WO2018129306A1 - Souches vaccinales de listeria recombinées et leurs méthodes d'utilisation en immunothérapie anticancéreuse - Google Patents

Souches vaccinales de listeria recombinées et leurs méthodes d'utilisation en immunothérapie anticancéreuse Download PDF

Info

Publication number
WO2018129306A1
WO2018129306A1 PCT/US2018/012570 US2018012570W WO2018129306A1 WO 2018129306 A1 WO2018129306 A1 WO 2018129306A1 US 2018012570 W US2018012570 W US 2018012570W WO 2018129306 A1 WO2018129306 A1 WO 2018129306A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigenic peptide
listeria strain
recombinant listeria
seq
hpv16
Prior art date
Application number
PCT/US2018/012570
Other languages
English (en)
Inventor
Robert Petit
Original Assignee
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 Advaxis, Inc. filed Critical Advaxis, Inc.
Publication of WO2018129306A1 publication Critical patent/WO2018129306A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/522Bacterial cells; Fungal cells; Protozoal cells avirulent or attenuated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6068Other bacterial proteins, e.g. OMP
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/86Lung
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • HPV-associate cancers include cervical cancer, anal cancer, head and neck cancer, or an oropharyngeal cancer.
  • Cervical cancer is the most common HPV-associated cancer among women, and oropharyngeal cancers (cancers of the back of the throat, including the base of the tongue and tonsils) are the most common among men.
  • E6 and E7 proteins from high-risk type HPV 16 and HPV 18 are oncoproteins that act by stimulating the destruction of many host cell key regulatory proteins.
  • HPV16 E6 associates with host E6-AP ubiquitin-protein ligase, and inactivates tumor suppressors TP53 and TP73 by targeting them to the 26S proteasome for degradation and this increases DNA damage and chromosomal instabilities and leads to cell proliferation and cancer development.
  • Standard chemoradiation regimens often used in advanced stage cancers can be associated with significant toxicity.
  • immunotherapy has the potential to reduce toxicity through de-escalation of chemoradiation regimens, and potentially enhance long- term disease control.
  • recombinant Listeria strains comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST-containing peptide fused to an HPV 16 antigenic peptide and an HPV 18 antigenic peptide, wherein the HPV 16 antigenic peptide and the HPV 18 antigenic peptide are operably linked in tandem.
  • the HPV 16 antigenic peptide in an HPV 16 E6 antigenic peptide or an HPV 16 E7 antigenic peptide is an HPV 18 E6 antigenic peptide or an HPV 18 E7 antigenic peptide.
  • the HPV 16 antigenic peptide is an HPV 16 E7 antigenic peptide and the HPV 18 antigenic peptide is an HPV 18 E7 antigenic peptide.
  • such fusion polypeptides and nucleic acids encoding such fusion polypeptides are also provided herein are immunogenic compositions,
  • compositions, or vaccines comprising a recombinant Listeria strain comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST-containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.
  • the HPV16 antigenic peptide in an HPV16 E6 antigenic peptide or an HPV16 E7 antigenic peptide is an HPV18 E6 antigenic peptide or an HPV18 E7 antigenic peptide.
  • the HPV16 antigenic peptide is an HPV16 E7 antigenic peptide and the HPV18 antigenic peptide is an HPV18 E7 antigenic peptide.
  • immunogenic compositions, pharmaceutical compositions, or vaccines comprising the fusion polypeptide or a nucleic acid encoding the fusion polypeptide.
  • kits for inducing an immune response against a tumor or cancer in a subject comprising administering to the subject a recombinant Listeria strain comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST- containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.
  • the HPV16 antigenic peptide in an HPV16 E6 antigenic peptide or an HPV16 E7 antigenic peptide is an HPV18 E6 antigenic peptide or an HPV18 E7 antigenic peptide.
  • the HPV16 antigenic peptide is an HPV16 E7 antigenic peptide and the HPV18 antigenic peptide is an HPV18 E7 antigenic peptide.
  • methods of inducing an immune response against a tumor or cancer in a subject comprising administering to the subject an immunogenic composition, a pharmaceutical composition, or a vaccine comprising such a recombinant Listeria strain.
  • Also provided are methods of inducing an immune response against a tumor or cancer in a subject comprising administering to the subject the fusion polypeptide or a nucleic acid encoding the fusion polypeptide, an immunogenic composition comprising the fusion polypeptide or the nucleic acid encoding the fusion polypeptide, a pharmaceutical composition comprising the fusion polypeptide or the nucleic acid encoding the fusion polypeptide, or a vaccine comprising the fusion polypeptide or the nucleic acid encoding the fusion polypeptide.
  • kits for preventing or treating a tumor or cancer in a subject comprising administering to the subject a recombinant Listeria strain comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST-containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.
  • the HPV16 antigenic peptide in an HPV16 E6 antigenic peptide or an HPV16 E7 antigenic peptide is an HPV18 E6 antigenic peptide or an HPV18 E7 antigenic peptide.
  • the HPV16 antigenic peptide is an HPV16 E7 antigenic peptide and the HPV18 antigenic peptide is an HPV18 E7 antigenic peptide. Also provided are methods of preventing or treating a tumor or cancer in a subject, comprising administering to the subject an immunogenic composition, a pharmaceutical composition, or a vaccine comprising such a recombinant Listeria strain.
  • Also provided are methods of preventing or treating a tumor or cancer in a subject comprising administering to the subject the fusion polypeptide, a nucleic acid encoding the fusion polypeptide, an immunogenic composition comprising the fusion polypeptide or the nucleic acid encoding the fusion polypeptide, a pharmaceutical composition comprising the fusion polypeptide or the nucleic acid encoding the fusion polypeptide, or a vaccine comprising the fusion polypeptide or the nucleic acid encoding the fusion polypeptide.
  • cell banks comprising one or more recombinant Listeria strains comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST- containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.
  • the HPV16 antigenic peptide in an HPV16 E6 antigenic peptide or an HPV16 E7 antigenic peptide is an HPV18 E6 antigenic peptide or an HPV18 E7 antigenic peptide.
  • the HPV16 antigenic peptide is an HPV16 E7 antigenic peptide and the HPV18 antigenic peptide is an HPV18 E7 antigenic peptide.
  • Figure 1 shows the study design for Example 3.
  • Figure 2 shows results for tumor volume. Mice treated with ADXS-DUAL (ADXS-602) show a significant decrease in tumor volume.
  • Figure 3 shows results for percent survival. Mice treated with ADXS-DUAL (ADXS-602) show a significant increase in percent survival.
  • protein refers to polymeric forms of amino acids of any length, including coded and non- coded amino acids and chemically or biochemically modified or derivatized amino acids.
  • the terms include polymers that have been modified, such as polypeptides having modified peptide backbones.
  • Proteins are said to have an "N-terminus” and a "C-terminus.”
  • N- terminus relates to the start of a protein or polypeptide, terminated by an amino acid with a free amine group (-NH2).
  • C-terminus relates to the end of an amino acid chain (protein or polypeptide), terminated by a free carboxyl group (-COOH).
  • fusion protein refers to a protein comprising two or more peptides linked together by peptide bonds or other chemical bonds.
  • the peptides can be linked together directly by a peptide or other chemical bond.
  • a chimeric molecule can be recombinantly expressed as a single-chain fusion protein.
  • the peptides can be linked together by a "linker” such as one or more amino acids or another suitable linker between the two or more peptides.
  • nucleic acid and “polynucleotide,” used interchangeably herein, refer to polymeric forms of nucleotides of any length, including ribonucleotides, deoxyribonucleotides, or analogs or modified versions thereof. They include single-, double-, and multi- stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, and polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases.
  • mononucleotides are reacted to make oligonucleotides in a manner such that the 5' phosphate of one mononucleotide pentose ring is attached to the 3' oxygen of its neighbor in one direction via a phosphodiester linkage.
  • An end of an oligonucleotide is referred to as the "5' end” if its 5' phosphate is not linked to the 3' oxygen of a mononucleotide pentose ring.
  • An end of an oligonucleotide is referred to as the "3' end” if its 3' oxygen is not linked to a 5' phosphate of another mononucleotide pentose ring.
  • a nucleic acid sequence even if internal to a larger oligonucleotide, also may be said to have 5' and 3' ends.
  • discrete elements are referred to as being "upstream” or 5' of the "downstream” or 3' elements.
  • Codon optimization refers to a process of modifying a nucleic acid sequence for enhanced expression in particular host cells by replacing at least one codon of the native sequence with a codon that is more frequently or most frequently used in the genes of the host cell while maintaining the native amino acid sequence.
  • a polynucleotide encoding a fusion polypeptide can be modified to substitute codons having a higher frequency of usage in a given Listeria cell or any other host cell as compared to the naturally occurring nucleic acid sequence. Codon usage tables are readily available, for example, at the "Codon Usage Database.” The optimal codons utilized by L.
  • plasmid or “vector” includes any known delivery vector including a bacterial delivery vector, a viral vector delivery vector, a peptide immunotherapy delivery vector, a DNA immunotherapy delivery vector, an episomal plasmid, an integrative plasmid, or a phage vector.
  • vector refers to a construct which is capable of delivering, and, optionally, expressing, one or more fusion polypeptides in a host cell.
  • episomal plasmid or "extrachromosomal plasmid” refers to a nucleic acid vector that is physically separate from chromosomal DNA (i.e., episomal or extrachromosomal and does not integrated into a host cell's genome) and replicates independently of chromosomal DNA.
  • a plasmid may be linear or circular, and it may be single- stranded or double-stranded.
  • Episomal plasmids may optionally persist in multiple copies in a host cell's cytoplasm (e.g., Listeria), resulting in amplification of any genes of interest within the episomal plasmid.
  • the term "genomically integrated” refers to a nucleic acid that has been introduced into a cell such that the nucleotide sequence integrates into the genome of the cell and is capable of being inherited by progeny thereof. Any protocol may be used for the stable incorporation of a nucleic acid into the genome of a cell.
  • stably maintained refers to maintenance of a nucleic acid molecule or plasmid in the absence of selection (e.g., antibiotic selection) for at least 10 generations without detectable loss.
  • the period can be at least 15 generations, 20 generations, at least 25 generations, at least 30 generations, at least 40 generations, at least 50 generations, at least 60 generations, at least 80 generations, at least 100 generations, at least 150 generations, at least 200 generations, at least 300 generations, or at least 500 generations.
  • Stably maintained can refer to a nucleic acid molecule or plasmid being maintained stably in cells in vitro (e.g., in culture), being maintained stably in vivo, or both.
  • An "open reading frame” or “ORF” is a portion of a DNA which contains a sequence of bases that could potentially encode a protein.
  • an ORF can be located between the start-code sequence (initiation codon) and the stop-codon sequence (termination codon) of a gene.
  • a "promoter” is a regulatory region of DNA usually comprising a TATA box capable of directing RNA polymerase II to initiate RNA synthesis at the appropriate transcription initiation site for a particular polynucleotide sequence.
  • a promoter may additionally comprise other regions which influence the transcription initiation rate.
  • the promoter sequences disclosed herein modulate transcription of an operably linked polynucleotide.
  • a promoter can be active in one or more of the cell types disclosed herein (e.g., a eukaryotic cell, a non-human mammalian cell, a human cell, a rodent cell, a pluripotent cell, a one-cell stage embryo, a differentiated cell, or a combination thereof).
  • a promoter can be, for example, a constitutively active promoter, a conditional promoter, an inducible promoter, a temporally restricted promoter (e.g., a developmentally regulated promoter), or a spatially restricted promoter (e.g., a cell-specific or tissue- specific promoter). Examples of promoters can be found, for example, in WO 2013/176772, herein incorporated by reference in its entirety.
  • “Operable linkage" or being “operably linked” refers to the juxtaposition of two or more components (e.g., a promoter and another sequence element) such that both components function normally and allow the possibility that at least one of the
  • a promoter can be operably linked to a coding sequence if the promoter controls the level of transcription of the coding sequence in response to the presence or absence of one or more transcriptional regulatory factors. Operable linkage can include such sequences being contiguous with each other or acting in trans (e.g., a regulatory sequence can act at a distance to control transcription of the coding sequence).
  • sequence identity in the context of two polynucleotides or polypeptide sequences makes reference to the residues in the two sequences that are the same when aligned for maximum correspondence over a specified comparison window.
