 generations. In another embodiment, the nucleic acid molecule or plasmid is maintained stably in vitro (e.g. in culture). In another embodiment, the nucleic acid molecule or plasmid is maintained stably in vivo. In another embodiment, the nucleic acid molecule or plasmid is maintained stably both in vitro and in vitro. [00209] The recombinant Listeria strain of methods and compositions disclosed herein is, in another embodiment, a recombinant Listeria monocytogenes strain. In another embodiment, the Listeria strain is a recombinant Listeria seeligeri strain. In another embodiment, the Listeria strain is a recombinant Listeria grayi strain. In another embodiment, the Listeria strain is a recombinant Listeria ivanovii strain. In another embodiment, the Listeria strain is a recombinant Listeria murrayi strain. In another embodiment, the Listeria strain is a recombinant Listeria welshimeri strain. In another embodiment, the Listeria strain is a recombinant strain of another Listeria species.
[00210] In one embodiment, attenuated Listeria strains, such as Lm ddta-actA mutant (Brundage et al, 1993, Proc. Natl. Acad. Sci., USA, 90: 11890-11894), L. monocytogenes delta-plcA (Camilli et al, 1991, J. Exp. Med., 173:751-754), or delta-actA, ddta-inlB (Brockstedt et 5 al, 2004, PNAS, 101:13832-13837) are used in the present invention. In another embodiment, attenuated Listeria strains are constructed by introducing one or more attenuating mutations, as will be understood by one of average skill in the art when equipped with the disclosure herein. Examples of such strains include, but are not limited to Listeria strains auxotrophic for aromatic amino acids (Alexander et al, 1993, Infection and Immunity 10 61:2245-2248) and mutant for the formation of lipoteichoic acids (Abachin et al, 2002, Mol. Microbiol. 43:1-14) and those attenuated by a lack of a virulence gene (see examples herein).
[00211] In another embodiment, a recombinant Listeria strain disclosed herein has been passaged through an animal host. In another embodiment, the passaging maximizes efficacy of the strain as a vaccine vector. In another embodiment, the passaging stabilizes the immunogenicity of the Listeria strain. In another embodiment, the passaging stabilizes the virulence of the Listeria strain. In another embodiment, the passaging increases the immunogenicity of the Listeria strain. In another embodiment, the passaging increases the virulence of the Listeria strain. In another embodiment, the passaging removes unstable sub-strains of the Listeria strain. In another embodiment, the passaging reduces the prevalence of unstable sub-strains of the Listeria strain. In another embodiment, the Listeria strain contains a genomic insertion of the gene encoding the antigen-containing recombinant peptide. In another embodiment, the Listeria strain carries a plasmid comprising the gene encoding the antigen-containing recombinant peptide. In another embodiment, the passaging is performed as described herein. In another embodiment, the passaging is performed by other methods known in the art.
[00212] In another embodiment, a recombinant nucleic acid disclosed herein is operably linked to a promoter/regulatory sequence that drives expression of the encoded peptide in the Listeria strain. Promoter/regulatory sequences useful for driving constitutive expression of a gene are well known in the art and include, but are not limited to, for example, the PhiyA, VactA, and p60 promoters of Listeria, the Streptococcus bac promoter, the Streptomyces griseus sgiA promoter, and the B. thuringiensis phaZ promoter.
[00213] In another embodiment, inducible and tissue specific expression of the nucleic acid encoding a peptide disclosed herein is accomplished by placing the nucleic acid encoding the peptide under the control of an inducible or tissue specific promoter/regulatory sequence. Examples of tissue specific or inducible promoter/regulatory sequences which are useful for his purpose include, but are not limited to the MMTV LTR inducible promoter, and the SV40 late enhancer/promoter. In another embodiment, a promoter that is induced in response to inducing agents such as metals, glucocorticoids, and the like, is utilized. Thus, it will be appreciated that the invention includes the use of any promoter/regulatory sequence, which is either known or unknown, and which is capable of driving expression of the desired protein operably linked thereto.
[00214] It will be appreciated by a skilled artisan that the term "heterologous" encompasses a nucleic acid, amino acid, peptide, polypeptide, or protein derived from a different species than the reference species. Thus, for example, a Listeria strain expressing a heterologous polypeptide, in one embodiment, would express a polypeptide that is not native or endogenous to the Listeria strain, or in another embodiment, a polypeptide that is not normally expressed by the Listeria strain, or in another embodiment, a polypeptide from a source other than the Listeria strain. In another embodiment, heterologous may be used to describe something derived from a different organism within the same species. In another embodiment, the heterologous antigen is expressed by a recombinant strain of Listeria, and is processed and presented to cytotoxic T-cells upon infection of mammalian cells by the recombinant strain. In another embodiment, the heterologous antigen expressed by Listeria species need not precisely match the corresponding unmodified antigen or protein in the tumor cell or infectious agent so long as it results in a T-cell response that recognizes the unmodified antigen or protein which is naturally expressed in the mammal. The term heterologous antigen may be referred to herein as "antigenic polypeptide", "heterologous protein", "heterologous protein antigen", "protein antigen", "antigen", and the like. In one embodiment, a heterologous antigen disclosed herein is a survivin polypeptide or antigenic portion thereof.
[00215] In one embodiment, the two molecules of the fusion protein (for example the LLO, ActA fragment or PEST sequence and the antigen, for example survivin or a portion thereof) 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. In another embodiment, the two molecules of the protein (for example, the LLO fragment and the antigen) are synthesized separately or unfused. In another embodiment, the two molecules of the protein are synthesized separately from the same nucleic acid. In yet another embodiment, the two molecules are individually synthesized from separate nucleic acids.
II. COMPOSITIONS
[00216] In one embodiment, the present invention provides a composition comprising an antagonist of immunosuppressive signaling molecules. In one embodiment, immunosuppressive signaling molecules mediate resistance of tumors to a host' s immune response. In another embodiment, an immunosuppressive signaling molecule comprises a CD28/CTLA-4 receptor superfamily. In another embodiment, an immunosuppressive signaling molecule comprises a B7 receptor family.
[00217] In another embodiment, an immunosuppressive molecule comprises PD- 1. In another embodiment, an immunosuppressive molecule comprises PD-L1. In another embodiment, an immunosuppressive molecule comprises PD-L2. In another embodiment, an immunosuppressive molecule comprises B7- 1.
[00218] In yet another embodiment, an antagonist disclosed herein is any chemical compound or biological molecule that blocks interaction of an immunosuppressive molecules with these molecules' receptors. In one embodiment, an antagonist is a small molecule. In another embodiment, the antagonist is an antibody. In another embodiment, the antagonist is a monoclonal antibody (mAb). In another embodiment, the antagonist is a polyclonal antibody. In yet another embodiment, the antagonist is an antigen-binding antibody fragment. Hence, in one embodiment, a composition disclosed herein comprises an antagonist antibody. [00219] It will be appreciated by a skilled artisan that the term "antibody" may encompass any form of immunoglobulin molecule that exhibits the desired biological or binding activity. Thus, it is used in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g. , bispecific antibodies), humanized, human antibodies, chimeric antibodies and camelized single domain antibodies. "Parental antibodies" are antibodies obtained by exposure of an immune system to an antigen prior to modification of the antibodies for an intended use, such as humanization of an antibody for use as a human therapeutic. In one embodiment, the term "antibody" encompasses not only intact polyclonal or monoclonal antibodies, but also, unless otherwise specified, any antigen binding portion thereof that competes with the intact antibody for specific binding, fusion proteins comprising an antigen binding portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site. Antigen binding portions include, for example, Fab, Fab', F(ab')2, Fd, Fv, domain antibodies (dAbs, e.g., shark and camelid antibodies), fragments including complementarity determining regions (CDRs), single chain variable fragment antibodies (scFv), maxibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide. An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant region of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG2, IgG3, IgG4, IgAi and IgA2. The heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. [00220] It will be appreciated by a skilled artisan that the term "antibody fragment" or
"antigen binding fragment" may encompass to antigen binding fragments of antibodies, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full- length antibody, e.g. fragments that retain one or more CDR regions. Examples of antibody binding fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and multispecific antibodies formed from antibody fragments.
[00221] It will be appreciated by a skilled artisan that the term "CDR" or "CDRs" may encompass complementarity determining region(s) in an immunoglobulin variable region, defined using the Kabat numbering system, unless otherwise indicated. [00222] "Kabat" as used herein means an immunoglobulin alignment and numbering system pioneered by Elvin A. Kabat ((1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.). [00223] It will be appreciated by a skilled artisan that the term "Monoclonal antibody" or
"mAb" or "Mab", may encompass a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, a monoclonal antibody to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The monoclonal antibody may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731.
[00224] In another embodiment, a mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region. In some embodiments, the human constant region is selected from the group consisting of IgGl, IgG2, IgG3 and IgG4 constant regions, and in some embodiments, the human constant region is an IgGl or IgG4 constant region. In some embodiments, the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab')2, scFv and Fv fragments.
[00225] It will be appreciated by a skilled artisan that the term "Chimeric antibody" may encompass an antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
[00226] It will be appreciated by a skilled artisan that the term "Human antibody" may encompass an antibody that comprises human immunoglobulin protein sequences only. A human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell. Similarly, "mouse antibody" or "rat antibody" refer to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
[00227] It will be appreciated by a skilled artisan that the term "Humanized antibody" may encompass forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. The prefix "hum", "hu" or "h" is added to antibody clone designations when necessary to distinguish humanized antibodies from parental rodent antibodies. The humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parental rodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability of the humanized antibody, or for other reasons. [00228] In another embodiment, an immunosuppressive antagonist molecule disclosed herein comprises PD-1 antagonists. In another embodiment, PD-1 antagonists interact with PD-1 and block binding of human PD-L1 expressed on a cancer cell to human PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also block binding of human PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1. Alternative names or synonyms for PD-1 include: PDCD1, PD1, CD279 and SLEB2. Exemplary human PD-1 amino acid sequences can be found in NCBI Locus No.: NP_005009.
[00229] Examples of mAbs that bind to human PD-1, and useful in the treatment method, medicaments and uses of the present invention, are described in US7488802, US7521051, US8008449, US8354509, US8168757, WO2004/004771, WO2004/072286, WO2004/056875, and US2011/0271358. Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses disclosed herein include:, nivolumab (BMS-936558), a human IgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 1, pages 68-69 (2013) and which comprises the heavy and light chain amino acid sequences shown in Figure 7; the humanized antibodies h409Al l, h409A16 and h409A17, which are described in WO2008/156712, and AMP-514, which is being developed by Medlmmune.
[00230] In another embodiment, the immunosuppressive antagonists molecules disclosed herein comprise PD-L1 antagonists. In another embodiment, PD-L1 antagonists interact with PD-L1 and block binding of human PD-1 expressed on an immune cell (T cell, B cell or NKT cell) to human PD-L1 expressed on a cancer cell. Alternative names or synonyms for PD-L1 include: PDCD1L1, PDL1, B7H1, B7-4, CD274 and B7-H. Exemplary human PD-L1 amino acid sequence can be found in NCBI Locus No.: NP_054862.
[00231] Examples of mAbs that bind to human PD-L1, and useful in the treatment method, medicaments and uses of the present invention, are described in WO2013/019906, W02010/077634 Al and US8383796.
[00232] It will be appreciated by a skilled artisan that the term "Variable regions" or "V region" may encompass the segment of IgG chains which is variable in sequence between different antibodies. It extends to Kabat residue 109 in the light chain and 113 in the heavy chain. A "variable region" of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. Typically, the variable regions of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope. In general, from N-terminal to C-terminal, both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al; National Institutes of Health, Bethesda, Md. ; 5th ed.; NIH Publ. No. 91-3242 (1991); Kabat (1978) Adv. Prot. Chem. 32: 1-75; Kabat, et al, (1977) J. Biol. Chem. 252:6609-6616; Chothia, et al, (1987) J Mol. Biol. 196:901- 917 or Chothia, et al, (1989) Nature 342:878-883.
[00233] It will be appreciated by a skilled artisan that the term "hypervariable region" may encompass the amino acid residues of an antibody that are responsible for antigen-binding. The hypervariable region comprises amino acid residues from a "complementarity determining region" or "CDR" {i.e. CDRL1, CDRL2 and CDRL3 in the light chain variable domain and CDRH1, CDRH2 and CDRH3 in the heavy chain variable domain). See Kabat et al. (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (defining the CDR regions of an antibody by sequence); see also Chothia and Lesk (1987) J. Mol. Biol. 196: 901-917 (defining the CDR regions of an antibody by structure). As used herein, the term "framework" or "FR" residues refers to variable domain residues other than the hypervariable region residues defined herein as CDR residues.
[00234] It will be appreciated by a skilled artisan that the term "Framework region" or
"FR" may encompass the immunoglobulin variable regions excluding the CDR regions.
[00235] In another embodiment, antagonists comprise PD-L2 antagonists. In another embodiment, PD-L2 antagonists interact with PD-L2 and block binding of human PD-1 expressed on an immune cell (T cell, B cell or NKT cell) to human PD-L2 expressed on a cancer cell. Alternative names or synonyms for PD-L2 include: PDCD1L2, PDL2, B7-DC, Btdc and CD273. Exemplary human PD-L2 amino acid sequences can be found in NCBI Locus No.: NP_079515. [00236] In another embodiment, antagonists comprise B7-1 antagonists.
[00237] Other PD-1 antagonists useful in any of the treatment methods, medicaments and uses disclosed herein include an immunoadhesin that specifically binds to human PD-1 or human PD-Ll, e.g. a fusion protein containing the extracellular or PD-1 binding portion of PD-Ll or PD-L2 fused to a constant region such as an Fc region of an immunoglobulin molecule. Examples of immunoadhesion molecules that specifically bind to PD-1 are described in WO2010/027827 and WO2011/066342. Specific fusion proteins useful as the PD-1 antagonist in the treatment method, medicaments and uses disclosed herein include AMP-224 (also known as B7-DCIg), which is a PD-L2-FC fusion protein and binds to human PD-1. In another embodiment, other PD-1/PD-L1 binding antagonists include a small molecule or a biomolecule that blocks or competes with critical binding or signaling of PD-1 and its ligand or signaling to the effector cell after this binding.
[00238] It will be appreciated by a skilled artisan an antibody that "specifically binds to" a specified target protein may encompass an antibody that exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity. An antibody is considered "specific" for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g. without producing undesired results such as false positives. Antibodies or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins. In one embodiment, an antibody is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human PD-1, human PD-L1, human PD-L2, or human B7-1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
[00239] In some embodiments of the treatment method, medicaments and uses of the present invention, the PD- 1 antagonist is a monoclonal antibody, or an antigen binding fragment thereof.
[00240] In other embodiments of the treatment method, medicaments and uses of the present invention, the PD-1 antagonist is a monoclonal antibody, or antigen binding fragment thereof, which specifically binds to human PD- 1 and comprises (a) a heavy chain variable region, and (b) a light chain variable.
[00241] In another embodiment, of the treatment method, medicaments and uses of the present invention, the PD-1 antagonist is a monoclonal antibody which specifically binds to human PD-1.
[00242] In yet another embodiment, of the treatment method, medicaments and uses disclosed herein, the PD-1 antagonist is a monoclonal antibody which specifically binds to human PD-1.