  • sequence identity or “identity” in the context of two polynucleotides or polypeptide sequences makes reference to the residues in the two sequences that are the same when aligned for maximum correspondence over a specified comparison window.
  • Sequences that differ by such conservative substitutions are said to have "sequence similarity" or "similarity.” Means for making this adjustment are well known to those of skill in the art. Typically, this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, California).
  • Percentage of sequence identity refers to the value determined by comparing two optimally aligned sequences (greatest number of perfectly matched residues) over a comparison window, wherein the portion of the polynucleotide sequence in the
  • comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison, and multiplying the result by 100 to yield the percentage of sequence identity.
  • the comparison window is the full length of the shorter of the two sequences being compared.
  • sequence identity/similarity values refer to the value obtained using GAP Version 10 using the following parameters: % identity and % similarity for a nucleotide sequence using GAP Weight of 50 and Length Weight of 3, and the nwsgapdna.cmp scoring matrix; % identity and % similarity for an amino acid sequence using GAP Weight of 8 and Length Weight of 2, and the BLOSUM62 scoring matrix; or any equivalent program thereof.
  • "Equivalent program” includes any sequence comparison program that, for any two sequences in question, generates an alignment having identical nucleotide or amino acid residue matches and an identical percent sequence identity when compared to the corresponding alignment generated by GAP Version 10.
  • conservative amino acid substitution refers to the substitution of an amino acid that is normally present in the sequence with a different amino acid of similar size, charge, or polarity.
  • conservative substitutions include the substitution of a non-polar (hydrophobic) residue such as isoleucine, valine, or leucine for another non- polar residue.
  • conservative substitutions include the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, or between glycine and serine.
  • substitution of a basic residue such as lysine, arginine, or histidine for another, or the substitution of one acidic residue such as aspartic acid or glutamic acid for another acidic residue are additional examples of conservative substitutions.
  • non-conservative substitutions include the substitution of a non-polar (hydrophobic) amino acid residue such as isoleucine, valine, leucine, alanine, or methionine for a polar (hydrophilic) residue such as cysteine, glutamine, glutamic acid or lysine and/or a polar residue for a non-polar residue.
  • Typical amino acid categorizations are summarized below. Alanine Ala A Nonpolar Neutral 1.8
  • a "homologous" sequence refers to a sequence that is either identical or substantially similar to a known reference sequence, such that it is, for example, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the known reference sequence.
  • wild type refers to entities having a structure and/or activity as found in a normal (as contrasted with mutant, diseased, altered, or so forth) state or context. Wild type gene and polypeptides often exist in multiple different forms (e.g., alleles).
  • isolated with respect to proteins and nucleic acid refers to proteins and nucleic acids that are relatively purified with respect to other bacterial, viral or cellular components that may normally be present in situ, up to and including a substantially pure preparation of the protein and the polynucleotide.
  • isolated also includes proteins and nucleic acids that have no naturally occurring counterpart, have been chemically synthesized and are thus substantially uncontaminated by other proteins or nucleic acids, or has been separated or purified from most other cellular components with which they are naturally accompanied (e.g., other cellular proteins, polynucleotides, or cellular components).
  • Exogenous or heterologous molecules or sequences are molecules or sequences that are not normally expressed in a cell or are not normally present in a cell in that form. Normal presence includes presence with respect to the particular developmental stage and environmental conditions of the cell.
  • An exogenous or heterologous molecule or sequence for example, can include a mutated version of a corresponding endogenous sequence within the cell or can include a sequence corresponding to an endogenous sequence within the cell but in a different form (i.e., not within a chromosome).
  • An exogenous or heterologous molecule or sequence in a particular cell can also be a molecule or sequence derived from a different species than a reference species of the cell or from a different organism within the same species. For example, in the case of a
  • the heterologous polypeptide could be a polypeptide that is not native or endogenous to the Listeria strain, that is not normally expressed by the Listeria strain, from a source other than the Listeria strain, derived from a different organism within the same species.
  • endogenous molecules or sequences or “native” molecules or sequences are molecules or sequences that are normally present in that form in a particular cell at a particular developmental stage under particular environmental conditions.
  • variant refers to an amino acid or nucleic acid sequence (or an organism or tissue) that is different from the majority of the population but is still sufficiently similar to the common mode to be considered to be one of them (e.g., splice variants).
  • isoform refers to a version of a molecule (e.g., a protein) with only slight differences compared to another isoform, or version (e.g., of the same protein).
  • protein isoforms may be produced from different but related genes, they may arise from the same gene by alternative splicing, or they may arise from single nucleotide polymorphisms.
  • fragment when referring to a protein means a protein that is shorter or has fewer amino acids than the full length protein.
  • fragment when referring to a nucleic acid means a nucleic acid that is shorter or has fewer nucleotides than the full length nucleic acid.
  • a fragment can be, for example, an N-terminal fragment (i.e., removal of a portion of the C-terminal end of the protein), a C-terminal fragment (i.e., removal of a portion of the N-terminal end of the protein), or an internal fragment.
  • a fragment can also be, for example, a functional fragment or an immunogenic fragment.
  • analog when referring to a protein means a protein that differs from a naturally occurring protein by conservative amino acid differences, by modifications which do not affect amino acid sequence, or by both.
  • the term "functional” refers to the innate ability of a protein or nucleic acid (or a fragment, isoform, or variant thereof) to exhibit a biological activity or function.
  • biological activities or functions can include, for example, the ability to elicit an immune response when administered to a subject.
  • biological activities or functions can also include, for example, binding to an interaction partner.
  • 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.
  • immunogenicity refers to the innate ability of a molecule (e.g., a protein, a nucleic acid, an antigen, or an organism) to elicit an immune response in a subject when administered to the subject. Immunogenicity can be measured, for example, by a greater number of antibodies to the molecule, a greater diversity of antibodies to the molecule, a greater number of T-cells specific for the molecule, a greater cytotoxic or helper T-cell response to the molecule, and the like.
  • a molecule e.g., a protein, a nucleic acid, an antigen, or an organism
  • Immunogenicity can be measured, for example, by a greater number of antibodies to the molecule, a greater diversity of antibodies to the molecule, a greater number of T-cells specific for the molecule, a greater cytotoxic or helper T-cell response to the molecule, and the like.
  • antigen is used herein to refer to a substance that, when placed in contact with a subject or organism (e.g., when present in or when detected by the subject or organism), results in a detectable immune response from the subject or organism.
  • An antigen may be, for example, a lipid, a protein, a carbohydrate, a nucleic acid, or combinations and variations thereof.
  • an "antigenic peptide” refers to a peptide that leads to the mounting of an immune response in a subject or organism when present in or detected by the subject or organism.
  • an "antigenic peptide” may encompass proteins that are loaded onto and presented on MHC class I and/or class II molecules on a host cell's surface and can be recognized or detected by an immune cell of the host, thereby leading to the mounting of an immune response against the protein.
  • an immune response may also extend to other cells within the host, such as diseased cells (e.g., tumor or cancer cells) that express the same protein.
  • epitope refers to a site on an antigen that is recognized by the immune system (e.g., to which an antibody binds).
  • An epitope can be formed from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of one or more proteins. Epitopes formed from contiguous amino acids (also known as linear epitopes) are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding (also known as conformational epitopes) are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols, in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed. (1996), herein incorporated by reference in its entirety for all purposes.
  • mutation refers to the any change of the structure of a gene or a protein.
  • a mutation can result from a deletion, an insertion, a substitution, or a rearrangement of chromosome or a protein.
  • An "insertion” changes the number of nucleotides in a gene or the number of amino acids in a protein by adding one or more additional nucleotides or amino acids.
  • a “deletion” changes the number of nucleotides in a gene or the number of amino acids in a protein by reducing one or more additional nucleotides or amino acids.
  • a "frameshift" mutation in DNA occurs when the addition or loss of nucleotides changes a gene's reading frame.
  • a reading frame consists of groups of 3 bases that each code for one amino acid.
  • a frameshift mutation shifts the grouping of these bases and changes the code for amino acids.
  • the resulting protein is usually nonfunctional. Insertions and deletions can each be frameshift mutations.
  • a "missense” mutation or substitution refers to a change in one amino acid of a protein or a point mutation in a single nucleotide resulting in a change in an encoded amino acid.
  • a point mutation in a single nucleotide that results in a change in one amino acid is a "nonsynonymous" substitution in the DNA sequence.
  • Nonsynonymous substitutions can also result in a "nonsense" mutation in which a codon is changed to a premature stop codon that results in truncation of the resulting protein.
  • a nonsense mutation in which a codon is changed to a premature stop codon that results in truncation of the resulting protein.
  • “synonymous” mutation in a DNA is one that does not alter the amino acid sequence of a protein (due to codon degeneracy).
  • the term "somatic mutation” includes genetic alterations acquired by a cell other than a germ cell (e.g., sperm or egg). Such mutations can be passed on to progeny of the mutated cell in the course of cell division but are not inheritable. In contrast, a germinal mutation occurs in the germ line and can be passed on to the next generation of offspring.
  • in vitro refers to artificial environments and to processes or reactions that occur within an artificial environment (e.g., a test tube).
  • in vivo refers to natural environments (e.g., a cell or organism or body) and to processes or reactions that occur within a natural environment.
  • compositions or methods "comprising” or “including” one or more recited elements may include other elements not specifically recited.
  • a composition that "comprises” or “includes” a protein may contain the protein alone or in combination with other ingredients.
  • Designation of a range of values includes all integers within or defining the range, and all subranges defined by integers within the range.
  • the term "about” encompasses values within a standard margin of error of measurement (e.g., SEM) of a stated value or variations + 0.5%, 1%, 5%, or 10% from a specified value.
  • an antigen or “at least one antigen” can include a plurality of antigens, including mixtures thereof.
  • recombinant fusion polypeptides comprising an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem (e.g., fused to a PEST- containing peptide).
  • the HPV16 antigenic peptide in an HPV16 E6 antigenic peptide or an HPV16 E7 antigenic peptide and the HPV18 antigenic peptide is an HPV18 E6 antigenic peptide or an HPV18 E7 antigenic peptide.
  • the HPV16 antigenic peptide is an HPV16 E7 antigenic peptide and the HPV18 antigenic peptide is an HPV18 E7 antigenic peptide.
  • nucleic acids encoding such fusion polypeptides; recombinant bacteria or Listeria strains comprising such fusion polypeptides or such nucleic acids; cell banks comprising such recombinant bacteria or Listeria strains; immunogenic compositions, pharmaceutical compositions, and vaccines comprising such fusion polypeptides, such nucleic acids, or such recombinant bacteria or Listeria strains; and methods of generating such fusion polypeptides, such nucleic acids, and such recombinant bacteria or Listeria strains.
  • HPV-associated cancers are attributable to HPV types 16 and 18. However, HPV-associated cancers are typically not positive for both HPV type 16 and HPV type 18. Because it is rare for a patient to be both HPV16-positive and HPV18- positive, there is a lack of motivation to create immunotherapies that simultaneously target both HPV 16- specific and HPV18-specific antigens. This is particularly true for Listeria- based immunotherapy platforms, which can be limited in their capacity for number and size of nucleic acid sequences encoding antigenic peptides that can inserted.
  • Listeria-based immunotherapies e.g., Lm technology
  • bioengineered to secrete an antigenic peptide from one type of HPV can surprisingly increase the twelve-month overall survival rate in patients with a cancer or tumor associated with a different type of HPV.
  • the Lm technology has a mechanism of action that incorporates potent innate immune stimulation, delivery of a target peptide directly into the cytosol of dendritic cells and antigen presenting cells, generation of a targeted T cell response, and reduced immune suppression by regulatory T cells and myeloid-derived suppressor cells in the tumor microenvironment. Multiple treatments can be given and/or combined without neutralizing antibodies.
  • the Lm technology can use, for example, live, attenuated, bioengineered Lm bacteria to stimulate the immune system to view tumor cells as potentially bacterial- infected cells and target them for elimination.
  • the technology process can start with a live, attenuated strain of Listeria and can add, for example, multiple copies of a plasmid that encodes a fusion protein sequence including a fragment of, for example, the LLO (listeriolysin O) molecule joined to the antigen of interest.
  • This fusion protein is secreted by the Listeria inside antigen-presenting cells. This results in a stimulation of both the innate and adaptive arms of the immune system that reduces tumor defense mechanisms and makes it easier for the immune system to attack and destroy the cancer cells. //. Recombinant Fusion Polypeptides
  • recombinant fusion polypeptides comprising a PEST- containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.
  • fusion polypeptides comprising an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem, and wherein the fusion polypeptide does not comprise a PEST-containing peptide.
  • fusion polypeptides comprising from N- terminal end to C-terminal end a bacterial secretion sequence, a ubiquitin (Ub) protein, and two or more antigenic peptides (i.e., in tandem, such as Ub-peptidel-peptide2), wherein an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.
  • Ub protein e.g., Ubl- peptidel ; Ub2-peptide2).
  • Nucleic acids encoding such recombinant fusion polypeptides are also disclosed.
  • Such minigene nucleic acid constructs can further comprise two or more open reading frames linked by a Shine-Dalgarno ribosome binding site nucleic acid sequence between each open reading frame.
  • a minigene nucleic acid construct can further comprise two to four open reading frames linked by a Shine-Dalgarno ribosome binding site nucleic acid sequence between each open reading frame.
  • Each open reading frame can encode a different polypeptide.
  • the codon encoding the carboxy terminus of the fusion polypeptide is followed by two stop codons to ensure termination of protein synthesis.
  • the bacterial signal sequence can be a Listerial signal sequence, such as an Hly or an ActA signal sequence, or any other known signal sequence. In other cases, the signal sequence can be an LLO signal sequence.
  • the signal sequence can be bacterial, can be native to a host bacterium (e.g., Listeria monocytogenes, such as a secAl signal peptide), or can be foreign to a host bacterium.
  • signal peptides include an Usp45 signal peptide from Lactococcus lactis, a Protective Antigen signal peptide from Bacillus anthracis, a secA2 signal peptide such the p60 signal peptide from Listeria monocytogenes, and a Tat signal peptide such as a B. subtilis Tat signal peptide (e.g., PhoD).
  • the secretion signal sequence is from a Listeria protein, such as an ActA 3 oo secretion signal or an ActAioo secretion signal.
  • the ubiquitin can be, for example, a full-length protein.
  • the ubiquitin expressed from the nucleic acid construct provided herein can be cleaved at the carboxy terminus from the rest of the recombinant fusion polypeptide expressed from the nucleic acid construct through the action of hydrolases upon entry to the host cell cytosol. This liberates the amino terminus of the fusion polypeptide, producing a peptide in the host cell cytosol.
  • the recombinant fusion polypeptides can comprise one or more tags.
  • the recombinant fusion polypeptides can comprise one or more peptide tags N- terminal and/or C-terminal to the combination of the HPV16 antigenic peptide and the HPV18 antigenic peptide operably linked in tandem.
  • a tag can be fused directly to an antigenic peptide or linked to an antigenic peptide via a linker (examples of which are disclosed elsewhere herein). Examples of tags include the following: FLAG tag,
  • SIINFEKL tag is set forth in SEQ ID NO: 16 (encoded by any one of the nucleic acids set forth in SEQ ID NO: 16 (encoded by any one of the nucleic acids set forth in SEQ ID NO: 16 (encoded by any one of the nucleic acids set forth in SEQ ID NO: 16 (encoded by any one of the nucleic acids set forth in SEQ ID NO: 16 (encoded by any one of the nucleic acids set forth in SEQ ID NO: 16 (encoded by any one of the nucleic acids set forth in SEQ ID NO: 16
  • 3xFLAG tag is set forth in SEQ ID NO: 32 (encoded by any one of the nucleic acids set forth in SEQ ID NOS: 17-31).
  • Other tags include chitin binding protein (CBP), maltose binding protein (MBP), glutathione-S-transferase (GST), thioredoxin (TRX), and poly(NANP).
  • CBP chitin binding protein
  • MBP maltose binding protein
  • GST glutathione-S-transferase
  • TRX thioredoxin
  • poly(NANP) poly(NANP).
  • Particular recombinant fusion polypeptides comprise a C-terminal SIINFEKL tag.
  • Such tags can allow for easy detection of the recombinant fusion protein, confirmation of secretion of the recombinant fusion protein, or for following the immunogenicity of the secreted fusion polypeptide by following immune responses to these "tag" sequence peptides.
  • immune response can be monitored using a number of reagents including, for example, monoclonal antibodies and DNA or RNA probes specific for these tags.
  • the recombinant fusion polypeptides disclosed herein can be expressed by recombinant Listeria strains or can be expressed and isolated from other vectors and cell systems used for protein expression and isolation.
  • Recombinant Listeria strains comprising expressing such antigenic peptides can be used, for example in immunogenic compositions comprising such recombinant Listeria and in vaccines comprising the recombinant Listeria strain and an adjuvant.
  • antigenic peptides as a fusion polypeptides with a nonhemolytic truncated form of LLO, ActA, or a PEST- like sequence in host cell systems in Listeria strains and host cell systems other than Listeria can result in enhanced immunogenicity of the antigenic peptides.
  • nucleic acids encoding such recombinant fusion polypeptides are also disclosed.
  • the nucleic acid can be in any form.
  • the nucleic acid can comprise or consist of DNA or RNA, and can be single- stranded or double- stranded.
  • the nucleic acid can be in the form of a plasmid, such as an episomal plasmid, a multicopy episomal plasmid, or an integrative plasmid.
  • the nucleic acid can be in the form of a viral vector, a phage vector, or in a bacterial artificial chromosome.
  • nucleic acids can have one open reading frame or can have two or more open reading frames (e.g., an open reading frame encoding the recombinant fusion polypeptide and a second open reading frame encoding a metabolic enzyme).
  • such nucleic acids can comprise two or more open reading frames linked by a Shine-Dalgarno ribosome binding site nucleic acid sequence between each open reading frame.
  • a nucleic acid can comprise two to four open reading frames linked by a Shine-Dalgarno ribosome binding site nucleic acid sequence between each open reading frame.
  • Each open reading frame can encode a different polypeptide.
  • the codon encoding the carboxy terminus of the fusion polypeptide is followed by two stop codons to ensure termination of protein synthesis.
  • the antigenic peptides used herein can include a combination of any HPV 16- specific peptide and any HPV18-specific peptide.
  • Such peptides can be HPV 16- specific and HPV18-specific full-length proteins or fragments thereof.
  • Exemplary HPV 16- specific and HPV18-specific proteins includes E6 and E7 proteins from HPV16 and HPV18.
  • the E6 and E7 proteins are oncoproteins that act by stimulating the destruction of many host cell key regulatory proteins. Examples of HPV16 and HPV18 E6 and E7 proteins include HPV 16 E7 (GenBank Accession Nos. AHK23257 and AAD33253; a 98 amino acid protein), HPV 16 E6 (GenBank Accession Nos.
  • HPV 18 E7 GenBank Accession Nos. AGM34461 and P06788; a 105 amino acid protein
  • HPV18 E6 GenBank Accession No. P06463; a 158 amino acid protein
  • An exemplary HPV16 E7 protein is set forth in SEQ ID NO: 96 (encoded by the DNA sequence set forth in SEQ ID NO: 95)
  • an exemplary HPV18 E7 protein is set forth in SEQ ID NO: 98 (encoded by the DNA sequence set forth in SEQ ID NO: 97).
  • a suitable HPV16 E7 peptide can be, for example, a protein that is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 96 or a fragment thereof.
  • a suitable HPV18 E7 peptide can be, for example, a protein that is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 98 or a fragment thereof.
  • the fusion polypeptide can include at least two antigenic peptides.
  • the fusion polypeptide can include 2, 3, 4, 5, 6, 7, 8, 9, or 10 antigenic peptides.
  • Each antigenic peptide can be of any length sufficient to induce an immune response, and each antigenic peptide can be the same length or the antigenic peptides can have different lengths.
  • an antigenic peptide disclosed herein can be about 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-110, 110-120, 120-130, 130-140, 140-150, 150-160, or 95 to 160 amino acids in length.
  • Each antigenic peptide can also be hydrophilic or can score up to or below a certain hydropathy threshold, which can be predictive of secretability in Listeria monocytogenes or another bacteria of interest.
  • a certain hydropathy threshold can be predictive of secretability in Listeria monocytogenes or another bacteria of interest.
  • antigenic peptides can be scored by a Kyte and Doolittle hydropathy index 21 amino acid window, and all scoring above a cutoff (around 1.6) can be excluded as they are unlikely to be secretable by
  • the combination of antigenic peptides or the fusion polypeptide can be hydrophilic or can score up to or below a certain hydropathy threshold, which can be predictive of secretability in Listeria monocytogenes or another bacteria of interest.
  • the antigenic peptides can be linked together in any manner.
  • the antigenic peptides can be fused directly to each other with no intervening sequence.
  • the antigenic peptides can be linked to each other indirectly via one or more linkers, such as peptide linkers.
  • some pairs of adjacent antigenic peptides can be fused directly to each other, and other pairs of antigenic peptides can be linked to each other indirectly via one or more linkers.
  • the same linker can be used between each pair of adjacent antigenic peptides, or any number of different linkers can be used between different pairs of adjacent antigenic peptides.
  • one linker can be used between a pair of adjacent antigenic peptides, or multiple linkers can be used between a pair of adjacent antigenic peptides.
  • Any suitable sequence can be used for a peptide linker.
  • a linker sequence may be, for example, from 1 to about 50 amino acids in length. Some linkers may be hydrophilic. The linkers can serve varying purposes. For example, the linkers can serve to increase bacterial secretion, to facilitate antigen processing, to increase flexibility of the fusion polypeptide, to increase rigidity of the fusion polypeptide, or any other purpose. In some cases, different amino acid linker sequences are distributed between the antigenic peptides or different nucleic acids encoding the same amino acid linker sequence are distributed between the antigenic peptides (e.g., SEQ ID NOS: 84-94) in order to minimize repeats.
  • peptide linker sequences may be chosen, for example, based on one or more of the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the antigenic peptides; and (3) the lack of hydrophobic or charged residues that might react with the functional epitopes.
  • peptide linker sequences may contain Gly, Asn and Ser residues.
  • linker sequences which may be usefully employed as linkers include those disclosed in Maratea et al. (1985) Gene 40:39-46; Murphy et al. (1986) Proc Natl Acad Sci USA 83:8258-8262; US 4,935,233; and US 4,751,180, each of which is herein incorporated by reference in its entirety for all purposes.
  • linkers include those in the following table (each of which can be used by itself as a linker, in a linker comprising repeats of the sequence, or in a linker further comprising one or more of the other sequences in the table), although others can also be envisioned ⁇ see, e.g., Reddy Chichili et al. (2013) Protein Science 22: 153-167, herein incorporated by reference in its entirety for all purposes). Unless specified, "n" represents an undetermined number of repeats in the listed linker.
  • the recombinant fusion proteins disclosed herein comprise a PEST-containing peptide.
  • the PEST-containing peptide may at the amino terminal (N-terminal) end of the fusion polypeptide (i.e., N-terminal to the antigenic peptides), may be at the carboxy terminal (C-terminal) end of the fusion polypeptide (i.e., C-terminal to the antigenic peptides), or may be embedded within the antigenic peptides.
  • a PEST containing peptide is not part of and is separate from the fusion polypeptide.
  • Fusion of an antigenic peptides to a PEST-like sequence, such as an LLO peptide can enhance the immunogenicity of the antigenic peptides and can increase cell- mediated and antitumor immune responses (i.e., increase cell- mediated and anti-tumor immunity). See, e.g., Singh et al. (2005) J Immunol 175(6):3663-3673, herein incorporated by reference in its entirety for all purposes.
  • a PEST-containing peptide is one that comprises a PEST sequence or a PEST- like sequence.
  • PEST sequences in eukaryotic proteins have long been identified. For example, proteins containing amino acid sequences that are rich in prolines (P), glutamic acids (E), serines (S) and threonines (T) (PEST), generally, but not always, flanked by clusters containing several positively charged amino acids, have rapid intracellular half- lives (Rogers et al. (1986) Science 234:364-369, herein incorporated by reference in its entirety for all purposes).
  • a PEST or PEST-like sequence can be identified using the PEST-find program.
  • a PEST-like sequence can be a region rich in proline (P), glutamic acid (E), serine (S), and threonine (T) residues.
  • the PEST-like sequence can be flanked by one or more clusters containing several positively charged amino acids.