[00243] "PD-L1" expression or "PD-L2" expression as used herein means any detectable level of expression of the designated PD-L protein on the cell surface or of the designated PD-L mRNA within a cell or tissue. PD-L protein expression may be detected with a diagnostic PD-L antibody in an IHC assay of a tumor tissue section or by flow cytometry. Alternatively, PD-L protein expression by tumor cells may be detected by PET imaging, using a binding agent (e.g., antibody fragment, antibody and the like) that specifically binds to the desired PD-L target, e.g., PD-L1 or PD-L2. Techniques for detecting and measuring PD-L mRNA expression include RT- PCR and realtime quantitative RT-PCR. [00244] Several approaches have been described for quantifying PD-L1 protein expression in IHC assays of tumor tissue sections. See, e.g., Thompson, R. H., et al., PNAS 101 (49); 17174- 17179 (2004); Thompson, R. H. et al., Cancer Res. 66:3381-3385 (2006); Gadiot, J., et al., Cancer 117:2192-2201 (2011); Taube, J. M. et al., Sci Transl Med 4, 127ra37 (2012); and Toplian, S. L. et al., New Eng. J Med. 366 (26): 2443-2454 (2012). [00245] One approach employs a simple binary end-point of positive or negative for PD- LI expression, with a positive result defined in terms of the percentage of tumor cells that exhibit histologic evidence of cell-surface membrane staining. A tumor tissue section is counted as positive for PD-Ll expression if IHC staining is observed in at least 1%, and preferably 5% of total tumor cells. In an embodiment, a pancreatic tumor sample is designated as having weak PD-Ll expression if 1% to 49% of the total tumor cells in the sample exhibit membrane staining and is designated as having strong PD-Ll expression if at least 50% of the tumor cells in the sample exhibit membrane staining, in each case as determined by IHC assay using the antibody 22C3 described in WO2014/100079.
[00246] In another approach, PD-Ll expression in the tumor tissue section is quantified in the tumor cells as well as in infiltrating immune cells, which predominantly comprise lymphocytes. The percentage of tumor cells and infiltrating immune cells that exhibit membrane staining are separately quantified as < 5%, 5 to 9%, and then in 10% increments up to 100%. For tumor cells, PD-Ll expression is counted as negative if the score is < 5% score and positive if the score is > 5%. PD-Ll expression in the immune infiltrate is reported as a semi-quantitative measurement called the adjusted inflammation score (AIS), which is determined by multiplying the percent of membrane staining cells by the intensity of the infiltrate, which is graded as none (0), mild (score of 1, rare lymphocytes), moderate (score of 2, focal infiltration of tumor by lymphohistiocytic aggregates), or severe (score of 3, diffuse infiltration). A tumor tissue section is counted as positive for PD-Ll expression by immune infiltrates if the AIS is > 5. [00247] The level of PD-L mRNA expression may be compared to the mRNA expression levels of one or more reference genes that are frequently used in quantitative RT-PCR, such as ubiquitin C.
[00248] In some embodiments, a level of PD-Ll expression (protein and/or mRNA) by malignant cells and/or by infiltrating immune cells within a tumor is determined to be "overexpressed" or "elevated" based on comparison with the level of PD-Ll expression (protein and/ or mRNA) by an appropriate control. For example, a control PD-Ll protein or mRNA expression level may be the level quantified in nonmalignant cells of the same type or in a section from a matched normal tissue. In some embodiments, PD-Ll expression in a tumor sample is determined to be elevated if PD-Ll protein (and/or PD-Ll mRNA) in the sample is at least 10%, 20%, or 30% greater than in the control.
[00249] In one embodiment, disclosed herein is a composition comprising a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to an additional polypeptide, and further comprising a PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and wherein administering the composition to a subject having a survivin-expressing cancer treats, ameliorates, or induces regression of the cancer. In another embodiment, disclosed herein is a pharmaceutical composition comprising a nucleic acid encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to an additional polypeptide and further comprising a PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist. In another embodiment, disclosed herein is a pharmaceutical composition comprising a recombinant Listeria vaccine strain comprising a nucleic acid encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to an additional polypeptide, the composition further comprising a PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist. In another embodiment, the additional polypeptide is a PEST- containing peptide or polypeptide. In another embodiment, the additional polypeptide is an N- terminal Listeriolysin O (LLO) fragment thereof. In another embodiment, the additional polypeptide is an N-terminal ActA fragment thereof. In another embodiment, the additional polypeptide is a PEST-containing amino acid sequence.
[00250] It will be appreciated by a skilled artisan that the term "Administration" and
"treatment," as it applies to an animal, human, experimental subject, cell, tissue, organ, or biological fluid, may encompass contacting of an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human, subject, cell, tissue, organ, or biological fluid. Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell. "Administration" and "treatment" also may encompass in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell. The term "subject" includes any organism, preferably an animal, more preferably a mammal {e.g., rat, mouse, dog, cat, rabbit) and most preferably a human.
[00251] It will be appreciated by a skilled artisan that the term "pharmaceutical composition" may encompass a therapeutically effective amount of the active ingredient or ingredients comprising the Listeria strain, an immunosuppressive molecule antagonist, or both together with a pharmaceutically acceptable carrier or diluent.
[00252] It will be appreciated by a skilled artisan that the terms "therapeutically effective amount", in reference to the treatment of tumor, may encompass an amount capable of invoking one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, slowing down and complete growth arrest; (2) reduction in the number of tumor cells; (3) reduction in tumor size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of tumor cell infiltration into peripheral organs; (5) inhibition (i.e., reduction, slowing down or complete stopping) of metastasis; (6) enhancement of anti-tumor immune response, which may, but does not have to, result in the regression or rejection of the tumor; and/or (7) relief, to some extent, of one or more symptoms associated with the disorder. A "therapeutically effective amount" of a vaccine disclosed herein for purposes of treatment of tumor may be determined empirically and in a routine manner.
[00253] It will be appreciated by a skilled artisan that the terms "immunogenic composition", "composition" and "pharmaceutical composition" may be used interchangeably. It is also to be understood that administration of such compositions enhances an immune response, or increase a T effector cell to regulatory T cell ratio or elicit an anti-tumor immune response, as further provided herein.
[00254] In another embodiment, a nucleic acid molecule of the anti-tumor composition disclosed herein further comprises a second open reading frame encoding a metabolic enzyme, wherein the metabolic enzyme complements a mutation in the chromosome of the recombinant Listeria strain.
[00255] In another embodiment, an immune response elicited by the methods and compositions disclosed herein comprises an immune response to at least one 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 at least one subdominant epitope. In another embodiment, the only measurable component of the immune response is an immune response to at least one subdominant epitope. In yet another embodiment, the immune response against the survivin-expressing cancer comprises an immune response to at least one dominant epitope of the survivin protein.
[00256] In one embodiment, a composition disclosed hereincomprises a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprises recombinant Listeria strain comprising a nucleic acid molecule, wherein the nucleic acid molecule comprises a first open reading frame encoding a polypeptide, wherein the polypeptide comprises a PEST-containing peptide or polypeptide fused to a heterologous antigen, wherein the PEST-containing peptide or polypeptide comprises an N-terminal fragment of Listeriolysin O (LLO), an N-terminal fragment of ActA, or a PEST-containing amino acid sequence, and wherein the heterologous antigen comprises a survivin antigen or an immunogenic fragment thereof. [00257] In another embodiment, administration of compositions disclosed herein increase the number of antigen- specific T cells. In another embodiment, administration of compositions activates co- stimulatory receptors on T cells. In another embodiment, administration of compositions induces proliferation of memory and/or effector T cells. In another embodiment, administration of compositions increases proliferation of T cells. In another embodiment, administration of compositions disclosed herein negate tumor immunosuppressive signaling. In another embodiment, administration of compositions suppresses PD-1 mediated immunosuppressive signaling. In another embodiment, administration of compositions counters tumor mediated attenuation of T-cell activation. In another embodiment, administration of compositions neutralizes tumor's immune surveillance evasion mechanisms.
[00258] In another embodiment, administration of compositions disclosed herein increase the number of active antigen- specific T cells. In another embodiment, administration of compositions enhances activation of co- stimulatory receptors on T cells. In another embodiment, administration of compositions induces proliferation of memory and/or effector T cells. In another embodiment, administration of compositions increases proliferation of T cells.
[00259] Compositions disclosed herein may be used in methods disclosed hereinin order to elicit an enhanced anti-tumor T cell response in a subject, in order to inhibit tumor-mediated immunosuppression in a subject, or for increasing the ratio or T effector cells to regulatory T cells (Tregs) in the spleen and tumor of a subject, or any combination thereof.
[00260] In another embodiment, a composition comprising a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and a recombinant attenuated a Listeria strain described hereinabove, further comprises an adjuvant. The adjuvant utilized in methods and compositions disclosed herein is, in another embodiment, a granulocyte/macrophage colony- stimulating factor (GM-CSF) protein. In another embodiment, the adjuvant comprises a GM-CSF protein. In another embodiment, the adjuvant is a nucleotide molecule encoding GM-CSF. In another embodiment, the adjuvant comprises a nucleotide molecule encoding GM-CSF. In another embodiment, the adjuvant is saponin QS21. In another embodiment, the adjuvant comprises saponin QS21. In another embodiment, the adjuvant is monophosphoryl lipid A. In another embodiment, the adjuvant comprises monophosphoryl lipid A. In another embodiment, the adjuvant is SBAS2. In another embodiment, the adjuvant comprises SBAS2. In another embodiment, the adjuvant is an unmethylated CpG-containing oligonucleotide. In another embodiment, the adjuvant comprises an unmethylated CpG-containing oligonucleotide. In another embodiment, the adjuvant is an immune- stimulating cytokine. In another embodiment, the adjuvant comprises an immune- stimulating cytokine. In another embodiment, the adjuvant is a nucleotide molecule encoding an immune- stimulating cytokine. In another embodiment, the adjuvant comprises a nucleotide molecule encoding an immune- stimulating cytokine. In another embodiment, the adjuvant is or comprises a quill glycoside. In another embodiment, the adjuvant is or comprises a bacterial mitogen. In another embodiment, the adjuvant is or comprises a bacterial toxin. Other adjuvants are known in the art.
[00261] It will be appreciated by a skilled artisan that the term "oligonucleotide" is interchangeable with the term "nucleic acid", and may encompass a molecule, which may include, but is not limited to, prokaryotic sequences, eukaryotic mRNA, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic (e.g., mammalian) DNA, and even synthetic DNA sequences. The term may encompass sequences that include any of the known base analogs of DNA and RNA.
[00262] In one embodiment, the present invention provides a composition comprising a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria strain expressing the antigen. In another embodiment, the antigen is survivin or an antigenic portion thereof. In another embodiment, the PD-1 antagonist is an anti-PD-1 antibody or an antigen binding fragment thereof. In another embodiment, PD-L1 antagonist is an anti-PD-Ll antibody or an antigen binding fragment thereof. In another embodiment, PD-L2 antagonist is an anti-PD-L2 antibody or an antigen binding fragment thereof. In another embodiment, B7-1 antagonist is an anti-B7-l antibody or an antigen binding fragment thereof. In another embodiment, the present invention provides immunogenic compositions comprising PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising recombinant Listeria provided herein. In another embodiment, the present invention provides pharmaceutical compositions comprising a PD- 1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising recombinant Listeria provided herein.
[00263] In another embodiment, of methods and compositions of the present invention, the fusion protein comprises the survivin antigen or immunogenic fragment thereof and an additional polypeptide. In another embodiment, the additional polypeptide disclosed herein is an N-terminal Listeriolysin O (LLO) fragment. In another embodiment, the additional polypeptide is an N-terminal ActA fragment. In another embodiment, the additional polypeptide is a PEST-containing amino acid sequence. In some embodiments, an additional polypeptide or an additional adjuvant polypeptide augments antigen presentation and immunity in a similar fashion to LLO.
[00264] In another embodiment, PD-L1 antagonist is an anti-PD-Ll antibody or an antigen binding fragment thereof. In another embodiment, PD-L2 antagonist is an anti-PD-L2 antibody or an antigen binding fragment thereof. In another embodiment, B7-1 antagonist is an anti-B7-l antibody or an antigen binding fragment thereof. In another embodiment, the PD-1 antagonist blocks interaction of PD-1 with any of its ligands. In another embodiment, the PD-1 antagonist blocks the interaction of PD-1 with PD-Ll, PD-L2, or both. In another embodiment, the PD-Ll antagonist blocks the interaction of PD-Ll with PD-1, B7-1, or both. In another embodiment, the PD-L2 antagonist blocks the interaction of PD-L2 with PD-1. In another embodiment, the B7-1 antagonist blocks the interaction of B7-1 with PD-Ll.
[00265] In one embodiment, disclosed herein is a composition comprising PD-1 antagonist, PD- Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria of the present invention. In another embodiment, disclosed herein is a composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-lantagonist comprising a recombinant attenuated Listeria expressing a fusion polypeptide of the present invention. In another embodiment, the composition further comprises formulation buffer suitable for administration to a subject.
[00266] In one embodiment, disclosed herein is a pharmaceutical composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria of the present invention. In another embodiment, disclosed herein is a composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist comprising a recombinant attenuated Listeria expressing a fusion polypeptide of the present invention. In another embodiment, the pharmaceutical composition further comprises formulation buffer suitable for administration to a subject.
[00267] In one embodiment, disclosed herein is an immunogenic composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprising a recombinant Listeria of the present invention. In another embodiment, disclosed herein is a composition comprising PD-1 antagonist, PD-Ll antagonist, a PD-L2 antagonist or B7-1 antagonist comprising a recombinant attenuated Listeria expressing a fusion polypeptide of the present invention. In another embodiment, said immunogenic composition further comprises formulation buffer suitable for administration to a subject.
[00268] The combination therapy may also comprise one or more additional therapeutic agents. The additional therapeutic agent may be, e.g., a chemo therapeutic, a biotherapeutic agent (including but not limited to antibodies to VEGF, VEGFR, EGFR, Her2/neu, other growth factor receptors, CD20, CD40, CD-40L, CTLA-4, OX-40, GITR, 4- IBB, and ICOS), an immunogenic agent (for example, attenuated cancerous cells, tumor antigens, antigen presenting cells such as dendritic cells pulsed with tumor derived antigen or nucleic acids, immune stimulating cytokines (for example, IL-2, IFNa2, GM-CSF), and cells transfected with genes encoding immune stimulating cytokines such as but not limited to GM-CSF). [00269] It will be appreciated by a skilled artisan that the term "Bio therapeutic agent" may encompass a biological molecule, such as an antibody or fusion protein, that blocks ligand / receptor signaling in any biological pathway that supports tumor maintenance and/or growth or suppresses the anti-tumor immune response.
[00270] Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin, especially calicheamicin gammall and calicheamicin phill, see, e.g., Agnew, Chem. Intl. Ed. Engl., 33: 183- 186 (1994); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino- doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6- azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2- ethylhydrazide; procarbazine; razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2, 2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g. paclitaxel and doxetaxel; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti- estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen, raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate, exemestane, formestane, fadrozole, vorozole, letrozole, and anastrozole; and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. [00271] It will be appreciated by a skilled artisan that the term "Chemotherapeutic agent" may encompass a chemical or biological substance that can cause death of cancer cells, or interfere with growth, division, repair, and/or function of cancer cells. Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytoxic/antitumor antibiotics, topisomerase inhibitors, photosensitizers, anti-estrogens and selective estrogen receptor modulators (SERMs), anti- progesterones,estrogen receptor down-regulators (ERDs), estrogen receptor antagonists, leutinizing hormone-releasing hormone agonists, anti-androgens, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, anti-sense oligonucleotides that inhibit expression of genes implicated in abnormal cell proliferation or tumor growth. Chemotherapeutic agents useful in the treatment methods disclosed herein include cytostatic and/or cytotoxic agents.
[00272] Each therapeutic agent in a combination therapy of the invention may be administered either alone or in a medicament (also referred to herein as a pharmaceutical composition) which comprises the therapeutic agent and one or more pharmaceutically acceptable carriers, excipients and diluents, according to standard pharmaceutical practice.