  • a PEST-like sequence can be defined as a hydrophilic stretch of at least 12 amino acids in length with a high local concentration of proline (P), aspartate (D), glutamate (E), serine (S), and/or threonine (T) residues.
  • P proline
  • D aspartate
  • E glutamate
  • S serine
  • T threonine residues.
  • a PEST-like sequence contains no positively charged amino acids, namely arginine (R), histidine (H), and lysine (K).
  • Some PEST-like sequences can contain one or more internal phosphorylation sites, and phosphorylation at these sites precedes protein degradation.
  • the PEST-like sequence fits an algorithm disclosed in Rogers et al. In another example, the PEST-like sequence fits an algorithm disclosed in
  • PEST-like sequences can also be identified by an initial scan for positively charged amino acids R, H, and K within the specified protein sequence. All amino acids between the positively charged flanks are counted, and only those motifs containing a number of amino acids equal to or higher than the window-size parameter are considered further.
  • a PEST-like sequence must contain at least one P, at least one D or E, and at least one S or T.
  • the quality of a PEST motif can be refined by means of a scoring parameter based on the local enrichment of critical amino acids as well as the motifs hydrophobicity.
  • Enrichment of D, E, P, S, and T is expressed in mass percent (w/w) and corrected for one equivalent of D or E, onel of P, and one of S or T.
  • Calculation of hydrophobicity can also follow in principle the method of Kyte and Doolittle (1982) J. Mol. Biol. 157: 105, herein incorporated by reference in its entirety for all purposes.
  • Kyte-Doolittle hydropathy indices which originally ranged from -4.5 for arginine to +4.5 for isoleucine, are converted to positive integers, using the following linear
  • a potential PEST motif's hydrophobicity can also be calculated as the sum over the products of mole percent and hydrophobicity index for each amino acid species.
  • a PEST-containing peptide can refer to a peptide having a score of at least +5 using the above algorithm. Alternatively, it can refer to a peptide having a score of at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, at least 30, at least 32, at least 35, at least 38, at least 40, or at least 45.
  • any other available methods or algorithms known in the art can also be used to identify PEST-like sequences. See, e.g., the CaSPredictor (Garay-Malpartida et al. (2005) Bioinformatics 21 Suppl l:il69-76, herein incorporated by reference in its entirety for all purposes).
  • Another method that can be used is the following: a PEST index is calculated for each stretch of appropriate length (e.g. a 30-35 amino acid stretch) by assigning a value of one to the amino acids Ser, Thr, Pro, Glu, Asp, Asn, or Gin.
  • the coefficient value (CV) for each of the PEST residues is one and the CV for each of the other AA (non-PEST) is zero.
  • Examples of PEST-like amino acid sequences are those set forth in SEQ ID NOS: 43-51.
  • One example of a PEST-like sequence is
  • KENS IS S M APP AS PP AS PKTPIEKKH ADEID K SEQ ID NO: 43.
  • KENSISSMAPPASPPASPK SEQ ID NO: 44.
  • any PEST or PEST-like amino acid sequence can be used.
  • PEST sequence peptides are known and are described, for example, in US 7,635,479; US 7,665,238; and US 2014/0186387, each of which is herein incorporated by reference in its entirety for all purposes.
  • the PEST-like sequence can be from a Listeria species, such as from Listeria monocytogenes.
  • the Listeria monocytogenes ActA protein contains at least four such sequences (SEQ ID NOS: 45-48), any of which are suitable for use in the compositions and methods disclosed herein.
  • Other similar PEST-like sequences include SEQ ID NOS: 52-54.
  • Streptolysin O proteins from Streptococcus sp. also contain a PEST sequence.
  • Streptococcus pyogenes streptolysin O comprises the PEST sequence KQNTASTETTTTNEQPK (SEQ ID NO: 49) at amino acids 35-51 and
  • Streptococcus equisimilis streptolysin O comprises the PEST-like sequence
  • KQNTANTETTTTNEQPK (SEQ ID NO: 50) at amino acids 38-54.
  • Another example of a PEST-like sequence is from Listeria seeligeri cytolysin, encoded by the Iso gene:
  • RSEVTISPAETPESPPATP (e.g., SEQ ID NO: 51).
  • the PEST-like sequence can be derived from other prokaryotic organisms.
  • Other prokaryotic organisms wherein PEST-like amino acid sequences would be expected include, for example, other Listeria species.
  • LLO listeriolysin O
  • An example of an LLO protein is the protein assigned GenBank Accession No. P13128 (SEQ ID NO: 55; nucleic acid sequence is set forth in GenBank Accession No. X15127).
  • SEQ ID NO: 55 is a proprotein including a signal sequence. The first 25 amino acids of the proprotein is the signal sequence and is cleaved from LLO when it is secreted by the bacterium, thereby resulting in the full-length active LLO protein of 504 amino acids without the signal sequence.
  • LLO peptide disclosed herein can comprise the signal sequence or can comprise a peptide that does not include the signal sequence.
  • Exemplary LLO proteins that can be used comprise, consist essentially of, or consist of the sequence set forth in SEQ ID NO: 55 or homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of variants, fragments of analogs, and fragments of isoforms of SEQ ID NO: 55. Any sequence that encodes a fragment of an LLO protein or a homologue, variant, isoform, analog, fragment of a homologue, fragment of a variant, or fragment of an analog of an LLO protein can be used.
  • a homologous LLO protein can have a sequence identity with a reference LLO protein, for example, of greater than 70%, 72%, 75%, 78%, 80%, 82%, 83%, 85%, 87%, 88%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, or 99%.
  • LLO proteins that can be used can comprise, consist essentially of, or consist of the sequence set forth in SEQ ID NO: 56 or homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of variants, fragments of analogs, and fragments of isoforms of SEQ ID NO: 56.
  • LLO protein is an LLO protein from the Listeria monocytogenes 10403S strain, as set forth in GenBank Accession No.: ZP_01942330 or EBA21833, or as encoded by the nucleic acid sequence as set forth in GenBank Accession No.: NZ_AARZ01000015 or AARZ01000015.1.
  • Another example of an LLO protein is an LLO protein from the Listeria monocytogenes 4b F2365 strain ⁇ see, e.g., GenBank Accession No.: YP_012823), EGD-e strain ⁇ see, e.g., GenBank Accession No.:
  • LLO protein from Flavobacteriales bacterium HTCC2170 ⁇ see, e.g., GenBank Accession No.: ZP_01106747 or EAR01433, or encoded by GenBank
  • LLO proteins that can be used can comprise, consist essentially of, or consist of any of the above LLO proteins or homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of variants, fragments of analogs, and fragments of isoforms of the above LLO proteins.
  • Proteins that are homologous to LLO, or homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of variants, fragments of analogs, and fragments of isoforms thereof, can also be used.
  • alveolysin which can be found, for example, in Paenibacillus alvei ⁇ see, e.g., GenBank Accession No.: P23564 or AAA22224, or encoded by GenBank Accession No.: M62709).
  • Other such homologous proteins are known.
  • the LLO peptide can be a full-length LLO protein or a truncated LLO protein or LLO fragment.
  • the LLO peptide can be one that retains one or more functionalities of a native LLO protein or lacks one or more functionalities of a native LLO protein.
  • the retained LLO functionality can be allowing a bacteria (e.g., Listeria) to escape from a phagosome or phagolysosome, or enhancing the immunogenicity of a peptide to which it is fused.
  • the retained functionality can also be hemolytic function or antigenic function.
  • the LLO peptide can be a non- hemolytic LLO.
  • Other functions of LLO are known, as are methods and assays for evaluating LLO functionality.
  • An LLO fragment can be a PEST-like sequence or can comprise a PEST-like sequence.
  • LLO fragments can comprise one or more of an internal deletion, a truncation from the C-terminal end, and a truncation from the N-terminal end. In some cases, an LLO fragment can comprise more than one internal deletion.
  • Other LLO peptides can be full-length LLO proteins with one or more mutations.
  • LLO proteins or fragments have reduced hemolytic activity relative to wild type LLO or are non-hemolytic fragments.
  • an LLO protein can be rendered non-hemolytic by deletion or mutation of the activation domain at the carboxy terminus, by deletion or mutation of cysteine 484, or by deletion or mutation at another location.
  • LLO proteins are rendered non-hemolytic by a deletion or mutation of the cholesterol binding domain (CBD) as detailed in US 8,771,702, herein incorporated by reference in its entirety for all purposes.
  • the mutations can comprise, for example, a substitution or a deletion.
  • the entire CBD can be mutated, portions of the CBD can be mutated, or specific residues within the CBD can be mutated.
  • the LLO protein can comprise a mutation of one or more of residues C484, W491, and W492 (e.g., C484, W491, W492, C484 and W491, C484 and W492, W491 and W492, or all three residues) of SEQ ID NO: 55 or corresponding residues when optimally aligned with SEQ ID NO: 55 (e.g., a corresponding cysteine or tryptophan residue).
  • a mutant LLO protein can be created wherein residues C484, W491, and W492 of LLO are substituted with alanine residues, which will substantially reduce hemolytic activity relative to wild type LLO.
  • the mutant LLO protein with C484A, W491A, and W492A mutations is termed "mutLLO.”
  • a mutant LLO protein can be created with an internal deletion comprising the cholesterol-binding domain.
  • the internal deletion can be a 1-11 amino acid deletion, an 11-50 amino acid deletion, or longer.
  • the mutated region can be 1-11 amino acids, 11-50 amino acids, or longer (e.g., 1-50, 1-11, 2-11, 3-11, 4-11, 5-11, 6-11, 7-11, 8-11, 9-11, 10-11, 1-2, 1-3, 1-4, 1-5, 1-6, 1- 7, 1-8, 1-9, 1-10, 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 2-9, 2-10, 3-4, 3-5, 3-6, 3-7, 3-8, 3-9, 3-10, 12-50, 11-15, 11-20, 11-25, 11-30, 11-35, 11-40, 11-50, 11-60, 11-70, 11-80, 11-90, 11- 100, 11-150, 15-20, 15-25, 15-30, 15-35, 15-40, 15-50, 15-60, 15-70, 15-80, 15-90, 15- 100, 15-150, 20-25, 20-30, 20-35, 20-40, 20-50, 20-60, 20-70, 20-80, 20-90, 20-100,
  • a mutated region consisting of residues 470-500, 470-510, or 480-500 of SEQ ID NO: 55 will result in a deleted sequence comprising the CBD (residues 483-493 of SEQ ID NO: 55).
  • the mutated region can also be a fragment of the CBD or can overlap with a portion of the CBD.
  • the mutated region can consist of residues 470-490, 480-488, 485-490, 486-488, 490-500, or 486-510 of SEQ ID NO: 55.
  • a fragment of the CBD (residues 484-492) can be replaced with a
  • the CBD (ECTGLAWEWWR; SEQ ID NO: 74) can be replaced with a CTL epitope from the antigen NY-ESO-1 (ESLLMWITQCR; SEQ ID NO: 75), which contains the HLA-A2 restricted epitope 157-165 from NY-ESO-1.
  • ctLLO The resulting LLO is termed "ctLLO.”
  • the mutated region can be replaced by a heterologous sequence.
  • the mutated region can be replaced by an equal number of heterologous amino acids, a smaller number of heterologous amino acids, or a larger number of amino acids (e.g., 1-50, 1-11, 2-11, 3-11, 4-11, 5-11, 6-11, 7-11, 8-11, 9- 11, 10-11, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 2-9, 2-10, 3-4, 3-5, 3-6, 3-7, 3-8, 3-9, 3-10, 12-50, 11-15, 11-20, 11-25, 11-30, 11-35, 11-40, 11-50, 11-60, 11-70, 11-80, 11-90, 11-100, 11-150, 15-20, 15-25, 15-30, 15-35, 15-40, 15-50, 15- 60, 15-70, 15-70, 15
  • an LLO peptide may have a deletion in the signal sequence and a mutation or substitution in the CBD.
  • LLO peptides are N-terminal LLO fragments (i.e., LLO proteins with a C-terminal deletion). Some LLO peptides are at least 494, 489, 492, 493, 500, 505, 510, 515, 520, or 525 amino acids in length or 492-528 amino acids in length.
  • the LLO fragment can consist of about the first 440 or 441 amino acids of an LLO protein (e.g., the first 441 amino acids of SEQ ID NO: 55 or 56, or a corresponding fragment of another LLO protein when optimally aligned with SEQ ID NO: 55 or 56).
  • N- terminal LLO fragments can consist of the first 420 amino acids of an LLO protein (e.g., the first 420 amino acids of SEQ ID NO: 55 or 56, or a corresponding fragment of another LLO protein when optimally aligned with SEQ ID NO: 55 or 56).
  • Other N-terminal fragments can consist of about amino acids 20-442 of an LLO protein (e.g., amino acids 20-442 of SEQ ID NO: 55 or 56, or a corresponding fragment of another LLO protein when optimally aligned with SEQ ID NO: 55 or 56).
  • Other N-terminal LLO fragments comprise any ALLO without the activation domain comprising cysteine 484, and in particular without cysteine 484.
  • the N-terminal LLO fragment can correspond to the first 425, 400, 375, 350, 325, 300, 275, 250, 225, 200, 175, 150, 125, 100, 75, 50, or 25 amino acids of an LLO protein (e.g., the first 425, 400, 375, 350, 325, 300, 275, 250, 225, 200, 175, 150, 125, 100, 75, 50, or 25 amino acids of SEQ ID NO: 55 or 56, or a corresponding fragment of another LLO protein when optimally aligned with SEQ ID NO: 55 or 56).
  • the fragment comprises one or more PEST-like sequences.
  • LLO fragments and truncated LLO proteins can contain residues of a homologous LLO protein that correspond to any one of the above specific amino acid ranges.
  • the residue numbers need not correspond exactly with the residue numbers enumerated above (e.g., if the homologous LLO protein has an insertion or deletion relative to a specific LLO protein disclosed herein).
  • Examples of N-terminal LLO fragments include SEQ ID NOS: 57, 58, and 59.
  • LLO proteins that can be used comprise, consist essentially of, or consist of the sequence set forth in SEQ ID NO: 57, 58, or 59 or homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of variants, fragments of analogs, and fragments of isoforms of SEQ ID NO: 57, 58, or 59.
  • the N-terminal LLO fragment set forth in SEQ ID NO: 59 is used.
  • An example of a nucleic acid encoding the N-terminal LLO fragment set forth in SEQ ID NO: 59 is SEQ ID NO: 60.
  • ActA is a surface- associated protein and acts as a scaffold in infected host cells to facilitate the
  • L. monocytogenes induces the polymerization of host actin filaments and uses the force generated by actin polymerization to move, first intracellularly and then from cell to cell.
  • ActA is responsible for mediating actin nucleation and actin-based motility.
  • the ActA protein provides multiple binding sites for host cytoskeletal components, thereby acting as a scaffold to assemble the cellular actin polymerization machinery.
  • the N-terminus of ActA binds to monomeric actin and acts as a constitutively active nucleation promoting factor by stimulating the intrinsic actin nucleation activity.
  • actA and hly genes are both members of the 10-kb gene cluster regulated by the transcriptional activator PrfA, and actA is upregulated approximately 226-fold in the mammalian cytosol.
  • Any sequence that encodes an ActA protein or a homologue, variant, isoform, analog, fragment of a homologue, fragment of a variant, or fragment of an analog of an ActA protein can be used.
  • a homologous ActA protein can have a sequence identity with a reference ActA protein, for example, of greater than 70%, 72%, 75%, 78%, 80%, 82%, 83%, 85%, 87%, 88%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, or 99%.
  • an ActA protein comprises, consists essentially of, or consists of the sequence set forth in SEQ ID NO: 61.
  • Another example of an ActA protein comprises, consists essentially of, or consists of the sequence set forth in SEQ ID NO: 62.
  • the first 29 amino acid of the proprotein corresponding to either of these sequences are the signal sequence and are cleaved from ActA protein when it is secreted by the bacterium.
  • An ActA peptide can comprise the signal sequence (e.g., amino acids 1-29 of SEQ ID NO: 61 or 62), or can comprise a peptide that does not include the signal sequence.
  • ActA proteins comprise, consist essentially of, or consist of homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of isoforms, or fragments of analogs of SEQ ID NO: 61 or 62.
  • an ActA protein is an ActA protein from the Listeria monocytogenes 10403S strain (GenBank Accession No.: DQ054585) the NICPBP 54002 strain (GenBank Accession No.: EU394959), the S3 strain (GenBank Accession No.: EU394960), NCTC 5348 strain (GenBank Accession No.: EU394961), NICPBP 54006 strain (GenBank Accession No.: EU394962), M7 strain (GenBank Accession No.:
  • LLO proteins that can be used can comprise, consist essentially of, or consist of any of the above LLO proteins or homologues, variants, isoforms, analogs, fragments, fragments of homologues, fragments of variants, fragments of analogs, and fragments of isoforms of the above LLO proteins.
  • ActA peptides can be full-length ActA proteins or truncated ActA proteins or ActA fragments (e.g., N-terminal ActA fragments in which a C-terminal portion is removed).
  • truncated ActA proteins comprise at least one PEST sequence (e.g., more than one PEST sequence).
  • truncated ActA proteins can optionally comprise an ActA signal peptide. Examples of PEST-like sequences contained in truncated ActA proteins include SEQ ID NOS: 45-48.
  • Some such truncated ActA proteins comprise at least two of the PEST-like sequences set forth in SEQ ID NOS: 45-48 or ho mo logs thereof, at least three of the PEST-like sequences set forth in SEQ ID NOS: 45- 48 or homo logs thereof, or all four of the PEST-like sequences set forth in SEQ ID NOS: 45-48 or homologs thereof.
  • Examples of truncated ActA proteins include those comprising, consisting essentially of, or consisting of about residues 30-122, about residues 30-229, about residues 30-332, about residues 30-200, or about residues 30-399 of a full length ActA protein sequence (e.g., SEQ ID NO: 62).
  • truncated ActA proteins include those comprising, consisting essentially of, or consisting of about the first 50, 100, 150, 200, 233, 250, 300, 390, 400, or 418 residues of a full length ActA protein sequence (e.g., SEQ ID NO: 62).
  • Other examples of truncated ActA proteins include those comprising, consisting essentially of, or consisting of about residues 200-300 or residues 300-400 of a full length ActA protein sequence (e.g., SEQ ID NO: 62).
  • the truncated ActA consists of the first 390 amino acids of the wild type ActA protein as described in US 7,655,238, herein incorporated by reference in its entirety for all purposes.
  • the truncated ActA can be 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 (SEQ ID NO: 73) substitution as described in US 2014/0186387, herein incorporated by references in its entirety for all purposes.
  • truncated ActA proteins can contain residues of a homologous ActA protein that corresponds to one of the above amino acid ranges or the amino acid ranges of any of the ActA peptides disclosed herein. The residue numbers need not correspond exactly with the residue numbers enumerated herein (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).
  • truncated ActA proteins include, for example, proteins
  • SEQ ID NO: 63 referred to as ActA/PESTl and consists of amino acids 30-122 of the full length ActA sequence set forth in SEQ ID NO: 62.
  • SEQ ID NO: 64 is referred to as ActA/PEST2 or LA229 and consists of amino acids 30-229 of the full length ActA sequence set forth in the full-length ActA sequence set forth in SEQ ID NO: 62.
  • SEQ ID NO: 65 is referred to as ActA/PEST3 and consists of amino acids 30-332 of the full-length ActA sequence set forth in SEQ ID NO: 62.
  • SEQ ID NO: 66 is referred to as
  • ActA/PEST4 and consists of amino acids 30-399 of the full-length ActA sequence set forth in SEQ ID NO: 62.
  • the truncated ActA protein consisting of the sequence set forth in SEQ ID NO: 64 can be used.
  • truncated ActA proteins include, for example, proteins comprising, consisting essentially of, or consisting of the sequence set forth in SEQ ID NO: 67, 69, 70, or 72 or homologues, variants, isoforms, analogs, fragments of variants, fragments of isoforms, or fragments of analogs of SEQ ID NO: 67, 69, 70, or 72.
  • the truncated ActA protein consisting of the sequence set forth in SEQ ID NO: 67 (encoded by the nucleic acid set forth in SEQ ID NO: 68) can be used.
  • the truncated ActA protein consisting of the sequence set forth in SEQ ID NO: 70 (encoded by the nucleic acid set forth in SEQ ID NO: 71) can be used.
  • SEQ ID NO: 71 is the first 1170 nucleotides encoding ActA in the Listeria monocytogenes 10403S strain.
  • the ActA fragment can be fused to a heterologous signal peptide.
  • SEQ ID NO: 72 sets forth an ActA fragment fused to an Hly signal peptide.
  • such methods can comprise selecting and designing antigenic peptides to include in the immunotherapy construct (and, for example, testing the hydropathy of the each antigenic peptide, and modifying or deselecting an antigenic peptide if it scores above a selected hydropathy index threshold value), designing one or more fusion polypeptides comprising each of the selected antigenic peptides, and generating a nucleic acid construct encoding the fusion polypeptide.
  • the antigenic peptides can be screened for hydrophobicity or hydrophilicity. Antigenic peptides can be selected, for example, if they are hydrophilic or if they score up to or below a certain hydropathy threshold, which can be predictive of secretability in a particular bacteria of interest (e.g., Listeria monocytogenes). For example, antigenic peptides can be scored by Kyte and Doolittle hydropathy index with a 21 amino acid window, all scoring above cutoff (around 1.6) are excluded as they are unlikely to be secretable by Listeria monocytogenes. See, e.g., Kyte-Doolittle (1982) J Mol Biol 157(1): 105-132; herein incorporated by reference in its entirety for all purposes.
  • an antigenic peptide scoring about a selected cutoff can be altered (e.g., changing the length of the antigenic peptide).
  • Other sliding window sizes that can be used include, for example, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27 or more amino acids.
  • the sliding window size can be 9-11 amino acids, 11-13 amino acids, 13-15 amino acids, 15-17 amino acids, 17-19 amino acids, 19-21 amino acids, 21-23 amino acids, 23-25 amino acids, or 25-27 amino acids.
  • cutoffs that can be used include, for example, the following ranges 1.2-1.4, 1.4-1.6, 1.6-1.8, 1.8-2.0, 2.0-2.2 2.2-2.5, 2.5- 3.0, 3.0-3.5, 3.5-4.0, or 4.0-4.5, or the cutoff can be 1.4, 1.5, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3,
  • the cutoff can vary, for example, depending on the genus or species of the bacteria being used to deliver the fusion polypeptide.
  • the antigenic peptides can be scored for their ability to bind to the subject human leukocyte antigen (HLA) type (for example by using the Immune Epitope Database (IED) available at www.iedb.org, which includes netMHCpan, ANN,
  • HLA human leukocyte antigen
  • SMMPMBEC SMM, CombLib_Sidney2008, PickPocket, and netMHCcons) and ranked by best MHC binding score from each antigenic peptide.
  • Other sources include TEpredict (tepredict.sourceforge.net/help.html) or other available MHC binding measurement scales. Cutoffs may be different for different expression vectors such as Salmonella.
  • the antigenic peptides can be screened for immunosuppressive epitopes (e.g., T-reg epitopes, IL-10-inducing T helper epitopes, and so forth) to deselect antigenic peptides or to avoid immunosuppressive influences.
  • immunosuppressive epitopes e.g., T-reg epitopes, IL-10-inducing T helper epitopes, and so forth
  • a predicative algorithm for immunogenicity of the epitopes can be used to screen the antigenic peptides.
  • these algorithms are at best 20% accurate in predicting which peptide will generate a T cell response.
  • the antigenic peptides can be screened for immunogenicity.
  • this can comprise contacting one or more T cells with an antigenic peptide, and analyzing for an immunogenic T cell response, wherein an immunogenic T cell response identifies the peptide as an immunogenic peptide.
  • This can also comprise using an immunogenic assay to measure secretion of at least one of CD25, CD44, or CD69 or to measure secretion of a cytokine selected from the group comprising IFN- ⁇ , TNF-a, IL-1, and IL-2 upon contacting the one or more T cells with the peptide, wherein increased secretion identifies the peptide as comprising one or more T cell epitopes.
  • the selected antigenic peptides can be arranged into one or more candidate orders for a potential fusion polypeptide. If there are more usable antigenic peptides than can fit into a single plasmid, different antigenic peptides can be assigned priority ranks as needed/desired and/or split up into different fusion polypeptides (e.g., for inclusion in different recombinant Listeria strains). Priority rank can be determined by factors such as relative size, priority of transcription, and/or overall hydrophobicity of the translated polypeptide.