[00273] it w[\\ be appreciated by a skilled artisan that the term "pharmaceutically acceptable carrier" may encompass any inactive substance that is suitable for use in a formulation for the administration to a human of a PD-1 antagonist and/or a live-attenuated Listeria strain that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells or a live-attenuated Listeria monocytogenes strain bioengineered, by transforming it with an expression vector to express a survivin antigen or immunogenic fragment thereof fused to a tLLO or ADXS31-265 {10403S da ] dat('] actA('] pADV265) strain. [00274] It will be appreciated by a skilled artisan that the term "Transforming," may encompass engineering a bacterial cell to take up a plasmid or other heterologous DNA molecule. In another embodiment, "transforming" refers to engineering a bacterial cell to express a gene of a plasmid or other heterologous DNA molecule.
III. THERAPEUTIC METHODS OF USE
[00275] In one embodiment, disclosed herein a method of treating a tumor or cancer in a subject, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration treats a tumor or a cancer in said subject. In another embodiment, the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer.
[00276] In another embodiment, disclosed herein is an immunogenic composition as described above, for treating cancer. For example, an immunogenic composition used in a method disclosed herein comprises a PD-1 antagonist, PD-L1 antagonist, a PD-L2 antagonist or B7-1 antagonist, and further comprises a Listeria strain expressing a fusion polypeptide as described throughout, wherein the fusion polypeptide comprises a survivin antigen.
[00277] It will be appreciated by a skilled artisan that in one embodiment, the term "treating" may encompass curing a disease. In another embodiment, "treating" may encompass preventing a disease. In another embodiment, "treating" may encompass reducing the incidence of a disease. In another embodiment, "treating" may encompass ameliorating symptoms of a disease. In another embodiment, "treating" may encompass increasing performance free survival or overall survival of a patient. In another embodiment, "treating" may encompass stabilizing the progression of a disease. In another embodiment, "treating" may encompass inducing remission. In another embodiment, "treating" may encompass slowing the progression of a disease. The terms "reducing", "suppressing" and "inhibiting" refer to lessening or decreasing.
[00278] It will be appreciated by a skilled artisan that the term "treating" may encompass both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or lessen the targeted pathologic condition or disorder as described herein. Thus, in one embodiment, treating may include directly affecting or curing, suppressing, inhibiting, preventing, reducing the severity of, delaying the onset of, reducing symptoms associated with the disease, disorder or condition, or a combination thereof. Thus, in one embodiment, "treating" may encompass inter alia delaying progression, expediting remission, inducing remission, augmenting remission, speeding recovery, increasing efficacy of or decreasing resistance to alternative therapeutics, or a combination thereof. In one embodiment, "preventing" or "impeding" may encompass, inter alia, delaying the onset of symptoms, preventing relapse to a disease, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, or a combination thereof. In one embodiment, "suppressing" or "inhibiting", may encompass, inter alia, reducing the severity of symptoms, reducing the severity of an acute episode, reducing the number of symptoms, reducing the incidence of disease-related symptoms, reducing the latency of symptoms, ameliorating symptoms, reducing secondary symptoms, reducing secondary infections, prolonging patient survival, or a combination thereof.
[00279] In one embodiment, symptoms are primary, while in another embodiment, symptoms are secondary. In one embodiment, "primary" refers to a symptom that is a direct result of a particular disease or disorder, while in one embodiment, "secondary" refers to a symptom that is derived from or consequent to a primary cause. In one embodiment, the compounds for use in the present invention treat primary or secondary symptoms or secondary complications. In another embodiment, "symptoms" may be any manifestation of a disease or pathological condition.
[00280] In one embodiment, a tumor or cancer being treated is a survivin-expressing tumor or survivin-expressing cancer. In another embodiment, the cancer being treated is breast cancer, a central nervous system (CNS) cancer, a head and neck cancer, an osteosarcoma (OSA), a canine osteosarcoma (OSA), or Ewing's sarcoma (ES). In another embodiment, the cancer is pancreatic cancer. In another embodiment, the cancer is ovarian cancer. In another embodiment, the cancer is gastric cancer. 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 a CNS 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 mesothelioma. In another embodiment, the cancer is malignant mesothelioma (MM). In another embodiment, the cancer is a melanoma. In another embodiment, the cancer is a glioma. In another embodiment, the cancer is a germ cell tumor. In another embodiment, the cancer is a choriocarcinoma. In another embodiment, the cancer is a lymphoma, leukemia, myeloma or any survivin-expressing cancer known in the art.
[00281] In another embodiment, the cancer is a pancreatic carcinoma. In another embodiment, the cancer is pancreatic ductal carcinoma. In another embodiment, the cancer is acinar cell carcinoma of the pancreas, or cystadenocarcinoma. In another embodiment, the cancer is pancreatic neuroendocrine tumor. In another embodiment, the cancer is insulinoma or gastrinoma. In another embodiment, the cancer is any prostate carcinoma known in the art.
[00282] In another embodiment, the cancer is refractory. In another embodiment, the cancer is advanced. In another embodiment, the cancer is a metastasis. In another embodiment, the cancer has metastasized. In another embodiment, the metastasis of pancreartic cancer, which in one embodiment, comprises metastasis to bone, and in another embodiment, comprises metastasis to other organs. In another embodiment, the cancer is any survivin-expressing pancreatic cancer. In another embodiment, a cancer or solid tumor is a result of relapse or metastatic disease. [00283] In another embodiment, cells of a tumor that is targeted by the methods and compositions disclosed herein express surviving or a fragment thereof. In another embodiment, the tumor is associated with survivin. In another embodiment, survivin expressed by a tumor cell is the target of the immune responses induced by methods and compositions of the present invention. In another embodiment, cells of the tumor that is targeted by methods and compositions disclosed herein express low levels of MHC. In another embodiment, cells of the tumor that is targeted by methods and compositions disclosed herein express high levels of PD-L1. In another embodiment, PD-L1 suppresses or attenuates the T effector cell response to survivin. In another embodiment, the PD-L1 mediated immunosuppression is neutralized by methods and compositions disclosed herein.
[00284] In one embodiment, cancer or tumors may be prevented in specific populations known to be susceptible to a particular cancer or tumor. In one embodiment, such susceptibilty may be due to environmental factors, such as smoking, which in one embodiment, may cause a population to be subject to lung cancer, while in another embodiment, such susceptbility may be due to genetic factors, for example a population with BRCAl/2 mutations may be susceptible, in one embodiment, to breast cancer, and in another embodiment, to ovarian cancer. In another embodiment, one or more mutations on chromosome 8q24, chromosome 17ql2, and chromosome 17q24.3 may increase susceptibility to pancreatic cancer, as is known in the art. Other genetic and environmental factors contributing to cancer susceptibility are known in the art.
[00285] Following the administration of an immunogenic composition provided herein, the methods disclosed herein induce the expansion of T effector cells in peripheral lymphoid organs leading to an enhanced presence of T effector cells at the tumor site. In another embodiment, the methods disclosed herein induce the expansion of T effector cells in peripheral lymphoid organs leading to an enhanced presence of T effector cells at the periphery. Such expansion of T effector cells leads to an increased ratio of T effector cells to regulatory T cells in the periphery and at the tumor site without affecting the number of Tregs. It will be appreciated by the skilled artisan that peripheral lymphoid organs include, but are not limited to, the spleen, peyer's patches, the lymph nodes, the adenoids, etc. In one embodiment, the increased ratio of T effector cells to regulatory T cells occurs in the periphery without affecting the number of Tregs. In another embodiment, the increased ratio of T effector cells to regulatory T cells occurs in the periphery, the lymphoid organs and at the tumor site without affecting the number of Tregs at these sites. In another embodiment, the increased ratio of T effector cells decreases the frequency of Tregs, but not the total number of Tregs at these sites. [00286] In one embodiment, an administration of an immunogenic compositions disclosed herein stimulates the anti-tumor activity of T effector cells. In another embodiment, the administration of an immunogenic compositions disclosed herein negates tumor-mediated immunosuppression of anti-survivin T effector cell response. In yet another embodiment, the administration of an immunogenic composition disclosed herein neutralizes tumor-mediated immunosuppressive signaling. In another embodiment, the administration of an immunogenic compositions disclosed herein desensitizes T effector cells to tumor-mediated immunosuppression.
[00287] In one embodiment, disclosed herein a method for treating a survivin-expressing cancer in a subject comprising administering to the subject a combination therapy which comprises a PD-1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or a B7-1 antagonist and further comprising a live-attenuated Listeria strain that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells. In another embodiment, disclosed herein a method of treating a survivin-expressing cancer in a subject, the method comprising the step of administering to the subject a combination therapy which comprises a PD- 1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or a B7-1 antagonist and further comprising a recombinant Listeria comprising a nucleic acid encoding a fusion polypeptide, wherein the fusion polypeptide comprises a survivin antigen or immunogenic fragment thereof fused to a PEST-containing peptide or polypeptide. In another embodiment, of the invention, disclosed herein a method for treating a survivin-expressing cancer in a subject comprising administering to the subject a combination therapy which comprises a PD-1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or a B7-1 antagonist and further comprises a live-attenuated Listeria monocytogenes strain comprising an expression vector encoding a survivin antigen or immunogenic fragment thereof fused to a tLLO. In yet another embodiment, of the invention, disclosed herein a method for treating a cancer in an individual comprising administering to the individual a combination therapy which comprises a PD-1 antagonist, a PD-L1 antagonist, a PD- L2 antagonist or a B7-1 antagonist and further comprising an ADXS-31-265 (10403S
Figure imgf000071_0001
actA{'} pADV265) strain.
[00288] In one embodiment, disclosed herein are methods of eliciting an enhanced immune response against a survivin-expressing cancer in a subject, the method comprising the step of administering to the subject a combination therapy provided herein.
[00289] In another embodiment, disclosed herein is a method of preventing a tumor or cancer in a subject the method comprising the step of administering to the subject a combination therapy provided herein. In another embodiment, the tumor or cancer is a survivin-expressing tumor or survivin-expressing cancer. In another embodiment, disclosed herein is a method of preventing a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy. In another embodiment, the administration of a survivin- expressing Listeria in the context of a fusion protein with a truncated LLO elicits an immune response against other tumor-associated antigens as a result of epitope spreading.
[00290] In one embodiment, disclosed herein is a method of treating a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy disclosed herein.
[00291] In one embodiment, disclosed herein is a method of delaying the progression of a survivin- expressing cancer in a subject, the method comprising the step of administering to the subject a combination therapy provided herein.
[00292] In one embodiment, disclosed herein is a method of delaying the progression of a survivin- expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination disclosed herein.
[00293] In one embodiment, disclosed herein is a method of prolonging the survival of a subject having a survivin-expressing cancer in a subject, the method comprising the step of administering to the subject a combination disclosed herein.
[00294] In one embodiment, disclosed herein is a method of prolonging the survival of a subject having a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy disclosed herein.
[00295] In one embodiment, disclosed herein is a method of inhibiting, impeding, or delaying metastatic disease in a subject having a survivin-expressing tumor growth in a subject, the method comprising the step of administering to the subject a combination therapy disclosed herein.
[00296] In one embodiment, disclosed herein is a method of inducing an anti-survivin immune response in a subject, the method comprising the step of administering to the subject a combination therapy provided herein. [00297] In one embodiment, disclosed herein is a method of augmenting an anti-survivin immune response in a subject, the method comprising the step of administering to the subject a combination therapy provided herein. [00298] In one embodiment, disclosed herein is a method of preventing an escape mutation in the treatment of survivin over-expressing cancers, the method comprising the step of administering to the subject a combination therapy provided herein.
[00299] In one embodiment, disclosed herein is a method of inducing regression of a survivin antigen-expressing cancer in a subject, the method comprising the step of administering to the subject a combination therapy provided herein.
[00300] In one embodiment, disclosed herein is a method of decreasing the frequency of intra- tumoral T regulatory cells, the method comprising the step of administering to the subject a combination therapy provided herein. [00301] In one embodiment, disclosed herein is a method of treating a metastatic disease coming from a survivin-expressing tumor in a subject, the method comprising the step of administering to the subject a combination therapy provided herein.
[00302] In one embodiment, disclosed herein is a method of breaking tolerance in a subject to a survivin-expressing tumor or cancer in the subject, the method comprising the step of administering to the subject a combination therapy provided herein.
[00303] In one embodiment, disclosed herein is a method of impeding growth of a survivin- expressing cancer in a subject the method comprising the step of administering to the subject a combination therapy provided herein.
[00304] In another embodiment, the methods disclosed herein of inducing an anti-survivin immune response allow treating a survivin-expressing tumor in a subject. In another embodiment, the methods provided herein, of inducing an anti-survivin immune response allow prolonging survival of a subject suffering from a survivin-expressing cancer. In another embodiment, the methods disclosed herein of inducing an anti-survivin immune response induce regression of a tumor or cancer in a subject suffering from a survivin-expressing cancer. In another embodiment, the methods disclosed herein of inducing an anti-survivin immune response break tolerance to survivin in a subject suffering from a survivin-expressing tumor. In another embodiment, the methods disclosed herein of inducing an anti-survivin immune response suppress formation of a survivin-expressing cancer in a subject. In another embodiment, a method of treating further comprises administering said combination therapy following a relapse or metastasis in the subject.
[00305] In one embodiment, any composition comprising an immunosuppressive molecule antagonist and a Listeria strain described herein may be used in the methods of this invention. Compositions comprising an immunosuppressive molecule antagonist Listeria strains have been described in detail above.
[00306] In one embodiment, the present invention provides a method for "epitope spreading" of an anti-tumor response. In another embodiment, the immunization using the compositions and methods disclosed herein induce epitope spreading onto other tumors bearing antigens other than the antigen carried in the vaccine of the present invention.
[00307] 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.
[00308] In one embodiment, disclosed herein is a method of treating, suppressing, or inhibiting a cancer or a tumor growth in a subject by epitope spreading wherein and in another embodiment, the cancer is associated with expression of an antigen or fragment thereof comprised in the composition of the present invention. In another embodiment, the method comprises administering to the subject a combination therapy of the present invention. In yet another embodiment, the subject mounts an immune response against the antigen-expressing cancer or the antigen-expressing tumor, thereby treating, suppressing, or inhibiting a cancer or a tumor growth in a subject.
[00309] In one embodiment, disclosed herein is a method of increasing a ratio of T effector cells to regulatory T cells (Tregs) in the spleen and tumor microenvironments of a subject, comprising administering the immunogenic composition provided herein. In another embodiment, increasing a ratio of T effector cells to regulatory T cells (Tregs) in the spleen and tumor microenvironments in a subject allows for a more profound anti-tumor response in the subject.
[00310] In another embodiment, the T effector cells comprise CD4+FoxP3- T cells. In another embodiment, the T effector cells are CD4+FoxP3- T cells. In another embodiment, the T effector cells comprise CD4+FoxP3- T cells and CD8+ T cells. In another embodiment, the T effector cells are CD4+FoxP3- T cells and CD8+ T cells. In another embodiment, the regulatory T cells is a CD4+FoxP3+ T cell.
[00311] In one embodiment, the present invention provides methods of treating, protecting against, and inducing an immune response against a tumor or a cancer, comprising the step of administering to a subject the combination therapy provided herein.