  • the antigenic peptides can be arranged so that they are joined directly together without linkers, or any combination of linkers between any number of pairs of antigenic peptides, as disclosed in more detail elsewhere herein.
  • the number of linear antigenic peptides to be included can be determined based on consideration of the number of constructs needed versus the mutational burden, the efficiency of translation and secretion of multiple epitopes from a single plasmid, and the MOI needed for each bacteria or Lm comprising a plasmid.
  • the combination of antigenic peptides or the entire fusion polypeptide i.e., comprising the antigenic peptides and the PEST-containing peptide and any tags
  • the entirety of the fused antigenic peptides or the entire fusion polypeptide can be scored for hydropathy by a Kyte and Doolittle hydropathy index with a sliding 21 amino acid window.
  • the antigenic peptides can be reordered or shuffled within the fusion polypeptide until an acceptable order of antigenic peptides is found (i.e., one in which no region scores above the cutoff).
  • any problematic antigenic peptides can be removed or redesigned to be of a different size.
  • one or more linkers between antigenic peptides as disclosed elsewhere herein can be added or modified to change the hydrophobicity.
  • window sizes can be used, or other cutoffs can be used (e.g., depending on the genus or species of the bacteria being used to deliver the fusion polypeptide).
  • cutoffs e.g., depending on the genus or species of the bacteria being used to deliver the fusion polypeptide.
  • other suitable hydropathy plots or other appropriate scales could be used.
  • the combination of antigenic peptides or the entire fusion polypeptide can be further screened for immunosuppressive epitopes (e.g., T-reg epitopes, IL-10-inducing T helper epitopes, and so forth) to deselect antigenic peptides or to avoid immunosuppressive influences.
  • immunosuppressive epitopes e.g., T-reg epitopes, IL-10-inducing T helper epitopes, and so forth
  • a nucleic acid encoding a candidate combination of antigenic peptides or fusion polypeptide can then be designed and optimized.
  • the sequence can be optimized for increased levels of translation, duration of expression, levels of secretion, levels of transcription, and any combination thereof.
  • the increase can be 2- fold to 1000-fold, 2-fold to 500-fold, 2-fold to 100-fold, 2-fold to 50-fold, 2-fold to 20- fold, 2-fold to 10-fold, or 3-fold to 5-fold relative to a control, non-optimized sequence.
  • the fusion polypeptide or nucleic acid encoding the fusion polypeptide can be optimized for decreased levels of secondary structures possibly formed in the oligonucleotide sequence, or alternatively optimized to prevent attachment of any enzyme that may modify the sequence.
  • Expression in bacterial cells can be hampered, for example, by transcriptional silencing, low mRNA half-life, secondary structure formation, attachment sites of oligonucleotide binding molecules such as repressors and inhibitors, and availability of rare tRNAs pools. The source of many problems in bacterial expressions is found within the original sequence.
  • RNAs may include modification of cis acting elements, adaptation of its GC-content, modifying codon bias with respect to non-limiting tRNAs pools of the bacterial cell, and avoiding internal homologous regions.
  • optimizing a sequence can entail, for example, adjusting regions of very high (> 80%) or very low ( ⁇ 30%) GC content.
  • Optimizing a sequence can also entail, for example, avoiding one or more of the following cis-acting sequence motifs: internal TATA-boxes, chi-sites, and ribosomal entry sites; AT-rich or GC-rich sequence stretches; repeat sequences and RNA secondary structures; (cryptic) splice donor and acceptor sites; branch points; or a combination thereof.
  • Optimizing expression can also entail adding sequence elements to flanking regions of a gene and/or elsewhere in the plasmid.
  • Optimizing a sequence can also entail, for example, adapting the codon usage to the codon bias of host genes (e.g., Listeria monocytogenes genes).
  • host genes e.g., Listeria monocytogenes genes.
  • the codons below can be used for Listeria monocytogenes.
  • a nucleic acid encoding a fusion polypeptide can be generated and introduced into a delivery vehicle such as a bacteria strain or Listeria strain.
  • a delivery vehicle such as a bacteria strain or Listeria strain.
  • Other delivery vehicles may be suitable for DNA immunotherapy or peptide immunotherapy, such as a vaccinia virus or virus-like particle.
  • recombinant bacterial strains such as a Listeria strain, comprising a recombinant fusion polypeptide disclosed herein or a nucleic acid encoding the recombinant fusion polypeptide as disclosed elsewhere herein.
  • the bacterial strain is a Listeria strain, such as a Listeria monocytogenes (Lm) strain.
  • Lm has a number of inherent advantages as a vaccine vector. The bacterium grows very efficiently in vitro without special requirements, and it lacks LPS, which is a major toxicity factor in gram- negative bacteria, such as Salmonella. Genetically attenuated Lm vectors also offer additional safety as they can be readily eliminated with antibiotics, in case of serious adverse effects, and unlike some viral vectors, no integration of genetic material into the host genome occurs.
  • the recombinant Listeria strain can be any Listeria strain.
  • suitable Listeria strains include Listeria seeligeri, Listeria grayi, Listeria ivanovii, Listeria murrayi, Listeria welshimeri, Listeria monocytogenes (Lm), or any other Listeria species known in the art.
  • the recombinant listeria strain is a strain of the species Listeria monocytogenes.
  • Listeria monocytogenes strains include the following: L. monocytogenes 10403S wild type (see, e.g., Bishop and Hinrichs (1987) Immunol 139:2005-2009; Lauer et al. (2002) J Bact 184:4177-4186); L.
  • monocytogenes DP-L4056 which is phage cured (see, e.g., Lauer et al. (2002) Bact 184:4177-4186); L. monocytogenes DP-L4027, which is phage cured and has an hly gene deletion (see, e.g., Lauer et al. (2002) Bact 184:4177- 4186; Jones and Portnoy (1994) Infect Immunity 65:5608-5613); L. monocytogenes DP-L4029, which is phage cured and has an actA gene deletion (see, e.g., Lauer et al. (2002) J Bact 184:4177-4186; Skoble et al.
  • L monocytogenes DP-L4042 (delta PEST) (see, e.g., Brockstedt et al. (2004) Proc Natl Acad Sci. USA 101: 13832-13837 and supporting information); L.
  • LLO-S44A monocytogenes DP-L4097
  • LLO-S44A monocytogenes DP-L4097
  • LLO-S44A monocytogenes DP-L4097
  • LLO-S44A monocytogenes DP- L4364
  • delta IplA lipoate protein ligase
  • L. monocytogenes DP-L4405 delta MA
  • L. MA monocytogenes DP-L4405
  • monocytogenes DP-L4406 (delta inlB) (see, e.g., Brockstedt et al. (2004) Proc Natl Acad Sci USA 101: 13832-13837 and supporting information); L. monocytogenes CS-LOOOl (delta actA; delta inlB) (see, e.g., Brockstedt et al. (2004) Proc Natl Acad Sci USA 101: 13832-13837 and supporting information); L. monocytogenes CS- L0002 (delta actA; delta IplA) (see, e.g., Brockstedt et al. (2004) Proc Natl Acad Sci USA 101: 13832-13837 and supporting information); L.
  • LLO L461T monocytogenes CS-L0003
  • delta IplA monocytogenes CS-L0003
  • LLO L461T delta IplA
  • LLO L461T monocytogenes DP-L4038
  • delta actA LLO L461T
  • LLO S44A LLO L461T
  • L. monocytogenes strain with an IplAl deletion encoding lipoate protein ligase LplAl
  • L. monocytogenes DP-L4017 10403S with LLO L461T
  • L. monocytogenes EGD see, e.g., GenBank Accession No. AL591824.
  • the Listeria strain is L. monocytogenes EGD-e (see GenBank Accession No.
  • L. monocytogenes DP-L4029 actA deletion, optionally in combination with uvrAB deletion (DP-L4029uvrAB) (see, e.g., US 7,691,393)
  • L. monocytogenes actA- linlB - double mutant see, e.g., ATCC Accession No. PTA-5562
  • L. monocytogenes IplA mutant or hly mutant see, e.g., US 2004/0013690
  • L. monocytogenes dalldat double mutant see, e.g., US 2005/0048081.
  • monocytogenes strains includes those that are modified (e.g., by a plasmid and/or by genomic integration) to contain a nucleic acid encoding one of, or any combination of, the following genes: hly (LLO; listeriolysin); iap (p60); inlA; inlB; inlC; dal (alanine racemase); dat (D-amino acid aminotransferase); plcA; plcB; actA; or any nucleic acid that mediates growth, spread, breakdown of a single walled vesicle, breakdown of a double walled vesicle, binding to a host cell, or uptake by a host cell.
  • the recombinant bacteria or Listeria can have wild-type virulence, can have attenuated virulence, or can be avirulent.
  • a recombinant Listeria of can be sufficiently virulent to escape the phagosome or phagolysosome and enter the cytosol.
  • Such Listeria strains can also be live-attenuated Listeria strains, which comprise at least one attenuating mutation, deletion, or inactivation as disclosed elsewhere herein.
  • the recombinant Listeria is an attenuated auxotrophic strain.
  • An auxotrophic strain is one that is unable to synthesize a particular organic compound required for its growth. Examples of such strains are described in US 8,114,414, herein incorporated by reference in its entirety for all purposes.
  • the recombinant Listeria strain lacks antibiotic resistance genes.
  • such recombinant Listeria strains can comprise a plasmid that does not encode an antibiotic resistance gene.
  • some recombinant Listeria strains provided herein comprise a plasmid comprising a nucleic acid encoding an antibiotic resistance gene.
  • Antibiotic resistance genes may be used in the conventional selection and cloning processes commonly employed in molecular biology and vaccine preparation. Exemplary antibiotic resistance genes include gene products that confer resistance to ampicillin, penicillin, methicillin, streptomycin, erythromycin, kanamycin, tetracycline, chloramphenicol (CAT), neomycin, hygromycin, and gentamicin.
  • CAT chloramphenicol
  • the recombinant bacterial strains (e.g., Listeria strains) disclosed herein comprise a recombinant fusion polypeptide disclosed herein or a nucleic acid encoding the recombinant fusion polypeptide as disclosed elsewhere herein.
  • nucleic acid in bacteria or Listeria strains comprising a nucleic acid encoding a recombinant fusion protein, the nucleic acid can be codon optimized. Examples of optimal codons utilized by L. monocytogenes for each amino acid are shown US 2007/0207170, herein incorporated by reference in its entirety for all purposes. A nucleic acid is codon- optimized if at least one codon in the nucleic acid is replaced with a codon that is more frequently used by L. monocytogenes for that amino acid than the codon in the original sequence.
  • the nucleic acid can be present in an episomal plasmid within the bacteria or Listeria strain and/or the nucleic acid can be genomically integrated in the bacteria or Listeria strain.
  • Some recombinant bacteria or Listeria strains comprise two separate nucleic acids encoding two recombinant fusion polypeptides as disclosed herein: one nucleic acid in an episomal plasmid, and one genomically integrated in the bacteria or Listeria strain.
  • the episomal plasmid can be one that is stably maintained in vitro (in cell culture), in vivo (in a host), or both in vitro and in vivo. If in an episomal plasmid, the open reading frame encoding the recombinant fusion polypeptide can be operably linked to a promoter/regulatory sequence in the plasmid. If genomically integrated in the bacteria or Listeria strain, the open reading frame encoding the recombinant fusion polypeptide can be operably linked to an exogenous promoter/regulatory sequence or to an endogenous promoter/regulatory sequence.
  • promoters/regulatory sequences useful for driving constitutive expression of a gene include, for example, an hly, hlyA, actA, prfA, and p60 promoters of Listeria, the Streptococcus bac promoter, the Streptomyces griseus sgiA promoter, and the B. thuringiensis phaZ promoter.
  • an inserted gene of interest is not interrupted or subjected to regulatory constraints which often occur from integration into genomic DNA, and in some cases, the presence of the inserted heterologous gene does not lead to rearrangement or interruption of the cell's own important regions.
  • Such recombinant bacteria or Listeria strains can be made by transforming a bacteria or Listeria strain or an attenuated bacteria or Listeria strain described elsewhere herein with a plasmid or vector comprising a nucleic acid encoding the recombinant fusion polypeptide.
  • the plasmid can be an episomal plasmid that does not integrate into a host chromosome.
  • the plasmid can be an integrative plasmid that integrates into a chromosome of the bacteria or Listeria strain.
  • the plasmids used herein can also be multicopy plasmids.
  • Methods for transforming bacteria include calcium-chloride competent cell-based methods, electroporation methods, bacteriophage- mediated transduction, chemical transformation techniques, and physical transformation techniques. See, e.g., de Boer et al. (1989) Cell 56:641-649; Miller et al. (1995) FASEB J. 9: 190-199; Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York; Ausubel et al. (1997) Current Protocols in Molecular
  • Bacteria or Listeria strains with genomically integrated heterologous nucleic acids can be made, for example, by using a site-specific integration vector, whereby the bacteria or Listeria comprising the integrated gene is created using homologous recombination.
  • the integration vector can be any site- specific integration vector that is capable of infecting a bacteria or Listeria strain.
  • Such an integration vector can comprise, for example, a PSA attPP' site, a gene encoding a PSA integrase, a U153 attPP' site, a gene encoding a U153 integrase, an A118 attPP' site, a gene encoding an A118 integrase, or any other known attPP' site or any other phage integrase.
  • Such bacteria or Listeria strains comprising an integrated gene can also be created using any other known method for integrating a heterologous nucleic acid into a bacteria or Listeria chromosome. Techniques for homologous recombination are well known, and are described, for example, in Baloglu et al. (2005) Vet Microbiol 109(1- 2): 11-17); Jiang et al. 2005) Acta Biochim Biophys Sin (Shanghai) 37(l): 19-24), and US 6,855,320, each of which is herein incorporated by reference in its entirety for all purposes.
  • transposon insertion Techniques for transposon insertion are well known, and are described, for example, for the construction of DP-L967 by Sun et al. (1990) Infection and Immunity 58: 3770-3778, herein incorporated by reference in its entirety for all purposes.
  • Transposon mutagenesis can achieve stable genomic insertion, but the position in the genome where the heterologous nucleic acids has been inserted is unknown.
  • Integration into a bacterial or Listerial chromosome can also be achieved using phage integration sites (see, e.g., Lauer et al. (2002) J Bacteriol 184(15):4177-4186, herein incorporated by reference in its entirety for all purposes).
  • phage integration sites see, e.g., Lauer et al. (2002) J Bacteriol 184(15):4177-4186, herein incorporated by reference in its entirety for all purposes.
  • an integrase gene and attachment site of a bacteriophage e.g., U153 or PSA listeriophage
  • a 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 can be cured from the utilized attachment site prior to integration of the heterologous nucleic acid.
  • Such methods can result, for example, in single-copy integrants.
  • a phage integration system based on PSA phage can be used (see, e.g., Lauer et al. (2002) J Bacteriol 184:4177-4186, herein incorporated by reference in its entirety for all purposes). Maintaining the integrated gene can require, for example, continuous selection by antibiotics.
  • a phage-based chromosomal integration system can be established that does not require selection with antibiotics.
  • an auxotrophic host strain can be complemented.
  • a phage-based chromosomal integration system for clinical applications can be used, where a host strain that is auxotrophic for essential enzymes, including, for example, D-alanine racemase is used (e.g., Lm dal(-)dat(-)).
  • Conjugation can also be used to introduce genetic material and/or plasmids into bacteria. Methods for conjugation are well known, and are described, for example, in Nikodinovic et al. (2006) Plasmid 56(3):223-227 and Auchtung et al. (2005) Proc Natl Acad Sci USA 102(35): 12554- 12559, each of which is herein incorporated by reference in its entirety for all purposes.
  • a recombinant bacteria or Listeria strain can comprise a nucleic acid encoding a recombinant fusion polypeptide genomically integrated into the bacteria or Listeria genome as an open reading frame with an endogenous actA sequence (encoding an ActA protein) or an endogenous hly sequence (encoding an LLO protein).
  • an endogenous actA sequence encoding an ActA protein
  • an endogenous hly sequence encoding an LLO protein
  • the expression and secretion of the fusion polypeptide can be under the control of the endogenous actA promoter and ActA signal sequence or can be under the control of the endogenous hly promoter and LLO signal sequence.
  • the nucleic acid encoding a recombinant fusion polypeptide can replace an actA sequence encoding an ActA protein or an hly sequence encoding an LLO protein.
  • Selection of recombinant bacteria or Listeria strains can be achieved by any means.
  • antibiotic selection can be used.
  • Antibiotic resistance genes may be used in the conventional selection and cloning processes commonly employed in molecular biology and vaccine preparation.
  • Exemplary antibiotic resistance genes include gene products that confer resistance to ampicillin, penicillin, methicillin, streptomycin, erythromycin, kanamycin, tetracycline, chloramphenicol (CAT), neomycin, hygromycin, and gentamicin.
  • auxotrophic strains can be used, and an exogenous metabolic gene can be used for selection instead of or in addition to an antibiotic resistance gene.
  • an exogenous metabolic gene can be used for selection instead of or in addition to an antibiotic resistance gene.
  • transformed auxotrophic bacteria can be grown in a medium that will select for expression of the gene encoding the metabolic enzyme (e.g., amino acid metabolism gene) or the complementing gene.
  • a temperature- sensitive plasmid can be used to select recombinants or any other known means for selecting recombinants.
  • the recombinant bacteria strains e.g., recombinant Listeria strains disclosed herein can be attenuated.
  • the term "attenuation" encompasses a diminution in the ability of the bacterium to cause disease in a host animal.
  • the pathogenic characteristics of an attenuated Listeria strain may be 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 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 that does not kill an animal to which it is administered, or is one that 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. Attenuated strains are
  • Attenuation can be accomplished by any known means.
  • such attenuated strains can be deficient in one or more endogenous virulence genes or one or more endogenous metabolic genes.
  • examples of such genes are disclosed herein, and attenuation can be achieved by inactivation of any one of or any combination of the genes disclosed herein. Inactivation can be achieved, for example, through deletion or through mutation (e.g., an inactivating mutation).
  • mutation includes any type of mutation or modification to the sequence (nucleic acid or amino acid sequence) and may encompass a deletion, a truncation, an insertion, a substitution, a disruption, or a translocation.
  • a mutation can include a frameshift mutation, a mutation which causes premature termination of a protein, or a 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 may be preferred because of the accompanying low probability of reversion.
  • the term "metabolic gene” refers to a gene encoding an enzyme involved in or required for synthesis of a nutrient utilized or required by a host bacteria. For example, the enzyme can be involved in or required for the synthesis of a nutrient required for sustained growth of the host bacteria.
  • viralence gene includes a gene whose presence or activity in an organism's genome that contributes to the pathogenicity of the organism (e.g., enabling the organism to achieve colonization of a niche in the host (including attachment to cells), immunoevasion
  • LmddA Listeria monocytogenes
  • LmddA Lm dal(-)dat(-) actA
  • LmddA Lm dal(-)dat(-) actA
  • Lm prfA ⁇ - Another specific example of an attenuated strain is Lm prfA ⁇ - or a strain having a partial deletion or inactivating mutation in the prfA gene.
  • the PrfA protein controls the expression of a regulon comprising essential virulence genes required by Lm to colonize its vertebrate hosts; hence the prfA mutation strongly impairs PrfA ability to activate expression of Prf A-dependent virulence genes.
  • Attenuated bacteria or Listeria strains include bacteria or Listeria strains deficient in one or more endogenous virulence genes. Examples of such genes include actA, prfA, plcB, plcA, inlA, inlB, inlC, inlJ, and bsh in Listeria. Attenuated Listeria strains can also be the double mutant or triple mutant of any of the above- mentioned strains. Attenuated Listeria strains can comprise a mutation or deletion of each one of the genes, or comprise a mutation or deletion of, for example, up to ten of any of the genes provided herein (e.g., including the actA, prfA, and dal/dat genes). For example, an attenuated Listeria strain can comprise a mutation or deletion of an endogenous internalin C (inlC) gene and/or a mutation or deletion of an endogenous actA gene.
  • inlC endogenous internalin C
  • an attenuated Listeria strain can comprise a mutation or deletion of an endogenous internalin B (inlB) gene and/or a mutation or deletion of an endogenous actA gene.
  • an attenuated Listeria strain can comprise a mutation or deletion of endogenous inlB, inlC, and actA genes. Translocation of Listeria to adjacent cells is inhibited by the deletion of the endogenous actA gene and/or the endogenous inlC gene or endogenous inlB gene, which are involved in the process, thereby resulting in high levels of attenuation with increased immunogenicity and utility as a strain backbone.
  • An attenuated Listeria strain can also be a double mutant comprising mutations or deletions of both plcA and plcB. In some cases, the strain can be constructed from the EGD Listeria backbone.
  • a bacteria or Listeria strain can also be an auxotrophic strain having a mutation in a metabolic gene.
  • the strain can be deficient in one or more endogenous amino acid metabolism genes.
  • the generation of auxotrophic strains of Listeria deficient in D-alanine may be accomplished in a number of ways that are well known, including deletion mutations, insertion mutations, frameshift mutations, mutations which cause premature termination of a protein, or mutation of regulatory sequences which affect gene expression. Deletion mutants may be 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 can be selected.
  • Examples of endogenous amino acid metabolism genes include a vitamin synthesis gene, a gene encoding pantothenic acid synthase, a D-glutamic acid synthase gene, a D-alanine amino transferase (dat) gene, a D-alanine racemase (dal) gene, dga, a gene involved in the synthesis of diaminopimelic acid (DAP), a gene involved in the synthesis of Cysteine synthase A (cysK), a vitamin-B 12 independent methionine synthase, trpA, trpB, trpE, astiB, gltD, gltB, leuA, argG, and thrC.
  • a vitamin synthesis gene a gene encoding pantothenic acid synthase, a D-glutamic acid synthase gene, a D-alanine amino transferase (dat) gene, a D-
  • the Listeria strain can be deficient in two or more such genes (e.g., dat and dal). D-glutamic acid synthesis is controlled in part by the dal gene, which is involved in the conversion of D-glu + pyr to alpha-ketoglutarate + D-ala, and the reverse reaction.
  • an attenuated Listeria strain can be deficient in an endogenous synthase gene, such as an amino acid synthesis gene.
  • endogenous synthase gene such as an amino acid synthesis gene.
  • genes include folP, a gene encoding a dihydro uridine synthase family protein, ispD, ispF, a gene encoding a phosphoenolpyruvate synthase, hisF, hisH,fliI, a gene encoding a ribosomal large subunit pseudouridine synthase, ispD, a gene encoding a bifunctional GMP synthase/glutamine amidotransferase protein, cobS, cobB, cbiD, a gene encoding a uroporphyrin-III C-methyltransferase/uroporphyrinogen-III synthase, cobQ, uppS, truB, dxs, mvaS,
  • Attenuated Listeria strains can be deficient in endogenous phoP, aroA, aroC, aroD, or plcB.
  • an attenuated Listeria strain can be deficient in an endogenous peptide transporter. Examples include genes encoding an ABC transporter/ ATP-binding/permease protein, an oligopeptide ABC transporter/oligopeptide- binding protein, an oligopeptide ABC transporter/permease protein, a zinc ABC
  • transporter/zinc-binding protein a sugar ABC transporter, a phosphate transporter, a ZIP zinc transporter, a drug resistance transporter of the EmrBIQacA family, a sulfate transporter, a proton-dependent oligopeptide transporter, a magnesium transporter, a formate/nitrite transporter, a spermidine/putrescine ABC transporter, a Na/Pi- cotransporter, a sugar phosphate transporter, a glutamine ABC transporter, a major facilitator family transporter, a glycine betaine/L-proline ABC transporter, a molybdenum ABC transporter, a techoic acid ABC transporter, a cobalt ABC transporter, an ammonium transporter, an amino acid ABC transporter, a cell division ABC transporter, a manganese ABC transporter, an iron compound ABC transporter, a maltose/maltodextrin ABC transporter, a drug resistance transporter of the Bcrl
  • Attenuated bacteria and Listeria strains can be deficient in an endogenous metabolic enzyme that metabolizes an amino acid that is used for a bacterial growth process, a replication process, cell wall synthesis, protein synthesis, metabolism of a fatty acid, or for any other growth or replication process.
  • an attenuated strain can be deficient in an endogenous metabolic enzyme that can catalyze the formation of an amino acid used in cell wall synthesis, can catalyze the synthesis of an amino acid used in cell wall synthesis, or can be involved in synthesis of an amino acid used in cell wall synthesis.