[00312] In one embodiment, the present invention provides a method of preventing or treating a tumor or cancer in a human subject, comprising the step of administering to the subject the combination therapy strain provided herein, the recombinant Listeria strain comprising a recombinant polypeptide comprising an N-terminal fragment of an LLO protein and tumor- associated antigen, whereby the recombinant Listeria strain induces an immune response against the tumor-associated antigen, thereby treating a tumor or cancer in a human subject. In another embodiment, the immune response is a T-cell response. In another embodiment, the T-cell response is a CD4+FoxP3- T cell response. In another embodiment, the T-cell response is a CD8+ T cell response. In another embodiment, the T-cell response is a CD4+FoxP3- and CD8+ T cell response. In another embodiment, the present invention provides a method of protecting a subject against a tumor or cancer, comprising the step of administering to the subject the combination therapy provided herein. In another embodiment, the present invention provides a method of inducing regression of a tumor in a subject, comprising the step of administering to the subject the combination therapy provided herein. In another embodiment, the present invention provides a method of reducing the incidence or relapse of a tumor or cancer, comprising the step of administering to the subject the combination therapy provided herein. In another embodiment, the present invention provides a method of suppressing the formation of a tumor in a subject, comprising the step of administering to the subject the combination therapy provided herein. In another embodiment, the present invention provides a method of inducing a remission of a cancer in a subject, comprising the step of administering to the subject the combination therapy provided herein. In another embodiment, the present invention provides a method of extending a remission of a tumor or cancer in a subject, comprising the step of administering to the subject the combination therapy provided herein. In another embodiment, the present invention provides a method of reducing the size of a tumor a subject, comprising the step of administering to the subject the combination therapy provided herein. In one embodiment, the nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide is integrated into the Listeria genome. In another embodiment, the nucleic acid is in a plasmid in the recombinant Listeria vaccine strain. In another embodiment, the nucleic acid molecule is in a bacterial artificial chromosome in the recombinant Listeria vaccine strain.
[00313] In another embodiment, a method of treating reduces tumor size. Reduction of tumor size may be partial or by 100%. In another embodiment, methods disclosed herein reduce tumor size by 90%. In another embodiment, methods disclosed herein reduce tumor size by 80%. In another embodiment, methods reduce tumor size by 70%. In another embodiment, methods reduce tumor size by 60%. In another embodiment, methods reduce tumor size by 50%.
[00314] In another embodiment, a method of treating increases the time to disease progression. In one embodiment, the time to disease progression was increased by at least 2 moths as compared to untreated subject. In one embodiment, the time to disease progression was increased by at least 4 moths as compared to untreated subject. In another embodiment, the time to disease progression was increased by at least 6 moths as compared to untreated subject. In one embodiment, the time to disease progression was increased by at least 1 year as compared to untreated subject. In one embodiment, the time to disease progression was increased by at least 2 years as compared to untreated subject. In one embodiment, the time to disease progression was increased by at least 3 years as compared to untreated subject. In one embodiment, the time to disease progression was increased by at least 4 years as compared to untreated subject. In one embodiment, the time to disease progression was increased by at least 5 years as compared to untreated subject.
[00315] In another embodiment, the present invention provides a method of impeding a growth of a survivin-expressing cancer in a subject, comprising administering to the subject a combination therapy provided herein, wherein the recombinant polypeptide comprising an N- terminal fragment of a LLO protein fused to a survivin antigen, and wherein the antigen has one or more subdominant CD8+ T cell epitopes. In another embodiment, the antigen does not contain any of the dominant CD8+ T cell epitopes. [00316] "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 or 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 CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 45% of the antigen- specific CD8+ 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 refers to an epitope recognized by over 60% of the antigen- specific CD8+ 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 term 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 term refers to an epitope recognized by over 80% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by over 85% of the antigen- specific CD 8+ T cells. In another embodiment, the term refers to an epitope recognized by over 90% of the antigen- specific CD8+ 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 CD8+ T cells.
[00317] "Subdominant CD8+ T cell epitope" refers to an epitope recognized by fewer than 30% of the antigen- specific CD8+ 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 CD8+ 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 over 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 over 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 term refers to an epitope recognized by fewer than 6% of the antigen- specific CD8+ 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 CD8+ 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 term refers to an epitope recognized by fewer than 1% of the antigen- specific CD8+ T cells. In another embodiment, the term refers to an epitope recognized by fewer than 0.5% of the antigen- specific CD8+ T cells.
[00318] In one embodiment, vaccination with recombinant Listeria expressing the LLO-antigen fusions disclosed herein induces epitope spreading. In another embodiment, vaccination with LLO- antigen fusions, even outside the context of survivin, induces epitope spreading as well.
[00319] 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.
[00320] The antigen in methods and compositions disclosed herein is, in one embodiment, expressed at a detectable level on a non-tumor 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. In another embodiment, the non-tumor cell is a germ cell.
[00321] "Detectable level" refers, in one embodiment, to a level that is detectable when using a standard assay. In one embodiment, the assay is an immunological assay. In one embodiment, the assay is enzyme-linked immunoassay (ELISA). In another embodiment, the assay is Western blot. In another embodiment, the assay is FACS. In yet another embodiment, the assay is Western blot. In another embodiment, the assay is PCR. It is to be understood by a skilled artisan that other assays available in the art can be used in the methods provided herein. 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.
[00322] 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 combination therapy of the present invention, thereby inducing formation of cytotoxic T cells in a host having cancer.
[00323] In one embodiment, disclosed herein is a method of administering a composition of the present invention. In another embodiment, disclosed herein is a method of administering a vaccine of the present invention. In another embodiment, disclosed herein is a method of administering a recombinant polypeptide or recombinant nucleotide of the present invention. In another embodiment, the step of administering a composition, recombinant polypeptide or recombinant nucleotide disclosed herein is performed with an attenuated recombinant form of Listeria comprising a recombinant nucleotide or expressing a recombinant polypeptide, each in its own discrete embodiment. In another embodiment, the administering is performed with a different attenuated bacterial vector. In another embodiment, the administering is performed with a DNA vaccine (e.g. a naked DNA vaccine). In another embodiment, administration of a recombinant polypeptide disclosed herein is performed by producing the protein recombinantly, then administering the recombinant protein to a subject.
[00324] In one embodiment, a 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, and then is transferred to the subject.
[00325] Various administration regimens are contemplated by this invention. Each therapeutic agent in a combination therapy of the invention may be administered simultaneously (i.e., in the same medicament), concurrently (i.e., in separate medicaments administered one right after the other in any order) or sequentially in any order. Sequential administration is particularly useful when the therapeutic agents in the combination therapy are in different dosage forms (one agent is a tablet or capsule and another agent is a sterile liquid) and/or are administered on different dosing schedules, e.g., a chemotherapeutic that is administered at least daily and a biotherapeutic that is administered less frequently, such as once weekly, once every two weeks, or once every three weeks.
[00326] It will be appreciated by a skilled artisan that the term "simultaneous administration" in relation to the administration of medicaments may encompass the administration of medicaments such that the individual medicaments are delivered to a subject at the same time, through a same or different routes of administration.
[00327] Various embodiments of dosage ranges are contemplated by this invention.
Dosage units for an immunosuppressive molecule antagonist (e.g., PD-1 antagonist antibody) may be expressed as a flat dose, i.e., 100 mg, 200 mg, 300 mg, or as a patient-specific dose, i.e., mg/kg (mg therapeutic agent/kg of body weight) or mg/m (quantity in milligrams per square meter of body surface area).
[00328] In one embodiment, the dose of the attenuated Listeria strain comprised by the immunogenic composition disclosed herein is administered to a subject at a dose of 1 x 10 - 3.31 x 1010 CFU. In another embodiment, the dose is 1 x 108 - 3.31 x 1010 CFU. In another embodiment, the dose is 1 x 109 - 3.31 x 1010 CFU. In another embodiment, the dose is 5-500 x
10 8 CFU. In another embodiment, the dose is 7-500 x 108 CFU. In another embodiment, the dose is 10-500 x 108 CFU. In another embodiment, the dose is 20-500 x 108 CFU. In another embodiment, the dose is 30-500 x 10 8 CFU. In another embodiment, the dose is 50-500 x 108 CFU. In another embodiment, the dose is 70-500 x 10 CFU. In another embodiment, the dose is 100-500 x 108 CFU. In another embodiment, the dose is 150-500 x 108 CFU. In another embodiment, the dose is 5-300 x 10 8 CFU. In another embodiment, the dose is 5-200 x 108 CFU. In another embodiment, the dose is 5-150 x 10 CFU. In another embodiment, the dose is 5-100 x
10 8 CFU. In another embodiment, the dose is 5-70 x 108 CFU. In another embodiment, the dose is 5-50 x 10 8 CFU. In another embodiment, the dose is 5-30 x 108 CFU. In another embodiment, the dose is 5-20 x 10 8 CFU. In another embodiment, the dose is 1-30 x 109 CFU. In another embodiment, the dose is 1-20 x 109 CFU. In another embodiment, the dose is 2-30 x 109 CFU. In another embodiment, the dose is 1-10 x 109 CFU. In another embodiment, the dose is 2-10 x 109 CFU. In another embodiment, the dose is 3-10 x 109 CFU. In another embodiment, the dose is 2- 7 x 109 CFU. In another embodiment, the dose is 2-5 x 109 CFU. In another embodiment, the dose is 3-5 x 109 CFU. [00329] In another embodiment, the dose is 1 x 10 organisms. In another embodiment, the dose is 1 x 10 8 organisms. In another embodiment, the dose is 1 x 109 organisms. In another embodiment, the dose is 1.5 x 109 organisms. In another embodiment, the dose is 2 x 109 organisms. In another embodiment, the dose is 3 x 109 organisms. In another embodiment, the dose is 4 x 109 organisms. In another embodiment, the dose is 5 x 109 organisms. In another embodiment, the dose is 6 x 109 organisms. In another embodiment, the dose is 7 x 109 organisms. In another embodiment, the dose is 8 x 109 organisms. In another embodiment, the dose is 10 x 109 organisms. In another embodiment, the dose is 1.5 x 1010 organisms. In another embodiment, the dose is 2 x 1010 organisms. In another embodiment, the dose is 2.5 x 1010 organisms. In another embodiment, the dose is 3 x 1010 organisms. In another embodiment, the dose is 3.3 x 1010 organisms. In another embodiment, the dose is 4 x 1010 organisms. In another embodiment, the dose is 5 x 1010 organisms. Each dose and range of doses represents a separate embodiment of the present invention.
[00330] In one embodiment, repeat administrations (doses) of compositions disclosed hereinmay be undertaken immediately following the first course of treatment or after an interval of days, weeks or months to achieve tumor regression. In another embodiment, repeat doses may be undertaken immediately following the first course of treatment or after an interval of days, weeks or months to achieve suppression of tumor growth. Assessment may be determined by any of the techniques known in the art, including diagnostic methods such as imaging techniques, analysis of serum tumor markers, biopsy, or the presence, absence or amelioration of tumor associated symptoms.
[00331] It will be appreciated by the skilled artisan that the term "Boosting" may encompass administering an additional strain or immunogenic composition or recombinant Listeria strain dose or immune checkpoint inhibitor alone or in combination to a subject. In another embodiment, of methods of the present invention, 2 boosts (or a total of 3 inoculations) are administered. In another embodiment, 3 boosts are administered. In another embodiment, 4 boosts are administered. In another embodiment, 5 boosts are administered. In another embodiment, 6 boosts are administered. In another embodiment, more than 6 boosts are administered. Each possibility represents a separate embodiment of the present invention.
[00332] In another embodiment, a method of present invention further comprises the step of boosting the subject with a recombinant Listeria strain or immune checkpoint inhibitor as provided herein. In another embodiment, the recombinant Listeria strain used in the booster inoculation is the same as the strain used in the initial "priming" inoculation. In another embodiment, the booster strain is different from the priming strain. In another embodiment, the recombinant immune checkpoint inhibitor used in the booster inoculation is the same as the inhibitor used in the initial "priming" inoculation. In another embodiment, the booster inhibitor is different from the priming inhibitor. In another embodiment, the same doses are used in the priming and boosting inoculations. In another embodiment, a larger dose is used in the booster. In another embodiment, a smaller dose is used in the booster. In another embodiment, the methods disclosed herein further comprise the step of administering to the subject a booster vaccination. In one embodiment, the booster vaccination follows a single priming vaccination. In another embodiment, a single booster vaccination is administered after the priming vaccinations. In another embodiment, two booster vaccinations are administered after the priming vaccinations. In another embodiment, three booster vaccinations are administered after the priming vaccinations. In one embodiment, the period between a prime and a boost strain is experimentally determined by the skilled artisan. In another embodiment, the period between a prime and a boost strain is 1 week, in another embodiment it is 2 weeks, in another embodiment, it is 3 weeks, in another embodiment, it is 4 weeks, in another embodiment, it is 5 weeks, in another embodiment it is 6-8 weeks, in yet another embodiment, the boost strain is administered 8-10 weeks after the prime strain.
[00333] In another embodiment, a method disclosed herein further comprises boosting the subject with a immunogenic composition comprising an attenuated Listeria strain provided herein. In another embodiment, a method disclosed herein comprises the step of administering a booster dose of the immunogenic composition comprising the attenuated Listeria strain provided herein. In another embodiment, the booster dose is an alternate form of the immunogenic composition. In another embodiment, the methods disclosed herein further comprise the step of administering to the subject a booster immunogenic composition. In one embodiment, the booster dose follows a single priming dose of the immunogenic composition. In another embodiment, a single booster dose is administered after the priming dose. In another embodiment, two booster doses are administered after the priming dose. In another embodiment, three booster doses are administered after the priming dose. In one embodiment, the period between a prime and a boost dose of an immunogenic composition comprising the attenuated Listeria disclosed herein is experimentally determined by the skilled artisan. In another embodiment, the dose is experimentally determined by a skilled artisan. In another embodiment, the period between a prime and a boost dose is 1 week, in another embodiment it is 2 weeks, in another embodiment, it is 3 weeks, in another embodiment, it is 4 weeks, in another embodiment, it is 5 weeks, in another embodiment it is 6-8 weeks, in yet another embodiment, the boost dose is administered 8-10 weeks after the prime dose of the immunogenic composition.
[00334] Heterologous "prime boost" strategies have been effective for enhancing immune responses and protection against numerous pathogens. Schneider et al., Immunol. Rev. 170:29-38 (1999); Robinson, H. L., Nat. Rev. Immunol. 2:239-50 (2002); Gonzalo, R. M. et al., Strain 20: 1226-31 (2002); Tanghe, A., Infect. Immun. 69:3041-7 (2001). Providing antigen in different forms in the prime and the boost injections appears to maximize the immune response to the antigen. DNA strain priming followed by boosting with protein in adjuvant or by viral vector delivery of DNA encoding antigen appears to be the most effective way of improving antigen specific antibody and CD4+ T-cell responses or CD8+ T-cell responses respectively. Shiver J. W. et al., Nature 415: 331-5 (2002); Gilbert, S. C. et al., Strain 20: 1039-45 (2002); Billaut-Mulot, O. et al., Strain 19:95-102 (2000); Sin, J. I. et al., DNA Cell Biol. 18:771-9 (1999). Recent data from monkey vaccination studies suggests that adding CRL1005 poloxamer (12 kDa, 5% POE), to DNA encoding the HIV gag antigen enhances T-cell responses when monkeys are vaccinated with an HIV gag DNA prime followed by a boost with an adenoviral vector expressing HIV gag (Ad5-gag). The cellular immune responses for a DNA/poloxamer prime followed by an Ad5-gag boost were greater than the responses induced with a DNA (without poloxamer) prime followed by Ad5-gag boost or for Ad5-gag only. Shiver, J. W. et al. Nature 415:331-5 (2002). U.S. Patent Appl. Publication No. US 2002/0165172 Al describes simultaneous administration of a vector construct encoding an immunogenic portion of an antigen and a protein comprising the immunogenic portion of an antigen such that an immune response is generated. The document is limited to hepatitis B antigens and HIV antigens. Moreover, U.S. Pat. No. 6,500,432 is directed to methods of enhancing an immune response of nucleic acid vaccination by simultaneous administration of a polynucleotide and polypeptide of interest. According to the patent, simultaneous administration means administration of the polynucleotide and the polypeptide during the same immune response, preferably within 0-10 or 3-7 days of each other. The antigens contemplated by the patent include, among others, those of Hepatitis (all forms), HSV, HIV, CMV, EBV, RSV, VZV, HPV, polio, influenza, parasites (e.g., from the genus Plasmodium), and pathogenic bacteria (including but not limited to M. tuberculosis, M. leprae, Chlamydia, Shigella, B. burgdorferi, enterotoxigenic E. coli, S. typhosa, H. pylori, V. cholerae, B. pertussis, etc.). All of the above references are herein incorporated by reference in their entireties. [00335] In another embodiment, the recombinant polypeptide of methods disclosed herein is expressed by the recombinant Listeria strain. In another embodiment, the expression is mediated by a nucleotide molecule carried by the recombinant Listeria strain. Each possibility represents a separate embodiment of the present invention.
[00336] In some instances, the immunosuppressive molecule antagonist and the live- attenuated Listeria strain that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells are combined in a single dosage form. In some instances, the immunosuppressive molecule antagonist and the live-attenuated Listeria monocytogenes strain bioengineered, by transforming it with an expression vector to express a survivin antigen fused to a tLLO are combined in a single dosage form. In some instances, the immunosuppressive molecule antagonist and the an ADXS31-265 (10403S
Figure imgf000083_0001
pADV265) strain are combined in a single dosage form.
[00337] In one embodiment, administration of an immunosuppressive molecule antagonist and a live-attenuated Listeria strain disclosed herein that is used to stimulate APCs capable of driving a cellular immune response to survivin expressing cells is maintained throughout a period of treatment or prevention. In another embodiment, anti-cancer activity is achieved by subsequent administration of either component in isolation, i.e.- the immunosuppressive molecule antagonist or the live-attenuated Listeria strain (or a composition comprising either component).
[00338] In some embodiments, the live-attenuated Listeria strain that is used to stimulate
APCs capable of driving a cellular immune response to survivin expressing cells (e.g. ADXS31- 265 (10403S
Figure imgf000084_0001
pADV265) strain) is administered before administration of the immunosuppressive molecule antagonist, while in other embodiments, one of the live-attenuated Listeria strains disclosed herein is administered after administration of the immunosuppressive molecule antagonist. Each possibility represents a separate embodiment of the methods and compositions as provided herein.
[00339] Various modes of sequential administration are contemplated by this invention. In one embodiment, an administration regimen comprises administering an immunosuppressive molecule antagonist disclosed herein followed by administration of a recombinant Listeria vaccine strain provided herein. In another embodiment, the order of administration of components of combination therapy is reversed. In yet another embodiment, administration of one component is immediately followed by administration of the other component. In yet another embodiment, there is an interval between administrations of components. In one embodiment, the interval is at least 1-2 hours. In another embodiment, the interval is at least 2-3 hours. In yet another embodiment, the interval is at least 3-4 hours. In another embodiment, the interval is at least 4-5 hours. In yet another embodiment, the interval is at least 5-6 hours. In another embodiment, the interval is at least 6-8 hours. In yet another embodiment, the interval at least is 8-10 hours. In another embodiment, the interval is at least 10-12 hours. In another embodiment, the interval is at least one day. In another embodiment, the interval is at least two days. In another embodiment, the interval is at least three days. In another embodiment, the interval is at least four days. In another embodiment, the interval is at least five days. In another embodiment, the interval is at least six days. In another embodiment, the interval is at least seven days. In yet another embodiment, the interval is more than seven days.
[00340] In some embodiments, at least one of the therapeutic agents in the combination therapy is administered using the same dosage regimen (dose, frequency and duration of treatment) that is typically employed when the agent is used as monotherapy for treating the same cancer. In other embodiments, the patient receives a lower total amount of at least one of the therapeutic agents in the combination therapy than when the agent is used as monotherapy, e.g., smaller doses, less frequent doses, and/or shorter treatment duration.
[00341] In one embodiment, the method comprises the step of co-administering a composition comprising a recombinant Listeria with an additional therapy. In another embodiment, the additional therapy is surgery, chemotherapy, an immunotherapy, a radiation therapy, an antibody based immunotherapy provided herein, or a combination thereof. In another embodiment, the additional therapy precedes administration of a composition comprising a recombinant Listeria. In another embodiment, the additional therapy follows administration of the composition comprising a recombinant Listeria. In another embodiment, the additional therapy is an antibody therapy. In another embodiment, a composition comprising a recombinant Listeria is administered in increasing doses in order to increase the T-effector cell to regulatory T cell ration and generate a more potent anti-tumor immune response. It will be appreciated by a skilled artisan that the anti-tumor immune response can be further strengthened by providing the subject having a tumor with cytokines including, but not limited to IFN-γ, TNF-a, and other cytokines known in the art to enhance cellular immune response, some of which can be found in US Patent Serial No. 6,991,785, incorporated by reference herein. In another embodiment, an immunosuppressive molecule, a recombinant Listeria and an additional therapy are administered separately. [00342] Various modes of sequential administration are contemplated by this invention. In one embodiment, an administration regimen comprises administering a combination therapy disclosed herein via any regimen described above, followed by administration of one of additional therapies described above. In another embodiment, the order of administration of combination therapy and one of the additional therapies is reversed. In yet another embodiment, administration of the combination therapy is immediately followed or preceded by administration of one of the additional therapies. In yet another embodiment, there is an interval between administrations therapies. In one embodiment, the interval is at least 1-2 hours. In another embodiment, the interval is 2-3 hours. In another embodiment, the interval is 3-4 hours. In another embodiment, the interval is 4-5 hours. In another embodiment, the interval is 5-6 hours. In another embodiment, the interval is 6-8 hours. In another embodiment, the interval is 7-10 hours. In another embodiment, the interval is 9-12 hours. In another embodiment, the interval is at least one day. In another embodiment, the interval is two to three days. In another embodiment, the interval is three to five days. In another embodiment, the interval is five to seven days. In another embodiment, the interval is seven to ten days. In yet another embodiment, the interval is ten to fifteen days.
[00343] In another embodiment, disclosed herein is a method of increasing antigen- specific T cells in a subject suffering from cancer or having a tumor, the method comprising the step of administering to the subject an immunogenic composition comprising a recombinant Listeria vaccine strain comprising a nucleic acid molecule, the nucleic acid molecule comprising a first open reading frame encoding fusion polypeptide, wherein the fusion polypeptide comprises a PEST sequence-containing polypeptide or PEST-sequence containing peptide fused to survivin antigen or fragment thereof, as described herein.
[00344] In some embodiments, a combination therapy of the invention is administered to a patient who has not been previously treated with a biotherapeutic or chemotherapeutic agent, i.e., is treatment-naive. In other embodiments, the combination therapy is administered to a patient who failed to achieve a sustained response after prior therapy with a biotherapeutic or chemotherapeutic agent, i.e., is treatment-experienced.
[00345] It will be appreciated by a skilled artisan that the term "RECIST 1.1 Response
Criteria" may encompass the definitions set forth in Eisenhauer et al., E.A. et al., Eur. J Cancer 45:228-247 (2009) for target lesions or non-target lesions, as appropriate based on the context in which response is being measured. [00346] It will be appreciated by a skilled artisan that the term "Sustained response" may encompass a sustained therapeutic effect after cessation of treatment with a therapeutic agent, or a combination therapy described herein. In some embodiments, the sustained response has a duration that is at least the same as the treatment duration, or at least 1.5, 2.0, 2.5 or 3 times longer than the treatment duration. [00347] It will be appreciated by a skilled artisan that the Bliss independence combined response C for two single compounds with effects A and B is C = A + B - A*B, where each effect is expressed as a fractional inhibition between 0 and 1. (Reference: Bliss (1939) Annals of Applied Biology) The Bliss value, defined to be the difference between the experimental response and the calculated Bliss Independence value, indicates whether the two compounds in combination are additive or synergistic.
[00348] A Bliss value of zero (0) is considered additive. The term "additive" means that the result of the combination of the two targeted agents is the sum of each agent individually.
[00349] It will be appreciated by a skilled artisan that the term"Chothia" may encompass an antibody numbering system described in Al-Lazikani et al., JMB 273:927-948 (1997). [00350] It will be appreciated by a skilled artisan that the terms "synergy" or "synergistic" may encompass the response of the combination of the two agents is more than the sum of each agent's individual response. More specifically, in the in vitro setting one measure of synergy is known as "Bliss synergy." Bliss synergy may encompass "excess over Bliss independence", as determined by the Bliss value defined above. When the Bliss value is greater than zero (0), or more preferably greater than 0.2, it is considered indicative of synergy. Of course, the use of "synergy" herein also encompasses in vitro synergy as measured by additional and/or alternate methods. References herein to a combination's in vitro biological effects, including but not limited to anti-cancer effects, being greater than, or equal to, the sum of the combination's components individually, may be correlated to Bliss values. Again, the use of "synergy" herein, including whether a combination of components demonstrates activity equal to or greater than the sum of the components individually, may be measured by additional and/or alternate methods and are known, or will be apparent, to those skilled in this art.
[00351] A combination therapy of the invention is typically used to treat a tumor that is large enough to be found by palpation or by imaging techniques well known in the art, such as MRI, ultrasound, or CAT scan. In some embodiments, a combination therapy of the invention is used to treat an advanced stage tumor having dimensions of at least about 200 mm 3 ' 300 mm 3 , 400 mm 3 , 500 mm 3 , 750 mm 3 , or up to 1000 mm 3.
[00352] In one embodiment, a combination therapy of the invention is administered to a patient diagnosed with a pancreatic cancer that tests positive for PD-L1 expression. In some embodiments, PD-L1 expression is detected using a diagnostic anti-human PD-L1 antibody, or antigen binding fragment thereof, in an IHC assay on an FFPE or frozen tissue section of a tumor sample removed from the patient. Typically, the patient's physician would order a diagnostic test to determine PD-L1 expression in a tumor tissue sample removed from the patient prior to initiation of treatment with the PD-1 antagonist or PD-L1 antagonist and the live-attenuated Listeria strains provided for herein, but it is envisioned that the physician could order the first or subsequent diagnostic tests at any time after initiation of treatment, such as for example after completion of a treatment cycle.
[00353] In some embodiments, selecting a dosage regimen (also referred to herein as an administration regimen) for a combination therapy of the invention depends on several factors, including the serum or tissue turnover rate of the entity, the level of symptoms, the immunogenicity of the entity, and the accessibility of the target cells, tissue or organ in the individual being treated. Preferably, a dosage regimen maximizes the amount of each therapeutic agent delivered to the patient consistent with an acceptable level of side effects. Accordingly, the dose amount and dosing frequency of each biotherapeutic and chemotherapeutic agent in the combination depends in part on the particular therapeutic agent, the severity of the cancer being treated, and patient characteristics. Guidance in selecting appropriate doses of antibodies, cytokines, and small molecules are available. See, e.g., Wawrzynczak (1996) Antibody Therapy, Bios Scientific Pub. Ltd, Oxfordshire, UK; Kresina (ed.) (1991) Monoclonal Antibodies, Cytokines and Arthritis, Marcel Dekker, New York, NY; Bach (ed.) (1993) Monoclonal Antibodies and Peptide Therapy in Autoimmune Diseases, Marcel Dekker, New York, NY; Baert et al. (2003) New Engl. J. Med. 348:601-608; Milgrom et al. (1999) New Engl. J. Med. 341 : 1966- 1973; Slamon et al. (2001) New Engl. J. Med. 344:783-792; Beniaminovitz et al. (2000) New Engl. J. Med. 342:613-619; Ghosh et al. (2003) New Engl. J. Med. 348:24-32; Lipsky et al. (2000) New Engl. J. Med. 343: 1594- 1602; Physicians' Desk Reference 2003 (Physicians' Desk Reference, 57th Ed); Medical Economics Company; ISBN: 1563634457; 57th edition (November 2002). Determination of the appropriate dosage regimen may be made by the clinician, e.g., using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment, and will depend, for example, the patient's clinical history (e.g., previous therapy), the type and stage of the cancer to be treated and biomarkers of response to one or more of the therapeutic agents in the combination therapy.
[00354] Biotherapeutic agents in a combination therapy of the invention may be administered by continuous infusion, or by doses at intervals of, e.g., daily, every other day, three times per week, or one time each week, two weeks, three weeks, monthly, bimonthly, etc. A total weekly dose is generally at least 0.05 μg/kg, 0.2 μg/kg, 0.5 μg/kg, 1 μg/kg, 10 μg/kg, 100 μg/kg, 0.2 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg body weight or more. See, e.g., Yang et al. (2003) New Engl. J. Med. 349:427-434; Herald et al. (2002) New Engl. J. Med. 346: 1692- 1698; Liu et al. (1999) J. Neurol. Neurosurg. Psych. 67:451-456; Portielji et al. (20003) Cancer Immunol. Immunother. 52: 133-144.
[00355] In some embodiments that employ an anti-human PD- 1 mAb as the PD- 1 antagonist in the combination therapy, the dosing regimen will comprise administering the anti- human PD-1 mAb at a flat dose of 100 to 500 mg or a weight-based dose of 1 to 10 mg/kg at intervals of about 14 days (+ 2 days) or about 21 days (+ 2 days) or about 30 days (+ 2 days) throughout the course of treatment.
[00356] In other embodiments that employ an anti-human PD- 1 mAb as the PD- 1 antagonist in the combination therapy, the dosing regimen will comprise administering the anti- human PD-1 mAb at a dose of from about 0.005 mg/kg to about 10 mg/kg, with intra-patient dose escalation. In other escalating dose embodiments, the interval between doses will be progressively shortened, e.g., about 30 days (+ 2 days) between the first and second dose, about 14 days (+ 2 days) between the second and third doses. In certain embodiments, the dosing interval will be about 14 days (+ 2 days), for doses subsequent to the second dose.
[00357] In one embodiment, the terms "treatment regimen", "dosing protocol" and "dosing regimen" are used interchangeably herein and encompass the dose and timing of administration of each therapeutic agent in a combination of the invention.
[00358] In certain embodiments, a subject will be administered an intravenous (IV) infusion of a medicament comprising any of the immunosuppressive molecules antagonists described herein.
[00359] In one embodiment, of the invention, the PD-1 antagonist in the combination therapy is nivolumab, which is administered intravenously at a dose selected from the group consisting of: 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W.
[00360] In another embodiment, of the invention, the PD-1 antagonist in the combination therapy PD-1 antagonist is administered in a liquid medicament at a dose selected from the group consisting of 200 mg Q3W, 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W or equivalents of any of these doses (e.g., a PK model of a PD-1 antagonist estimates that the fixed dose of 200 mg Q3W provides exposures that are consistent with those obtained with 2 mg/kg Q3W). In some embodiments, a PD-1 antagonist is administered as a liquid medicament which comprises 25 mg/ml the PD-1 antagonist, 7% (w/v) sucrose, 0.02% (w/v) polysorbate 80 in 10 mM histidine buffer pH 5.5, and the selected dose of the medicament is administered by IV infusion over a time period of 30 minutes +/-10 min.
[00361] In another embodiment, of the invention, the attenuated bacterial or attenuated
Listeria in the combination therapy is a live-attenuated Listeria strain disclosed herein (e.g. ADXS31-265), which is administered in a liquid medicament at a dose selected from the group consisting of 1 x 109, 5 x 109 and 1 x 1010 CFU. In some embodiments, a dose ranges from about 1 x 109 CFU up to 3.31 x 1010 CFU, from about 5 x 108 CFU up to 5 x 1010 CFU, from about 7 x 108 CFU up to 5 x 1010 CFU, from about 1 x 109 CFU up to 5 x 1010 CFU, from about 2 x 109 CFU up to 5 x 1010 CFU, from about 3 x 109 CFU up to 5 x 1010 CFU, from about 5 x 109 CFU up to 5 x 1010 CFU, from about 7 x 109 CFU up to 5 x 1010 CFU, from about 1 x 1010 CFU up to 5 x 1010 CFU, from about 1.5 x 109 CFU up to 5 x 1010 CFU, from about 5 x 10° CFU - up to 3 x 1010 CFU, from about 5 x 10° CFU up to 2 x 1010 CFU, from about 5 x 10° CFU up to 1.5 x 109 CFU, from about 5 x 10° CFU up to 1 x 1010 CFU, from about 5 x 10° CFU up to 7 x 109 CFU, from about 5 x 10° CFU up to 5 x 109 CFU, from about 5 x 10° CFU up to 3 x 109 CFU, from about 5 x 10° CFU up to 2 x 109 CFU, from about 1 x 109 CFU up to 3 x 1010 CFU, from about 1 x 109 CFU up to 2x 1010CFU, from about 2 x 109 CFU up to 3 x 1010 CFU, from about 1 x 109 CFU up to lx 1010 CFU, from about 2 x 109 CFU up to 1 x 1010 CFU, from about 3 x 109 CFU up to 1 x 1010 CFU, from about 2 x 109 CFU up to 7 x 109 CFU, from about 2 x 109 CFU up to 5 x 109 CFU. Each possibility represents a separate embodiment of the present invention.
[00362] In other embodiments, a dose of recombinant Listeria ranges from about 1 x 10 organisms to aboutl.5 x 10 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 10 8 organisms to about 1.5 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 109 organisms to about 2 x 109 organisms. -, In another embodiment, a dose of recombinant Listeria ranges from about 2 x 109 organisms to about 5 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 2 x 109 organisms to about 1 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 3 x 109 organisms to about 1 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 4 x 109 organisms to about 1 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 1 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 6 x 109 organisms to about 1 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 7 x 109 organisms to about 1 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 109 organisms to about 5 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 109 organisms to about 4 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 1 x 109 organisms to about 3 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 8 x 109 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 1.5 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 2 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 2.5 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 3 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 3.5 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 4 x 1010 organisms. In another embodiment, a dose of recombinant Listeria ranges from about 5 x 109 organisms to about 5 x 1010 organisms. In another embodiment, the dose ranges
7 10
from 1 x 10 organisms-5 x 10 organisms.
[00363] In some embodiments, pharmaceutical compositions containing strains and compositions disclosed herein are 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, subcutaneously, intra-peritonealy, intra- ventricularly, intra-cranially, intra-vaginally or intra-tumorally.
[00364] The antibodies and compositions provided by the present disclosure can be administered via any suitable enteral route or parenteral route of administration. It will be appreciated by a skilled artisan that the term "enteral route" of administration may encompass the administration via any part of the gastrointestinal tract. Examples of enteral routes include oral, mucosal, buccal, and rectal route, or intragastric route. It will also be appreciated by a skilled artisan that the term "Parenteral route" of administration may encompass a route of administration other than enteral route. Examples of parenteral routes of administration include intravenous, intramuscular, intradermal, intraperitoneal, intratumor, intravesical, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, transtracheal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal, subcutaneous, or topical administration. The antibodies and compositions of the disclosure can be administered using any suitable method, such as by oral ingestion, nasogastric tube, gastrostomy tube, injection, infusion, implantable infusion pump, and osmotic pump. The suitable route and method of administration may vary depending on a number of factors such as the specific antibody being used, the rate of absorption desired, specific formulation or dosage form used, type or severity of the disorder being treated, the specific site of action, and conditions of the patient, and can be readily selected by a person skilled in the art.
[00365] In another embodiment, of the methods and compositions provided herein, the strains 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 formulations 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 disclosed herein comprise, in addition to the active compound and the inert carrier or diluent, a hard gelating capsule.
[00366] In another embodiment, a composition comprising a Listeria strain is 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.
[00367] In another embodiment, a vaccine or composition disclosed herein is 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. [00368] In one embodiment, a vaccine of the methods and compositions disclosed herein may be administered to a host vertebrate animal, preferably a mammal, and more preferably a human, either alone or in combination with a pharmaceutically acceptable carrier. In another embodiment, a vaccine is administered in an amount effective to induce an immune response to the Listeria strain itself or to a heterologous antigen which the Listeria species has been modified to express. In another embodiment, the amount of vaccine or immunogenic composition to be administered may be routinely determined by one of skill in the art when in possession of the present disclosure. In another embodiment, a pharmaceutically acceptable carrier may include, but is not limited to, sterile distilled water, saline, phosphate buffered solutions or bicarbonate buffered solutions. In another embodiment, the pharmaceutically acceptable carrier selected and the amount of carrier to be used will depend upon several factors including the mode of administration, the strain of Listeria and the age and disease state of the vaccinee. In another embodiment, administration of the vaccine may be by an oral route, or it may be parenteral, intranasal, intramuscular, intravascular, intrarectal, intraperitoneal, or any one of a variety of well-known routes of administration. In another embodiment, the route of administration may be selected in accordance with the type of infectious agent or tumor to be treated.
[00369] In another embodiment, the present invention provides a method of treating, suppressing, or inhibiting at least one tumor in a subject comprising administering the immunogenic composition provided herein.
[00370] In some embodiments an attenuated bacteria, or attenuated Listeria, or ADXS31-
265 is administered as a liquid medicament, and the selected dose of the medicament is administered by IV infusion over a time period of 30 minutes +/-10 min. [00371] The optimal dose for a combination therapy comprising an anti-PD-1 antibody disclosed herein in combination with a live-attenuated Listeria strain disclosed herein is identified by dose escalation of one or both of these agents. In another embodiment, the optimal dose for a composition comprising either the anti-PD-1 antibody disclosed herein or the live- attenuated Listeria strain disclosed herein is identified by dose escalation of one or both of these agents.
[00372] In one embodiment, a patient is treated with the combination therapy disclosed herein on day 1 of weeks 1, 4 and 7 in a 12 week cycle, starting with anti-PD-1 antibody is administered at a starting dose of 50, 100, 150, or 200 mg, and a live-attenuated Listeria strain disclosed herein at a starting dose of ranging from about 1 x 10 7 CFU to about 3.5 x 1010 CFU. [00373] In an embodiment, the anti-PD-1 antibody infusion is administered first, followed by a NSAIDS, e.g., naproxen or ibuprofen, and oral antiemetic medication within a predetermined amount of time prior to administration of a live-attenuated Listeria strain provided herein. In another embodiment, the predetermined amount of time is 5-10 min, 11-20 min, 21-40 min, 41-60 min. In another embodiment, the predetermined amount of time is at least one hour. In another embodiment, the predetermined amount of time is 1-2 hours, 2-4 hours, 4-6 hours, 6- 10 hours. In another embodiment, administrations of a NSAIDS, e.g., naproxen or ibuprofen, and oral antiemetic medication is repeated on a need basis to the subject, prior to administration of a live-attenuated Listeria strain provided herein.
[00374] In another embodiment, anti-PD-1 antibody is administered at a starting dose of
50, 100, 150 or 200 mg Q3W and a live-attenuated Listeria strain disclosed herein is administered Q3W at a starting dose of between 1 x 10 7 and 3.5 x 1010 CFU.
[00375] In another embodiment, a live-attenuated Listeria strain disclosed herein is administered at a starting dose of 5 x 109 Q3W and an anti-PD-1 antibody is administered at a starting dose of 200 mg Q3W, and if the starting dose of the combination is not tolerated by the patient, then the dose of the live-attenuated Listeria strain disclosed herein is reduced to 1 x 109 cfu Q3W or the dose of the anti-PD-1 antibody is reduced to 150 mg Q3W. It is to be understood by a skilled artisan that the doses of any of the components of a combination therapy disclosed herein may be incrementally adjusted to a lower or higher dose, as further provided herein, based on a subjects response to the combination therapy.
[00376] In some embodiments, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed, as determined by those skilled in the art.
[00377] In some embodiments, a treatment cycle begins with the first day of combination treatment and lasts for at least 12 weeks, 24 weeks or 48 weeks. On any day of a treatment cycle that the drugs are co-administered, the timing between the separate IV infusions of an anti-PDl antibody and a live-attenuated Listeria strain disclosed herein is between about 15 minutes to about 45 minutes. The invention contemplates that an anti-PD-1 antibody and a live-attenuated Listeria strain disclosed herein may be administered in either order or by simultaneous IV infusion.
[00378] In some embodiments, the combination therapy is administered for at least 2 to 4 weeks after the patient achieves a CR.
[00379] In some embodiments, a patient selected for treatment with the combination therapy of the invention has been diagnosed with a metastatic prostate cancer and the patient has progressed or become resistant to no more than 2 prior systemic treatment regimens. In some embodiments, a patient selected for treatment with the combination therapy of the invention has been diagnosed with a metastatic prostate cancer and the patient has progressed or become resistant to no more than 3 prior systemic treatment regimens. [00380] The present invention also provides a medicament which comprises a PD-1 antagonist disclosed herein and a pharmaceutically acceptable excipient. When the PD-1 antagonist is a biotherapeutic agent, e.g., a mAb, the antagonist may be produced in a producing cell line known in the art, such as, but not limited to CHO cells using conventional cell culture and recovery/purification technologies.
[00381] In some embodiments, a medicament comprising an anti-PD-1 antibody disclosed herein may be provided as a liquid formulation or prepared by reconstituting a lyophilized powder with sterile water for injection prior to use. WO 2012/135408 describes the preparation of liquid and lyophilized medicaments comprising an anti-PD-1 antibody that are suitable for use in the present invention. In some embodiments, a medicament comprising an anti-PD-1 antibody is provided in a glass vial which contains about 50 mg of anti-PD-1 antibody.
[00382] The present invention also provides a medicament which comprises a live- attenuated Listeria strain disclosed herein and a pharmaceutically acceptable excipient.
[00383] The PD-1 antagonist medicament and the live-attenuated Listeria strain disclosed herein medicament may be provided as a kit which comprises a first container and a second container and a package insert. The first container contains at least one dose of a medicament comprising an anti-PD-1 antibody, the second container contains at least one dose of a medicament comprising a live-attenuated Listeria strain provided herein, and the package insert, or label, which comprises instructions for treating a patient for a prostate cancer using the medicaments. The first and second containers may be comprised of the same or different shape (e.g., vials, syringes and bottles) and/or material (e.g., plastic or glass). The kit may further comprise other materials that may be useful in administering the medicaments, such as diluents, filters, IV bags and lines, needles and syringes. In some embodiments of the kit, the anti-PD-1 antagonist is an anti-PD-1 antibody and the instructions state that the medicaments are intended for use in treating a patient having a prostate cancer that tests positive for PD-L1 expression by an IHC assay.
[00384] It will be appreciated by a skilled artisan that the term "comprise" or grammatical forms thereof, may encompass the inclusion of the indicated active agent, such as the Lm strains of this invention, as well as inclusion of other active agents, such as an antibody or functional fragment thereof, and pharmaceutically acceptable carriers, excipients, emollients, stabilizers, etc., as are known in the pharmaceutical industry. In some embodiments, the term "consisting essentially of may encompass a composition, whose only active ingredient is the indicated active ingredient, however, other compounds may be included which are for stabilizing, preserving, etc. the formulation, but are not involved directly in the therapeutic effect of the indicated active ingredient. In some embodiments, the term "consisting essentially of may encompass components, which exert a therapeutic effect via a mechanism distinct from that of the indicated active ingredient. In some embodiments, the term "consisting essentially of may encompass components, which exert a therapeutic effect and belong to a class of compounds distinct from that of the indicated active ingredient. In some embodiments, the term "consisting essentially of may encompass components, which exert a therapeutic effect and may be distinct from that of the indicated active ingredient, by acting via a different mechanism of action. In some embodiments, the term "consisting essentially of may encompass components which facilitate the release of the active ingredient. In some embodiments, the term "consisting" may encompass a composition, which contains the active ingredient and a pharmaceutically acceptable carrier or excipient.
[00385] It is understood that wherever embodiments are described herein with the language "comprising", otherwise analogous embodiments described in terms of "consisting of and/or "consisting essentially of are also provided.
[00386] In one embodiment, the singular forms of words such as "a," "an," and "the," include their corresponding plural references unless the context clearly dictates otherwise.
[00387] Throughout this application, various embodiments disclosed hereinmay be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
[00388] Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases "ranging/ranges between" a first indicate number and a second indicate number and "ranging/ranges from" a first indicate number "to" a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals there between.
[00389] It will be appreciated by a skilled artisan that the term "About" when used to modify a numerically defined parameter (e.g., the dose of a PD-1 antagonist or the length of treatment time with a combination therapy described herein) may encompass variation of the parameter in quantitative terms plus or minus 5%, or in another embodiment plus or minus 10%, or in another embodiment plus or minus 15%, or in another embodiment plus or minus 20% of stated numerical value for that parameter. For example, a dose of about 200 mg of the PD-1 antagonist, may vary between 180 mg and 220 mg.
[00390] It is to be understood by the skilled artisan that the term "subject" can encompass a mammal including an adult human or a human child, teenager or adolescent in need of therapy for, or susceptible to, a condition or its sequelae, and also may include non-human mammals such as dogs, cats, pigs, cows, sheep, goats, horses, rats, and mice. It will also be appreciated that the term may encompass livestock. The term "subject" does not exclude an individual that is normal in all respects.
[00391] It will be appreciated by the skilled artisan that the term "mammal" for purposes of treatment refers to any animal classified as a mammal, including, but not limited to, humans, domestic and farm animals, and zoo, sports, or pet animals, such as canines, including dogs, and horses, cats, cattle, pigs, sheep, etc.
[00392] In the following examples, numerous specific details are set forth in order to provide a thorough understanding of the invention. However, it will be understood by those skilled in the art that the present invention may be practiced without these specific details. In other instances, well- known methods, procedures, and components have not been described in detail so as not to obscure the present invention. Thus these examples should in no way be construed, as limiting the broad scope of the invention.
EXAMPLES Materials and Methods
[00393] pADV265.5
[00394] DNA sequence of plasmid pAdv265.5 comprises
CGGAGTGTATACTGGCTTACTATGTTGGCACTGATGAGGGTGTCAGTGAAGTGCTTCATGTGGCAGGAGA AAAAAGGCTGCACCGGTGCGTCAGCAGAATATGTGATACAGGATATATTCCGCTTCCTCGCTCACTGACT CGCTACGCTCGGTCGTTCGACTGCGGCGAGCGGAAATGGCTTACGAACGGGGCGGAGATTTCCTGGAAG ATGCCAGGAAGATACTTAACAGGGAAGTGAGAGGGCCGCGGCAAAGCCGTT77TCC TAGGCTCCGCCCC CCTGACAAGCATCACGAAATCTGACGCTCAAATCAGTGGTGGCGAAACCCGACAGGACTATAAAGATACCAGGC GTTTCCCCCTGGCGGCTCCCTCGTGCGCTCTCCTGTTCCTGCCTTTCGGTTTACCGGTGTCATTCCGCTGTTATG GCCGCGTTTGTCTCATTCCACGCCTGACACTCAGTTCCGGGTAGGCAGTTCGCTCCAAGCTGGACTGTATGCAC GAACCCCCCGTTCAGTCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGAAAGACATGC AAAAGCACCACTGGCAGCAGCCACTGGTAATTGATTTAGAGGAGTTAGTCTTGAAGTCATGCGCCGGTTAAGGCT AAACTGAAAGGACAAGTTTTGGTGACTGCGCTCCTCCAAGCCAGTTACCTCGGTTCAAAGAGTTGGTAGCTCAGA GAACCTTCGAAAAACCGCCCTGCAAGGCGGTTTTTTCGTTTTCAGAGCAAGAGATTACGCGCAGACCAAAACGAT
CrCAAGAAGATCATCTTATTAATCAGATAAAATATTTCTAGCCCTCCTTTGATTAGTATATTCCTATCTTAA AGTTACTTTTATGTGGAGGCATTAACATTTGTTAATGACGTCAAAAGGATAGCAAGACTAGAATAAAGCT ATAAAGCAAGCATATAATATTGCGTTTCATCTTTAGAAGCGAATTTCGCCAATATTATAATTATCAAAAG AGAGGGGTGGCAAACGGTATTTGGCATTATTAGGTTAAAAAATGTAGAAGGAGAGTGAAACCCATGAAA AAAATAATGCTAGTTTTTATTACACTTATATTAGTTAGTCTACCAATTGCGCAACAAACTGAAGCAA AGGATGCATCTGCATTCAATAAAGAAAATTCAATTTCATCCATGGCACCACCAGCATCTCCGCCTG CAAGTCCTAAGACGCCAATCGAAAAGAAACACGCGGATGAAATCGATAAGTATATACAAGGATTGG ATTACAATAAAAACAATGTATTAGTATACCACGGAGATGCAGTGACAAATGTGCCGCCAAGAAAAG GTTACAAAGATGGAAATGAATATATTGTTGTGGAGAAAAAGAAGAAATCCATCAATCAAAATAATG CAGACATTCAAGTTGTGAATGCAATTTCGAGCCTAACCTATCCAGGTGCTCTCGTAAAAGCGAATT CGGAATTAGTAGAAAATCAACCAGATGTTCTCCCTGTAAAACGTGATTCATTAACACTCAGCATTGA TTTGCCAGGTATGACTAATCAAGACAATAAAATAGTTGTAAAAAATGCCACTAAATCAAACGTTAAC AACGCAGTAAATACATTAGTGGAAAGATGGAATGAAAAATATGCTCAAGCTTATCCAAATGTAAGT GCAAAAATTGATTATGATGACGAAATGGCTTACAGTGAATCACAATTAATTGCGAAATTTGGTACA GCATTTAAAGCTGTAAATAATAGCTTGAATGTAAACTTCGGCGCAATCAGTGAAGGGAAAATGCA AGAAGAAGTCATTAGTTTTAAACAAATTTACTATAACGTGAATGTTAATGAACCTACAAGACCTTCC AGATTTTTCGGCAAAGCTGTTACTAAAGAGCAGTTGCAAGCGCTTGGAGTGAATGCAGAAAATCCT CCTGCATATATCTCAAGTGTGGCGTATGGCCGTCAAGTTTATTTGAAATTATCAACTAATTCCCATA GTACTAAAGTAAAAGCTGCTTTTGATGCTGCCGTAAGCGGAAAATCTGTCTCAGGTGATGTAGAAC TAACAAATATCATCAAAAATTCTTCCTTCAAAGCCGTAATTTACGGAGGTTCCGCAAAAGATGAAGT TCAAATCATCGACGGCAACCTCGGAGACTTACGCGATATTTTGAAAAAAGGCGCTACTTTTAATCG AGAAACACCAGGAGTTCCCATTGCTTATACAACAAACTTCCTAAAAGACAATGAATTAGCTGTTATT AAAAACAACTCAGAATATATTGAAACAACTTCAAAAGCTTATACAGATGGAAAAATTAACATCGATC ACTCTGGAGGATACGTTGCTCAATTCAACATTTCTTGGGATGAAGTAAATTATGATCTCGAGGGTG CCCGACGTTGCCCCCTGCCTGGCAGCCCTTTCTCAAGGACCACCGCATCTCTACATTCAAGAACTGGCCCTTCTT GGAGGGCTGCGCCTGCGCCCCGGAGCGGATGGCCGAGGCTGGCTTCATCCACTGCCCCACTGAGAACGAGCC AGACTTGGCCCAGTGTTTCTTCTGCTTCAAGGAGCTGGAAGGCTGGGAGCCAGATGACGACCCCATAGAGGAAC ATAAAAAGCATTCGTCCGGTTGCGCTTTCCTTTCTGTCAAGAAGCAGTTTGAAGAATTAACCCTTGGTGAATT^
AAACTGGACAGAGAAAGAGCCAAGAACAAAATTGCAAAGGAAACCAACAATAAGAAGAAAGAATTTGAGGAAACT GCGAAGAAAGTGCGCCGTGCCATCGAGCAGCTGGCTGCCATGGATTGACCCGGGCCA T AA TCAACG T AG TAGTGGATTTAATCCCAAATGAGCCAACAGAACCAGAACCAGAAACAGAACAAGTAACATTGGAGTTAG AAATGGAAGAAGAAAAAAGCAATGATTTCGTGTGAATAATGCACGAAATCATTGCTTATTTTTTTAAAAA GCGATATACTAGATATAACGAAACAACGAACTGAATAAAGAATACAAAAAAAGAGCCACGACCAGTTAA AGCCTGAGAAACTTTAACTGCGAGCCTTAATTGATTACCACCAATCAATTAAAGAAGTCGAGACCCAAAA TTTGGTAAAGTATTTAATTACTTTATTAATCAGATACTTAAATATCTGTAAACCCATTATATCGGGTTTTTG AGGGGATTTCAAGTCTTTAAGAAGATACCAGGCAATCAATTAAGAAAAACTTAGTTGATTGCCTTTTTTG TTGTGATTCAACTTTGATCGTAGCTTCTAACTAATTAATTTTCGTAAGAAAGGAGAACAGCTGAATOAArA TCCCTTTTGTTGTAGAAACTGTGCTTCATGACGGCTTGTTAAAGTACAAATTTAAAAATAGTAAAATTCGCTC AATCACTACCAAGCCAGGTAAAAGTAAAGGGGCTATTTTTGCGTATCGCTCAAAAAAAAGCATGATTGGCGG ACGTGGCGTTGTTCTGACTTCCGAAGAAGCGATTCACGAAAATCAAGATACATTTACGCATTGGACACCAAA CGTTTATCGTTATGGTACGTATGCAGACGAAAACCGTTCATACACTAAAGGACATTCTGAAAACAATTTAAG ACAAATCAATACCTTCTTTATTGATTTTGATATTCACACGGAAAAAGAAACTATTTCAGCAAGCGATATTTTA ACAACAGCTATTGATTTAGGTTTTATGCCTACGTTAATTATCAAATCTGATAAAGGTTATCAAGCATATTTTG TTTTAGAAACGCCAGTCTATGTGACTTCAAAATCAGAATTTAAATCTGTCAAAGCAGCCaaaataatctCQCaaaat atccgagaatattttggaaagtctttgccagttgatctaacgreCAArCArrrreGGATTGCrCGrArACCAAGAACGGACAAr GTAGAATTTTTTGATCCCAATTACCGTTATTCTTTCAAAGAATGGCAAGATTGGTCTTTCAAACAAACAGATA ATAAGGGCTTTACTCGTTCAAGTCTAACGGTTTTAAGCGGTACAGAAGGCAAAAAACAAGTAGATGAACCCT GGTTTAATCTCTTATTGCACGAAACGAAATTTTCAGGAGAAAAGGGTTTAGTAGGGCGCAATAGCGTTATGT TTACCCTCTCTTTAGCCTACTTTAGTTCAGGCTATTCAATCGAAACGTGCGAATATAATATGTTTGAGTTTAA TAATCGATTAGATCAACCCTTAGAAGAAAAAGAAGTAATCAAAATTGTTAGAAGTGCCTATTCAGAAAACTA TCAAGGGGCTAATAGGGAATACATTACCATTCTTTGCAAAGCTTGGGTATCAAGTGATTTAACCAGTAAAGA TTTATTTGTCCGTCAAGGGTGGTTTAAATTCAAGAAAAAAAGAAGCGAACGTCAACGTGTTCATTTGTCAGA ATGGAAAGAAGATTTAATGGCTTATATTAGCGAAAAAAGCGATGTATACAAGCCTTATTTAGCGACGACCAA AAAAGAGATTAGAGAAGTGCTAGGCATTCCTGAACGGACATTAGATAAATTGCTGAAGGTACTGAAGGCGA ATCAGGAAATTTTCTTTAAGATTAAACCAGGAAGAAATGGTGGCATTCAACTTGCTAGTGTTAAATCATTGTT GCTATCGATCATTAAATTAAAAAAAGAAGAACGAGAAAGCTATATAAAGGCGCTGACAGCTTCGTTTAATTT AGAACGTACATTTATTCAAGAAACTCTAAACAAATTGGCAGAACGCCCCAAAACGGACCCACAACTCGATTT
GrrTAGCTACGATACAGGCTGAAAATAAAACCCGCACTATGCCATTACATTTATATCTATGATACGTGTTT GTTTTTCTTTGCTGGCTAGCTTAATTGCTTatatttacctgcaataaaggatttcttacttccattatactcccattttccaaaaacatacggggaacacgg gaacttattgtacaggccacctcatagttaatggtttcgagccttcctgcaatctcatccatggaaatatattcatccccctgccggcctattaatgtgacttttgtgcccggcggatatt cctgatccagctccaccataaattggtccatgcaaattcggccggcaattttcaggcgttttcccttcacaaggatgtcggtccctttcaattttcggagccagccgtccgcatagcct acaggcaccgtcccgatccatgtgtctttttccgctgtgtactcggctccgtagctgacgctctcgccttttctgatcagtttgacatgtgacagtgtcgaatgcagggtaaatgccgg acgcagctgaaacggtatctcgtccgacatgtcagcagacgggcgaaggccatacatgccgatgccgaatctgactgcattaaaaaagccttttttcagccggagtccagcggc gctgttcgcgcagtggaccattagattctttaacggcagcggagcaatcagctctttaaagcgctcaaactgcattaagaaatagcctctttctttttcatccgctgtcgcaaaatggg taaatacccctttgcactttaaacgagggttgcggtcaagaattgccatcacgttctgaacttcttcctctgtttttacaccaagtctgttcatccccgtatcgaccttcagatgaaaatga agagaaccttttttcgtgtggcgggctgcctcctgaagccattcaacagaataacctgttaaggtcacgtcatactcagcagcgattgccacatactccgggggaaccgcgccaa gcaccaatataggcgccttcaatccctttttgcgcagtgaaatcgcttcatccaaaatggccacggccaagcatgaagcacctgcgtcaagagcagcctttgctgtttctgcatcac catgcccgtaggcgtttgctttcacaactgccatcaagtggacatgttcaccgatatgttttttcatattgctgacattttcctttatcgcggacaagtcaatttccgcccacgtatctctgt aaaaaggttttgtgctcatggaaaactcctctcttttttcagaaaatcccagtacgtaattaagtatttgagaattaattttatattgattaatactaagtttacccagttttcacctaaaaaac aaatgatgagataatagctccaaaggctaaagaggactataccaactatttgttaATTAA (SEQ ID NO: 16). Sequence Legend key:
-Normal UPPERCASE sequences : hly promoter.
-Italicized UPPERCASE sequences: pl5 origin.
-Bolded UPPERCASE sequences: t-LLO ORF.
-Italicized and underlined UPPERCASE sequences: survivin.
[00395] Mice
[00396] LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-l-Cre (KPC) mice were generated as previously described (Hingorani SR et al. Cancer Cell 2005 7:469). Normal healthy wild-type littermates
72/ +
(LSL-Trp53 , Pdxl-Cre) used in experiments were generated during the routine breeding of KPC mice. C57BL/6 mice were ordered from Jackson Laboratories. All animal protocols were reviewed and approved by the Institute of Animal Care and Use Committee (IACUC) of the University of Pennsylvania and housed in pathogen free conditions at the University of Pennsylvania.
[00397] Subcutaneous tumor model
[00398] 6 week old female C57BL/6 mice were allowed to acclimate one week in the animal facility prior to tumor cell implantation. Mice were implanted subcutaneously (s.c.) with 200,000 cells obtained from a KPC-derived tumor cell line maintained at below passage 10. Tumors were longitudinally measured with calipers every 3-4 days. Tumor volumes were calculated as Volume (V)= ½ (L x S ) where L = longest dimension and S = shortest dimension. For overall survival studies, mice were euthanized when tumors reached 1000 mm . Mice were also euthanized if they became moribund or exhibited ulcerations larger than allowable in the IACUC approved animal protocol.
[00399] Collection of tissue samples from mice
[00400] Preparation of single cell suspension pancreatic tumors:
[00401] Tumor pieces were harvested from primary pancreatic tumors arising spontaneously in KPC mice. Tumors were minced to lxl mm pieces and incubated with collagenase (1 mg/mL) and DNAse I (75ug/mL,Roche #10104159001) for thirty minutes at 37°C with 5% C02. The digested tumor was homogenized and passed through a 70 μιη filter and washed with Dulbecco's Modified Eagle's medium (DMEM).
[00402] Preparation of single cell suspension from spleen: [00403] Spleens were excised and placed in DMEM supplemented with 10% fetal calf serum (FCS). Spleens were then gently homogenized to create a single cell suspension, which was passed through a 70μΜ cell strainer, followed by red blood cell lysis with ACK Lysis Buffer (Cambrex/BioWhittaker). Cells were washed with DMEM supplemented with 10% FCS.
[00404] Preparation of single cell suspension from peri-tumoral and mesenteric lymph nodes (MLN):
[00405] Peri-tumoral LN were defined as lymph nodes that were physically attached to pancreatic tumors in KPC mice. Peri-tumoral LN from individual mice were removed from tumors and pooled for homogenization by gentle mashing on a 70μΜ cell strainer. Mesenteric lymph nodes were excised and homogenized by gentle mashing on a 70μΜ cell strainer. Single cell suspensions were washed and resuspended in DMEM supplemented with 10% FCS.
[00406] Listeria infections and in vivo antibody administration
[00407] Listeria monocytogenes (Lm) stocks were kindly provided by Advaxis Inc. LmddA-265 (encoding human Survivin protein), ADXS 11-001 (encoding the HPV-E7 protein). For intraperitoneal injections, Lm Stocks were washed and resuspended in sterile PBS for prior to injection. Mice were injected i.p. with 1x10 8 CFU of Lm and 2x108 CFU of Lm for the second immunization. KPC mice received 1 Lm immunization of 1x10 CFU. For PD-1 blockade, 100μg of anti-mouse PD-1 (clone RMP1-14; BioXCell BE0146) or control 2A3 antibody (BioXCell BE0089) were diluted in sterile PBS and administered i.p.
[00408] Indirect Immunofluorescence microscopy
[00409] Tissues were flash frozen in Tissue-Tek O.C.T. Compound (V.W.R. 25608-930) and stored at -80°C. Frozen tissue sections were prepared by the University of Pennsylvania Cancer Histology Core. For PD-L1 and Her2 staining, tissue samples were thawed and immediately fixed with 3% buffered formaldehyde for 5 minutes at room temperature. Slides were then washed 2 times with PBST (PBS with 0.1% Tween-20) for 2 minutes each wash. Slides were then blocked with 10% goat serum in PBS for 45 minutes at room temperature, followed by a wash in PBST. Primary rat anti-mouse B7-H1 antibody (clone MIH5; eBiosciencel4-5982-81) was diluted at 1:50 in 10% goat serum + PBST. Primary antibody incubation was performed for 60 minutes at room temperature. For control "no primary" antibody staining, slides were incubated in 10% goat serum + TBST only. Slides were then washed prior to incubation with secondary goat anti-rat AF488 antibody (Life Technologies) for 30 minutes at room temperature at a dilution of 1:200 in 10% goat serum + PBST. Slides were washed with TBST. Slides were then counterstained with DAPI (4', 6-diamidino-2-phenylindole) solution at 0.2ug/mL for eight minutes in the dark. Slides were washed and mounted with VECTASHIELD Mounting Medium (Vector Laboratories). Images were collected on an Olympus 1X83 inverted microscope using CellSensDigital Imaging Software(Olympus). The same exposure times and acquisition settings were applied to all images to allow for comparison of fluorescence.
[00410] Immunohistochemistry
[00411] Tissues were flash frozen in Tissue-Tek O.C.T. Compound (V.W.R. 25608-930) and stored at -80C°. Frozen tissue sections were prepared by the University of Pennsylvania Cancer Histology Core and the University of Pennsylvania Molecular Pathology Imaging Core. Tissue samples were thawed and immediately fixed with either 3% buffered formaldehyde for 15 minutes at room temperature (for CD45 and mSurvivin staining) or with methanol at -20°C for 10 minutes (for CD3, CD4, CD8 and Foxp3 staining). Slides were then washed 2 times with PBST (PBS with 0.1% Tween-20) for 2 minutes each wash. Endogenous peroxidase activity was blocked by incubating slides in a solution of 0.3% hydrogen peroxide (diluted in distilled H20) for 10 minutes at room temperature. Slides were washed as described above. Slides were then blocked with 10% goat serum in PBS for 45 minutes at room temperature. The following primary antibodies were diluted in 10% goat serum in PBST:rat anti-mouse CD45 (clone 30-F11; BD 550539), rat anti-mouse CD3 antibody (clone KT3; Serotec/Bio-Rad MCA500GA) at a dilution of 1:50, rat anti-mouse CD4 antibody (clone GK1.5; BioXCell) at a dilution of 15ug/mL, rat anti-mouse CD8 antibody (clone 2.43; BioXCell) at a dilution of 15ug/mL, rat anti-mouse Foxp3 antibody (clone FJK-16s;eBioscience 14-5773-82) at a dilution of 1: 100, rat anti-mouse Survivin (CST 71G4B7). All primary incubations were performed overnight at 4°C. Slides were then washed two times in TBST for 2 minutes each. For control "no primary" antibody staining, slides were incubated in 10% goat serum + TBST only. Incubation with secondary biotinylated goat anti-rat antibody (polyclonal; BD 559286) was performed for 1 hour at room temperature at a dilution of 1:200 in 10% goat serum + PBST. Slides were washed 2 times for 2 minutes in PBS. VECTASTAIN ABC Reagent (Vector Laboratories PK-6100) was prepared according to manufacturer's instructions and applied to slides for 30 minutes at room temperature. Slides were washed 2 times in PBS. Detection was performed with DAB (3, 3 '-diaminobenzidine) HRP substrate that was prepared according to manufacturer instructions (Vector Laboratories SK- 4100). Slides were washed in distilled H20 and counterstained with Hematoxylin (Hematoxylin Stain-Harris; Sigma- Aldrich HHS 16) diluted 1: 1 in distilled H20 for forty seconds. Slides were rinsed with H20 and dehydrated by sequential washes of 95% ethanol, 100% ethanol and xylenes. After drying, slides were mounted using Cytoseal 60 (Fisher 23-244257). Images were acquired on an Olympus BX43 microscope using CellSens Digital Imaging Software (Olympus). Quantification was performed by averaging the number of positive events counted in 4 high powered fields (40X magnification).
[00412] Generation of tumor cell lines and cell culture
[00413] Tumor pieces were harvested from primary PDA tumors that spontaneously arose in KPC mice backcrossed onto the C57BL/6 genetic background. Tumor was minced to lxl mm pieces and incubated with collagenase (lmg/mL) for thirty minutes at 37°C with 5% C02. The digested tumor was homogenized and passed through a 70 μιη filter and washed with DMEM. Cells were spun at 1600 RPM for 5 minutes and washed 2 times with DMEM. Cells were resuspended in serum-free media and plated at a density of 1-3 xlO6 cells/well of a 6-well plate. After two weeks in culture, cells were cultured in DMEM supplemented with 5% fetal calf serum, 1% L-glutamine, and gentamycin. Cells were used for in vitro studies after at least 5 passages. For IFN-γ treatment of cell lines, cells were plated in 12 well plates 24 hours prior to IFN-γ addition. Mouse recombinant IFN-γ (R&D 485 Ml/CF) was added at 100 ng/niL and incubated for 12-72 hours.
[00414] Flow cytometry [00415] Antibodies and Flow cytometric analysis for cell surface antigens:
[00416] Flow cytometric analysis PDA tumor cell lines was performed by dissociating adherent cells with 0.05% Trypsin/EDTA (Gibco Life Technologies), followed by washes in DMEM and PBS. Single cell suspensions from tissue samples were prepared as describe above. lxlO6 cells were added to a round bottom 96-well plate for antibody incubations. Cells were first stained with Live/Dead Fixable Aqua Dead Cell Stain (Life Technologies L34957) according to manufacturer instructions. The following antibodies were used for staining of surface antigens: MHC1 FITC (H-2Db clone KH95; BD 553573) at 1:400, MHCII PE (I-A/I-E clone M5/115.15.2; BD 557000) at 1:200, PD-L1 PE(clone MIH5; BD 558091) at 1: 100, CD3 DPerCP/Cy5.5 (clone 145-2C11; eBioscience 561008) at 1: 150, CD4 PE/Cy7 (clone GK1.5; eBioscience 25-0041-82), CD44 APC e-Fluor 780 (clone IM7; eBioscience 47-0441-80) at 1:200, PD1 PE/Cy7 (clone RMPl-30; Biolegend 109110) at 1: 100, CD8b PE (clone H35-17.2; eBio 53-0083-82)at 1: 150 or CD8 D Pacific Blue (clone 53-6.7; eBio 48-0081082) at 1: 100, CD86 (clone GL1; BD 553692; 1:200), CD40 (clone 30-F11; BD 553791; 1:200), CD45 APC (clone 30-F11; BD 559864) at 1:800 or CD45 PE (clone 30-Fl l;BD 553081) at 1:800. All antibodies were diluted in FACS buffer (PBS + 2% FCS + 2mM EDTA) and added to cells for 20 minutes at room temperature. Cells were resuspended in FACS buffer for analysis on a BD FACSCanto. Data was analyzed using FloJo software (Tree Star Inc.).
[00417] Intracelluar Survivin staining:
[00418] Cells were first stained with Live/Dead Fixable Aqua Dead Cell Stain prior to fixation/permeabilization. The Foxp3 / Transcription Factor Staining Buffer Set (eBioscience 00- 5523-00) was used for intracellular Survivin staining. Fixation and permeabilization was then performed according to manufacture instructions for lhour at room temperature and in the dark. Primary rat anti-mouse Survivin (CST 71G4B7) was diluted at 1:200 in permeabilization buffer and incubated with cells for 20 minutes at room temperature. No primary antibody was added to "no primary" control samples. Cells were washed 2 times with permeabilization buffer. Secondary biotinylated goat anti-rat antibody (polyclonal; BD 559286) was diluted at 1:200 in permeabilization buffer and incubation was performed for 20 minutes at room temperature. Cells were washed 2 times in permeabilization buffer. Streptavidin PE (BD 554061), diluted 1:300 in permeabilization buffer, was added to cells for 20 minutes at room temperature. Cells were washed 2 times in permeabilization buffer, followed by wash in FACS buffer. Cells were resuspended in FACS buffer for analysis on a BD FACSCanto. Data was analyzed using FloJo software (Tree Star Inc.). [00419] Statistical analysis
[00420] Statistical analysis was performed with Prism Software (GraphPad). Subcutaneous tumor growth curves were calculated by taking the average volume of tumors for each group of mice at each time point. Unless otherwise noted, p values were calculated using the Student's t- test and the F-test was used to determine equality of variance. Survival curves were compared using the log-rank (Mantel-Cox) test.
RESULTS
EXAMPLE 1: HER2/NEU EXPRESSION IN KPC MICE AND PDA CELL LINES
[00421] High levels of expression of Her 2/Neu was observed both in spontaneous tumors arising in KPC mice and in PDA cell lines implanted subcutaneously with anti-CD40 treatment inducing T cells specific for Her2-specific peptides (Figs. 1A-D).
EXAMPLE 2: LM-LLO HER2/NEU TREATMENT OF PDA TUMORS IMPLANTED IN
MICE
[00422] 7861. PDA cells were implanted into mice. The resulting tumors were treated with Lm-LLO and Lm-LLO-cHER2 and the tumor size was observed over 20 day period. The treatment had no noticeable effect on tumor size (Figs. 2B and C).
EXAMPLE 3: PROPHYLACTIC LM-LLO HER2/NEU AND LM-LLO- CHER2
TREATMENT OF PDA TUMORS IMPLANTED IN MICE
[00423] Mice were treated with Lm-LLO and Lm-LLO-cHER2 prior to PDA tumor implantation (Fig. 3A) and the growth of subsequently implanted PDA tumors was assessed. The preventive treatment has no observable effect on growth of 7861. PDA cell line-derived tumors introduced in mice (Fig. 3B).
EXAMPLE 4: LM-LLO SURVIVIN AND LM-LLO-E7 TREATMENT OF PDA TUMORS
STIMULATES T CELL INFILTRATION [00424] PDA tumor cells were examined for expression of survivin and found to express high levels of survivin (Fig. 4A). The effect of, Lm-LLO-Survivin treatment on tumor growth and T cell infiltration was examined. Surprisingly, the treatment had no effect on growth of 69.PDA cell line- derived tumors established in mice (Fig. 4B) despite clear increase of T-cell infiltration into these tumors (Figs. 4C and D).
EXAMPLE 5: PD-Ll/PD-1 SIGNALING IS LIKELY ACTIVE IN KPC MOUSE TUMORS
[00425] The expression of the components of PD-Ll/PD-1 signaling pathway was investigated. While mouse tumors express high levels of PD-Ll (Fig. 5B), the T cells isolated from mesenteric versus peritumoral lymph nodes show high level of PD-1 expression (Fig. 5C).
EXAMPLE 6: INTERFERON-GAMMA TREATMENT OF TUMOR CELLS INDUCES
MHC AND PD-L-1 EXPRESSION
[00426] The effects of IFN-gamma treatment on MHC and PD-Ll expression in tumor cells was monitored. The treatment was observed to strongly stimulate expression of both molecules (Figs. 6A-D).
EXAMPLE 7: COMBINATION LM-LLO-SURVIVIN-ANTI PD1 TREATMENT OF PDA
TUMORS AND MICE
[00427] The effect of combination treatment involving Lm-LLO-Survivin-anti PD1 antibody was monitored in Mice. Mice were pretreated with the combination therapy as outlined in (Fig. 7A). A significant decrease in tumor growth rate (Figs. 7B and C) and prolonged survival were observed (Fig. 7D).
[00428] While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.

Claims

What is claimed is:
1. A method of eliciting an anti-tumor or anti-cancer immune response against a tumor or cancer in a subject, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist, thereby eliciting an anti-tumor or anti-cancer immune response in said subject.
2. The method of claim 1, wherein said immunosuppressive molecule antagonist is an antibody or functional fragment thereof.
3. The method of claim 2, wherein said antibody or fragment thereof comprises a polyclonal antibody, a monoclonal antibody, an Fab fragment, an F(ab')2 fragment, an Fv fragment, a single chain antibody (SCA), or any combination thereof.
4. The method of any one of claims 1-3, wherein said immunosuppressive molecule comprises a member of the CD28/CTLA-4 receptor superfamily, or a member of B7 receptor family.
5. The method of claim 4, wherein said member of the CD28/CTLA-4 receptor superfamily is PD-1.
6. The method of claim 4, wherein said member of B7 receptor family is selected from the group consisting of B7-1, PD-L1, and PD-L2.
7. A method of any of the claims 1-6, wherein said recombinant Listeria strain comprises a nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide, wherein the said fusion polypeptide comprises a PEST sequence-containing polypeptide fused to survivin antigen or immunogenic fragment thereof.
8. The method of any one of claims 1-7, wherein said survivin antigen is encoded by a nucleic acid sequence as set forth in SEQ ID NO: 17.
9. The method of any of the claims 1-8 wherein said survivin antigen comprises the amino acid sequence as set forth in SEQ ID NO: 19.
10. The method of any one of claims 1-9, wherein said nucleic acid molecule comprises a second open reading frame encoding a metabolic enzyme, and wherein said metabolic enzyme complements a mutation in the chromosome of said recombinant Listeria strain.
11. The method of claim 10, wherein said metabolic enzyme is an alanine racemase enzyme or a D-amino acid transferase enzyme.
12. The method of any one of claim 1-11, wherein said nucleic acid molecule is integrated into the Listeria genome.
13. The method of any one of claims 1-12, wherein said nucleic acid molecule is in a plasmid in said recombinant Listeria strain, and wherein said plasmid is stably maintained in said recombinant Listeria strain in the absence of antibiotic selection.
14. The method of any one of claims 1-13, wherein said recombinant Listeria comprises a mutation in the act A virulence gene.
15. The method of any one of claims 1-14, wherein said PEST-containing polypeptide is selected from the group consisting of an N-terminal Listeriolysin O (LLO) fragment, an N-terminal ActA fragment and a PEST-containing peptide.
16. The method of any one of claims 1-15, wherein said recombinant attenuated Listeria is a Listeria monocytogenes.
17. The method of any one of claims 1-16, wherein said recombinant attenuated Listeria is ADXS31-265.
18. The method of any one of claims 1-17, wherein said subject is a human.
19. The method of any one of claims 1-18, further comprising administration of an independent adjuvant.
20. The method of claim 19 wherein said adjuvant comprises a granulocyte/macrophage colony- stimulating factor (GM-CSF) protein, a nucleotide molecule encoding a GM-CSF protein, saponin QS21, monophosphoryl lipid A, or an unmethylated CpG-containing oligonucleotide.
21. The method of any one of claims 1-20, wherein said tumor or cancer comprises a pancreatic tumor or cancer.
22. The method of claim 21, wherein said cancer is a malignant or recurrent cancer.
23. The method of claim 22, wherein said malignant cancer is pancreatic ductal carcinoma.
24. The method of claim 23, wherein said cancer is acinar cell carcinoma of the pancreas, or cystadenocarcinoma.
25. The method of claim 21, wherein said cancer is pancreatic neuroendocrine tumor.
26. The method of claim 21, wherein said cancer is insulinoma or gastrinoma.
27. The method of any one of claims 1-26, wherein said immunosuppressive molecule antagonist is administered prior to, concurrent with or following the administration of said recombinant attenuated Listeria strain.
28. A method of any one of claims 1-27, wherein said administration comprises at least two administrations of said combination therapy.
29. The method of any one of claims 1-28, further comprising a step of administering said combination therapy following a relapse or metastasis of said cancer in said subject.
30. The method of any one of claims 1-29, further comprising administering a booster of either component or both components of said combination therapy.
31. A method of preventing or treating a tumor or cancer in a subject, said method comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration prevents or treats a survivin-expressing tumor or cancer in said subject.
32. The method of claim 31, wherein said immunosuppressive molecule antagonist is an antibody or functional fragment thereof.
33. The method of claim 32, wherein said antibody or fragment thereof comprises a polyclonal antibody, a monoclonal antibody, an Fab fragment, an F(ab')2 fragment, an Fv fragment, a single chain antibody (SCA), or any combination thereof.
34. The method of any one of claims 31-33, wherein said immunosuppressive antagonist molecule comprises a member of the CD28/CTLA-4 receptor superfamily, or a member of B7 receptor family.
35. The method of claim 34, wherein said member of the CD28/CTLA-4 receptor superfamily is PD-1.
36. The method of claim 34, wherein said member of B7 receptor family is selected from the group consisting of B7-1, PD-L1, and PD-L2.
37. A method of any of the claims 31-36, wherein said recombinant Listeria strain comprises a nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide, wherein the said fusion polypeptide comprises a PEST sequence-containing polypeptide fused to survivin antigen to immunogenic fragment thereof.
38. The method of any one of claims 31-37, wherein said survivin antigen is encoded by a nucleic acid sequence as set forth in SEQ ID NO: 17.
39. The method of any of the claims 31-38 wherein said survivin antigen comprises the amino acid sequence as set forth in SEQ ID NO: 19.
40. The method of any one of claims 31-39, wherein said nucleic acid molecule comprises a second open reading frame encoding a metabolic enzyme, and wherein said metabolic enzyme complements a mutation in the chromosome of said recombinant Listeria strain.
41. The method of claim 40, wherein said metabolic enzyme is an alanine racemase enzyme or a D-amino acid transferase enzyme.
42. The method of any one of claim 31-41, wherein said nucleic acid molecule is integrated into the Listeria genome.
43. The method of any one of claims 31-42, wherein said nucleic acid molecule is in a plasmid in said recombinant Listeria strain, and wherein said plasmid is stably maintained in said recombinant Listeria strain in the absence of antibiotic selection.
44. The method of any one of claims 31-43, wherein said recombinant Listeria comprises a mutation in the act A virulence gene.
45. The method of any one of claims 31-44, wherein said PEST-containing polypeptide is selected from the group consisting of an N-terminal Listeriolysin O (LLO) fragment, an N- terminal ActA fragment and a PEST-containing amino acid sequence.
46. The method of any one of claims 31-45, wherein said recombinant attenuated Listeria is a Listeria monocytogenes.
47. The method of any one of claims 31-46, wherein said recombinant attenuated Listeria is ADXS31-265.
48. The method of any one of claims 31-47, wherein said subject is a human.
49. The method of any one of claims 31-48, further comprising administration of an independent adjuvant.
50. The method of claim 49, wherein said adjuvant comprises a granulocyte/macrophage colony- stimulating factor (GM-CSF) protein, a nucleotide molecule encoding a GM-CSF protein, saponin QS21, monophosphoryl lipid A, or an unmethylated CpG-containing oligonucleotide.
51. The method of any one of claims 31-50, wherein said tumor or cancer comprises a pancreatic tumor or cancer.
52. The method of claim 51, wherein said cancer is a malignant or recurrent cancer.
53. The method of claim 52, wherein said malignant cancer is pancreatic ductal carcinoma.
54. The method of claim 53, wherein said cancer is acinar cell carcinoma of the pancreas, or cystadenocarcinoma.
55. The method of claim 54, wherein said cancer is pancreatic neuroendocrine tumor.
56. The method of claim 55, wherein said cancer is insulinoma or gastrinoma.
57. The method of any one of claims 31-56, wherein said immunosuppressive antagonist molecule is administered prior to, concurrent with or following the administration of said composition comprising said recombinant attenuated Listeria strain.
58. A method of any of the claims 31-57 wherein said administration comprises at least two administrations of said combination therapy.
59. The method of any one of claims 31-58, further comprising a step of administering said combination therapy following a relapse or metastasis of said cancer in said subject.
60. The method of any one of claims 31-59, further comprising administering a booster of either component or both components of said combination therapy.
61. The method of any one of claims 31-60, wherein said treating results in progression free survival.
62. The method of any one of claims 1-61, wherein said method comprises breaking tolerance to survivin in a subject having said tumor or cancer.
63. The method of any one of claims 1-62, wherein said method comprises delaying the onset of metastases in said subject.
64. The method of any one of claims 1-63, wherein said method comprises prolonging survival of a subject having said tumor or cancer.
65. A combination therapy for use in eliciting an anti-tumor or anti-cancer immune response against a tumor or cancer in a subject, said combination therapy comprising the step of administering to said subject an effective amount of a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist, thereby eliciting an anti-tumor or anti-cancer immune response in said subject.
66. The use of claim 65, wherein said immunosuppressive molecule antagonist is an antibody or functional fragment thereof.
67. The use of claim 66, wherein said antibody or fragment thereof comprises a polyclonal antibody, a monoclonal antibody, an Fab fragment, an F(ab')2 fragment, an Fv fragment, a single chain antibody (SCA), or any combination thereof.
Ill
68. The use of any one of claims 65-67, wherein said immunosuppressive molecule comprises a member of the CD28/CTLA-4 receptor superfamily, or a member of B7 receptor family.
69. The use of claim 68, wherein said member of the CD28/CTLA-4 receptor superfamily is PD- 1.
70. The use of claim 69, wherein said member of B7 receptor family is selected from the group consisting of B7-1, PD-L1, and PD-L2
71. The use of any of the claims 65-70, wherein said recombinant Listeria strain comprises a nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide, wherein the said fusion polypeptide comprises a PEST sequence-containing polypeptide fused to survivin antigen or immunogenic fragment thereof.
72. The use of any one of claims 65-71, wherein said survivin antigen is encoded by a nucleic acid sequence as set forth in SEQ ID NO: 17.
73. The use of any of the claims 65-72, wherein said survivin antigen comprises the amino acid sequence as set forth in SEQ ID NO: 19.
74. The use of any one of claims 65-73, wherein said nucleic acid molecule comprises a second open reading frame encoding a metabolic enzyme, and wherein said metabolic enzyme complements a mutation in the chromosome of said recombinant Listeria strain.
75. The use of claim 74, wherein said metabolic enzyme is an alanine racemase enzyme or a D- amino acid transferase enzyme.
76. The use of any one of claim 65-75, wherein said nucleic acid molecule is integrated into the Listeria genome.
77. The use of any one of claims 65-76, wherein said nucleic acid molecule is in a plasmid in said recombinant Listeria strain, and wherein said plasmid is stably maintained in said recombinant Listeria strain in the absence of antibiotic selection.
78. The use of any one of claims 65-77, wherein said recombinant Listeria comprises a mutation in the act A virulence gene.
79. The use of any one of claims 65-78, wherein said PEST-containing polypeptide is selected from the group consisting of an N-terminal Listeriolysin O (LLO) fragment, an N-terminal ActA fragment and a PEST-containing peptide.
80. The use of any one of claims 65-79, wherein said recombinant attenuated Listeria is a Listeria monocytogenes.
81. The use of any one of claims 65-80, wherein said recombinant attenuated Listeria is ADXS31-265.
82. The use of any one of claims 65-81, wherein said subject is a human.
83. The use of any one of claims 65-82, further comprising administration of an independent adjuvant.
84. The use of claim 20 wherein said adjuvant comprises a granulocyte/macrophage colony- stimulating factor (GM-CSF) protein, a nucleotide molecule encoding a GM-CSF protein, saponin QS21, monophosphoryl lipid A, or an unmethylated CpG-containing oligonucleotide.
85. The use of any one of claims 65-84, wherein said tumor or cancer comprises a pancreatic tumor or cancer.
86. The use of claim 85, wherein said cancer is a malignant or recurrent cancer.
87. The use of claim 86, wherein said malignant cancer is pancreatic ductal carcinoma.
88. The use of claim 87, wherein said cancer is acinar cell carcinoma of the pancreas, or cystadenocarcinoma.
89. The use of claim 88, wherein said cancer is pancreatic neuroendocrine tumor.
90. The use of claim 89, wherein said cancer is insulinoma or gastrinoma.
91. The use of any one of claims 65-90, wherein said immunosuppressive molecule antagonist is administered prior to, concurrent with or following the administration of said recombinant attenuated Listeria strain.
92. Use of any one of claims 65-91, wherein said administration comprises at least two administrations of said combination therapy.
93. The use of any one of claims 65-92, further comprising a step of administering said combination therapy following a relapse or metastasis of said cancer in said subject.
94. The use of any one of claims 65-93, further comprising administering a booster of either component or both components of said combination therapy.
95. Use of a combination therapy for preventing or treating a tumor or cancer in a subject, said use comprising the step of administering to said subject an effective amount of a combination therapy comprising a recombinant Listeria strain expressing a survivin antigen and an immunosuppressive molecule antagonist wherein said administration prevents or treats a survivin-expressing tumor or cancer in said subject.
96. The use of claim 95, wherein said immunosuppressive molecule antagonist is an antibody or functional fragment thereof.
97. The use of claim 96, wherein said antibody or fragment thereof comprises a polyclonal antibody, a monoclonal antibody, an Fab fragment, an F(ab')2 fragment, an Fv fragment, a single chain antibody (SCA), or any combination thereof.
98. The use of any one of claims 95-97, wherein said immunosuppressive antagonist molecule comprises a member of the CD28/CTLA-4 receptor superfamily, or a member of B7 receptor family.
99. The use of claim 98, wherein said member of the CD28/CTLA-4 receptor superfamily is PD- 1.
100. The use of claim 99, wherein said member of B7 receptor family is selected from the group consisting of B7-1, PD-L1, and PD-L2
101. Use of any of the claims 95-100, wherein said recombinant Listeria strain comprises a nucleic acid molecule comprising a first open reading frame encoding a fusion polypeptide, wherein the said fusion polypeptide comprises a PEST sequence-containing polypeptide fused to survivin antigen to immunogenic fragment thereof.
102. The use of any one of claims 95-101, wherein said survivin antigen is encoded by a nucleic acid sequence as set forth in SEQ ID NO: 17.
103. The use of any of the claims 95-102, wherein said survivin antigen comprises the amino acid sequence as set forth in SEQ ID NO: 19.
104. The use of any one of claims 95-103, wherein said nucleic acid molecule comprises a second open reading frame encoding a metabolic enzyme, and wherein said metabolic enzyme complements a mutation in the chromosome of said recombinant Listeria strain.
105. The use of claim 104, wherein said metabolic enzyme is an alanine racemase enzyme or a D-amino acid transferase enzyme.
106. The use of any one of claim 95-105, wherein said nucleic acid molecule is integrated into the Listeria genome.
107. The use of any one of claims 95-106, wherein said nucleic acid molecule is in a plasmid in said recombinant Listeria strain, and wherein said plasmid is stably maintained in said recombinant Listeria strain in the absence of antibiotic selection.
108. The use of any one of claims 95-107, wherein said recombinant Listeria comprises a mutation in the act A virulence gene.
109. The use of any one of claims 95-108, wherein said PEST-containing polypeptide is selected from the group consisting of an N-terminal Listeriolysin O (LLO) fragment, an N- terminal ActA fragment and a PEST-containing amino acid sequence.
110. The use of any one of claims 95-109, wherein said recombinant attenuated Listeria is a Listeria monocytogenes.
111. The use of any one of claims 95-110, wherein said recombinant attenuated Listeria is ADXS31-265.
112. The use of any one of claims 95-111, wherein said subject is a human.
113. The use of any one of claims 95-112, further comprising administration of an independent adjuvant.
114. The use of claim 113, wherein said adjuvant comprises a granulocyte/macrophage colony- stimulating factor (GM-CSF) protein, a nucleotide molecule encoding a GM-CSF protein, saponin QS21, monophosphoryl lipid A, or an unmethylated CpG-containing oligonucleotide.
115. The use of any one of claims 95-114, wherein said tumor or cancer comprises a pancreatic tumor or cancer.
116. The use of claim 115, wherein said cancer is a malignant or recurrent cancer.
117. The use of claim 116, wherein said malignant cancer is pancreatic ductal carcinoma.
118. The use of claim 117, wherein said cancer is acinar cell carcinoma of the pancreas, or cystadenocarcinoma.
119. The use of claim 118, wherein said cancer is pancreatic neuroendocrine tumor.
120. The use of claim 119, wherein said cancer is insulinoma or gastrinoma.
121. The use of any one of claims 95-120, wherein said immunosuppressive antagonist molecule is administered prior to, concurrent with or following the administration of said composition comprising said recombinant attenuated Listeria strain.
122. A use of any of the claims 95-121, wherein said administration comprises at least two administrations of said combination therapy.
123. The use of any one of claims 95-122, further comprising a step of administering said combination therapy following a relapse or metastasis of said cancer in said subject.
124. The use of any one of claims 95-123, further comprising administering a booster of either component or both components of said combination therapy.
125. The use of any one of claims 95-124, wherein said treating results in progression free survival.
126. The use of any one of claims 95-125, wherein said use comprises breaking tolerance to survivin in a subject having said tumor or cancer.
127. The use of any one of claims 95-126, wherein said use comprises delaying the onset of metastases in said subject.
128. The use of any one of claims 95-127, wherein said use comprises prolonging survival of a subject having said tumor or cancer.
PCT/US2016/023969 2015-03-26 2016-03-24 Combination of a pd-1 antagonist and a listeria based vaccine for treating pancreatic cancer WO2016154412A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562138644P 2015-03-26 2015-03-26
US62/138,644 2015-03-26

Publications (2)

Publication Number Publication Date
WO2016154412A2 true WO2016154412A2 (en) 2016-09-29
WO2016154412A9 WO2016154412A9 (en) 2016-11-17

Family

ID=56978635

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/023969 WO2016154412A2 (en) 2015-03-26 2016-03-24 Combination of a pd-1 antagonist and a listeria based vaccine for treating pancreatic cancer

Country Status (2)

Country Link
TW (1) TW201705968A (en)
WO (1) WO2016154412A2 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10055540B2 (en) 2015-12-16 2018-08-21 Gritstone Oncology, Inc. Neoantigen identification, manufacture, and use
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
US10336824B2 (en) 2015-03-13 2019-07-02 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of thereof
US10513558B2 (en) 2015-07-13 2019-12-24 Cytomx Therapeutics, Inc. Anti-PD1 antibodies, activatable anti-PD1 antibodies, and methods of use thereof
US10900044B2 (en) 2015-03-03 2021-01-26 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, 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
US11264117B2 (en) 2017-10-10 2022-03-01 Gritstone Bio, Inc. Neoantigen identification using hotspots
US11446369B2 (en) 2007-05-10 2022-09-20 Advaxis, Inc. Compositions and methods comprising KLK3 or FOLH1 antigen
US11885815B2 (en) 2017-11-22 2024-01-30 Gritstone Bio, Inc. Reducing junction epitope presentation for neoantigens
US11897927B2 (en) 2016-11-30 2024-02-13 Advaxis, Inc. Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11446369B2 (en) 2007-05-10 2022-09-20 Advaxis, Inc. Compositions and methods comprising KLK3 or FOLH1 antigen
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
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
US10336824B2 (en) 2015-03-13 2019-07-02 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of thereof
US10669339B2 (en) 2015-03-13 2020-06-02 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of use thereof
US11174316B2 (en) 2015-03-13 2021-11-16 Cytomx Therapeutics, Inc. Anti-PDL1 antibodies, activatable anti-PDL1 antibodies, and methods of use thereof
US10513558B2 (en) 2015-07-13 2019-12-24 Cytomx Therapeutics, Inc. Anti-PD1 antibodies, activatable anti-PD1 antibodies, and methods of use thereof
US10847252B2 (en) 2015-12-16 2020-11-24 Gritstone Oncology, Inc. Neoantigen identification, manufacture, and use
US10847253B2 (en) 2015-12-16 2020-11-24 Gritstone Oncology, Inc. Neoantigen identification, manufacture, and use
US11183286B2 (en) 2015-12-16 2021-11-23 Gritstone Bio, Inc. Neoantigen identification, manufacture, and use
US10055540B2 (en) 2015-12-16 2018-08-21 Gritstone Oncology, Inc. Neoantigen identification, manufacture, and use
US11897927B2 (en) 2016-11-30 2024-02-13 Advaxis, Inc. Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, 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
US11264117B2 (en) 2017-10-10 2022-03-01 Gritstone Bio, Inc. Neoantigen identification using hotspots
US11885815B2 (en) 2017-11-22 2024-01-30 Gritstone Bio, Inc. Reducing junction epitope presentation for neoantigens

Also Published As

Publication number Publication date
TW201705968A (en) 2017-02-16
WO2016154412A9 (en) 2016-11-17

Similar Documents

Publication Publication Date Title
AU2015289533B2 (en) Combination of a PD-1 antagonist and a Listeria-based vaccine for treating prostate cancer
WO2016154412A2 (en) Combination of a pd-1 antagonist and a listeria based vaccine for treating pancreatic cancer
US20210069326A1 (en) Pd-l1 antagonist combination treatments
US20220324979A1 (en) Combination of a pd-1 antagonist and a vegfr inhibitor for treating cancer
US10695426B2 (en) Combination of a PD-1 antagonist and an ALK inhibitor for treating cancer
EP3102604B1 (en) Combination of a pd-1 antagonist and a 4-1bb agonist for treating cancer
US10023649B2 (en) Method of treating cancer with a combination of an anti-CCR4 antibody and a 4-1BB agonist
CN105451770B (en) Treatment of cancer using a combination of a PD-1 antagonist and dinaciclib
US20180104284A1 (en) Immunogenic Listeria-Based Compositions Comprising Truncated Acta-Antigen Fusions And Methods Of Use Thereof
CN106573977B (en) Humanized antibodies against CEACAM1
KR20190031492A (en) NANT CANCER VACCINE
CA2955432A1 (en) Listeria-based immunogenic compositions for eliciting anti-tumor responses
KR20170066546A (en) Combination therapies
CN112312970A (en) Methods of treating cancer with bispecific anti-CD 3xMUC16 antibodies and anti-PD-1 antibodies
WO2017147368A1 (en) Redirecting immune responses
KR20210006405A (en) Inhibition of the combination of PD-1/PD-L1, TGFβ and DNA-PK for the treatment of cancer
US20200061167A1 (en) Combination of listeria-based vaccine with anti-ctla-4 or anti-cd137 antibodies
JP2022543710A (en) Combination therapy with antibodies against claudin 18.2 and immune checkpoint inhibitors to treat cancer
CN113939309A (en) Treatment of cancer with sEphB4-HSA fusion protein
CN113677707A (en) IL-4/IL-13 pathway inhibitors for enhanced efficacy in the treatment of cancer
NZ737018B2 (en) Pd-l1 antagonist combination treatments

Legal Events

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

Ref document number: 16769672

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16769672

Country of ref document: EP

Kind code of ref document: A2