  • the amino acid can be used in cell wall biogenesis.
  • the metabolic enzyme is a synthetic enzyme for D-glutamic acid, a cell wall component.
  • Attenuated Listeria strains can be deficient in metabolic enzymes encoded by a D-glutamic acid synthesis gene, dga, an air (alanine racemase) gene, or any other enzymes that are involved in alanine synthesis.
  • metabolic enzymes for which the Listeria strain can be deficient include enzymes encoded by serC (a phospho serine aminotransferase), asd (aspartate betasemialdehyde dehydrogenase; involved in synthesis of the cell wall constituent diaminopimelic acid), the gene encoding gsaB- glutamate-l-semialdehyde aminotransferase (catalyzes the formation of 5- aminolevulinate from (S)-4-amino-5-oxopentanoate), hemL (catalyzes the formation of 5- aminolevulinate from (S)-4-amino-5-oxopentanoate), aspB (an aspartate aminotransferase that catalyzes the formation of oxalozcetate and L-glutamate from L-aspartate and 2- oxoglutarate), argF-1 (involved in arginine biosynthesis), aroE (involved involved
  • An attenuated Listeria strain can be generated by mutation of other metabolic enzymes, such as a tRNA synthetase.
  • the metabolic enzyme can be encoded by the trpS gene, encoding tryptophanyltRNA synthetase.
  • the host strain bacteria can be A(trpS aroA), and both markers can be contained in an integration vector.
  • metabolic enzymes include aspartate aminotransferase, histidinol-phosphate aminotransferase (GenBank Accession No. NP_466347), or the cell wall teichoic acid glycosylation protein GtcA.
  • the component can be, for example, UDP-N-acetylmuramylpentapeptide, UDP-N-acetylglucosamine, MurNAc-(pentapeptide)-pyrophosphoryl-undecaprenol, GlcNAc-p-(l,4)-MurNAc-(pentapeptide)-pyrophosphorylundecaprenol, or any other peptidoglycan component or precursor.
  • the metabolic enzyme can be any other synthetic enzyme for a peptidoglycan component or precursor.
  • the metabolic enzyme can also be a trans-glycosylase, a trans-peptidase, a carboxy-peptidase, any other class of metabolic enzyme, or any other metabolic enzyme.
  • the metabolic enzyme can be any other Listeria metabolic enzyme or any other Listeria monocytogenes metabolic enzyme.
  • the attenuated bacteria or Listeria strains disclosed herein can further comprise a nucleic acid comprising a complementing gene or encoding a metabolic enzyme that complements an attenuating mutation (e.g., complements the auxotrophy of the auxotrophic Listeria strain).
  • a nucleic acid having a first open reading frame encoding a fusion polypeptide as disclosed herein can further comprise a second open reading frame comprising the complementing gene or encoding the complementing metabolic enzyme.
  • a first nucleic acid can encode the fusion polypeptide and a separate second nucleic acid can comprise the complementing gene or encode the complementing metabolic enzyme.
  • the complementing gene can be extrachromosomal or can be integrated into the bacteria or Listeria genome.
  • the auxotrophic Listeria strain can comprise an episomal plasmid comprising a nucleic acid encoding a metabolic enzyme. Such plasmids will be contained in the Listeria in an episomal or extrachromosomal fashion.
  • the auxotrophic Listeria strain can comprise an integrative plasmid (i.e., integration vector) comprising a nucleic acid encoding a metabolic enzyme.
  • integrative plasmids can be used for integration into a Listeria chromosome.
  • the episomal plasmid or the integrative plasmid lacks an antibiotic resistance marker.
  • the metabolic gene can be used for selection instead of or in addition to an antibiotic resistance gene.
  • transformed auxotrophic bacteria in order to select for auxotrophic bacteria comprising a plasmid encoding a metabolic enzyme or a complementing gene provided herein, transformed auxotrophic bacteria can be grown in a medium that will select for expression of the gene encoding the metabolic enzyme (e.g., amino acid metabolism gene) or the complementing gene.
  • a bacteria auxotrophic for D-glutamic acid synthesis can be 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 a plasmid comprising a 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 and are available commercially.
  • the bacteria can be propagated in the presence of a selective pressure. Such propagation can comprise growing the bacteria in media without the auxotrophic factor.
  • the presence of the plasmid expressing the metabolic enzyme or the complementing gene in the auxotrophic bacteria ensures that the plasmid will replicate along with the bacteria, thus continually selecting for bacteria harboring the plasmid.
  • Production of the bacteria or Listeria strain can be readily scaled up by adjusting the volume of the medium in which the auxotrophic bacteria comprising the plasmid are growing.
  • the attenuated strain is a strain having a deletion of or an inactivating mutation in dal and dat (e.g., Listeria monocytogenes (Lm) dal ⁇ -)dat ⁇ -) (Lmdd) or Lm dal(-)dat(-) actA (LmddA)), and the complementing gene encodes an alanine racemase enzyme (e.g., encoded by dal gene) or a D-amino acid aminotransferase enzyme (e.g., encoded by dat gene).
  • dal and dat e.g., Listeria monocytogenes (Lm) dal ⁇ -)dat ⁇ -) (Lmdd) or Lm dal(-)dat(-) actA (LmddA)
  • the complementing gene encodes an alanine racemase enzyme (e.g., encoded by dal gene) or a D-amino acid aminotransferase enzyme
  • An exemplary alanine racemase protein can have the sequence set forth in SEQ ID NO: 76 (encoded by SEQ ID NO: 78; GenBank Accession No: AF038438) or can be a homologue, variant, isoform, analog, fragment, fragment of a homologue, fragment of a variant, fragment of an analog, or fragment of an isoform of SEQ ID NO: 76 .
  • the alanine racemase protein can also be any other Listeria alanine racemase protein.
  • the alanine racemase protein can be any other gram- positive alanine racemase protein or any other alanine racemase protein.
  • An exemplary D- amino acid aminotransferase protein can have the sequence set forth in SEQ ID NO: 77 (encoded by SEQ ID NO: 79; GenBank Accession No: AF038439) or can be a
  • the D- amino acid aminotransferase protein can also be any other Listeria D-amino acid aminotransferase protein.
  • the D-amino acid aminotransferase protein can be any other gram-positive D-amino acid aminotransferase protein or any other D-amino acid aminotransferase protein.
  • the attenuated strain is a strain having a deletion of or an inactivating mutation in prfA (e.g., Lm prfA(-)), and the complementing gene encodes a PrfA protein.
  • the complementing gene can encode a mutant PrfA (D133V) protein that restores partial PrfA function.
  • SEQ ID NO: 80 encoded by nucleic acid set forth in SEQ ID NO: 81
  • SEQ ID NO: 82 an example of a D133V mutant PrfA protein is set forth in SEQ ID NO: 82 (encoded by nucleic acid set forth in SEQ ID NO: 83).
  • the complementing PrfA protein can be a homologue, variant, isoform, analog, fragment, fragment of a homologue, fragment of a variant, fragment of an analog, or fragment of an isoform of SEQ ID NO: 80 or 82.
  • the PrfA protein can also be any other Listeria PrfA protein.
  • the PrfA protein can be any other gram-positive PrfA protein or any other PrfA protein.
  • the bacteria strain or Listeria strain can comprise a deletion of or an inactivating mutation in an actA gene, and the complementing gene can comprise an actA gene to complement the mutation and restore function to the Listeria strain.
  • auxotroph strains and complementation systems can also be adopted for the use with the methods and compositions provided herein.
  • the recombinant bacteria strain (e.g., Listeria strain) optionally has been passaged through an animal host.
  • Such passaging can maximize efficacy of the Listeria strain as a vaccine vector, can stabilize the immunogenicity of the Listeria strain, can stabilize the virulence of the Listeria strain, can increase the immunogenicity of the Listeria strain, can increase the virulence of the Listeria strain, can remove unstable sub- strains of the Listeria strain, or can reduce the prevalence of unstable sub- strains of the Listeria strain.
  • Methods for passaging a recombinant Listeria strain through an animal host are well known in the art and are described, for example, in US 2006/0233835, herein incorporated by reference in its entirety for all purposes.
  • the recombinant bacteria strain can be stored in a frozen cell bank or stored in a lyophilized cell bank.
  • a cell bank can be, for example, a master cell bank, a working cell bank, or a Good Manufacturing Practice (GMP) cell bank.
  • GMP Good Manufacturing Practice
  • Examples of "Good Manufacturing Practices” include those defined by 21 CFR 210-211 of the United States Code of Federal Regulations. However, “Good Manufacturing Practices” can also be defined by other standards for production of clinical-grade material or for human consumption, such as standards of a country other than the United States.
  • Such cell banks can be intended for production of clinical-grade material or can conform to regulatory practices for human use.
  • Recombinant bacteria strains can also be from a batch of vaccine doses, from a frozen stock, or from a lyophilized stock.
  • Such cell banks, frozen stocks, or batches of vaccine doses can, for example, exhibit viability upon thawing of greater than 90%.
  • the thawing for example, can follow storage for cryopreservation or frozen storage for 24 hours.
  • the storage can last, for example, for 2 days, 3 days, 4 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 5 months, 6 months, 9 months, or 1 year.
  • the cell bank, frozen stock, or batch of vaccine doses can be cryopreserved, for example, by a method that comprises growing a culture of the bacteria strain (e.g., Listeria strain) in a nutrient media, freezing the culture in a solution comprising glycerol, and storing the Listeria strain at below -20°C.
  • the temperature can be, for example, about - 70°C or between about -70 to about -80°C.
  • the cell bank, frozen stock, or batch of vaccine doses can be cryopreserved by a method that comprises growing a culture of the Listeria strain in a defined medium, freezing the culture in a solution comprising glycerol, and storing the Listeria strain at below -20°C.
  • the temperature can be, for example, about -70°C or between about -70 to about -80°C. Any defined microbiological medium may be used in this method.
  • the culture e.g., the culture of a Listeria vaccine strain that is used to produce a batch of Listeria vaccine doses
  • the culture can be inoculated, for example, from a cell bank, from a frozen stock, from a starter culture, or from a colony.
  • the culture can be inoculated, for example, at mid-log growth phase, at approximately mid-log growth phase, or at another growth phase.
  • the solution used for freezing optionally contain another colligative additive or additive with anti- freeze properties in place of glycerol or in addition to glycerol.
  • additives include, for example, mannitol, DMSO, sucrose, or any other colligative additive or additive with anti-freeze properties.
  • the nutrient medium utilized for growing a culture of a bacteria strain can be any suitable nutrient medium.
  • suitable media include, for example, LB; TB; a modified, animal-product-free Terrific Broth; or a defined medium.
  • the step of growing can be performed by any known means of growing bacteria.
  • the step of growing can be performed with a shake flask (such as a baffled shake flask), a batch fermenter, a stirred tank or flask, an airlift fermenter, a fed batch, a continuous cell reactor, an immobilized cell reactor, or any other means of growing bacteria.
  • a constant pH is maintained during growth of the culture (e.g. in a batch fermenter).
  • the pH can be maintained at about 6.0, at about 6.5, at about 7.0, at about 7.5, or about 8.0.
  • the pH can be, for example, from about 6.5 to about 7.5, from about 6.0 to about 8.0, from about 6.0 to about 7.0, from about 6.0 to about 7.0, or from about 6.5 to about 7.5.
  • a constant temperature can be maintained during growth of the culture.
  • the temperature can be maintained at about 37°C or at 37°C.
  • the temperature can be maintained at 25°C, 27°C, 28°C, 30°C, 32°C, 34°C, 35°C, 36°C, 38°C, or 39°C.
  • a constant dissolved oxygen concentration can be maintained during growth of the culture.
  • the dissolved oxygen concentration can be maintained at 20% of saturation, 15% of saturation, 16% of saturation, 18% of saturation, 22% of saturation, 25% of saturation, 30% of saturation, 35% of saturation, 40% of saturation, 45% of saturation, 50% of saturation, 55% of saturation, 60% of saturation, 65% of saturation, 70% of saturation, 75% of saturation, 80% of saturation, 85% of saturation, 90% of saturation, 95% of saturation, 100% of saturation, or near 100% of saturation.
  • Methods for lyophilization and cryopreservation of recombinant bacteria strains are known.
  • a Listeria culture can be flash- frozen in liquid nitrogen, followed by storage at the final freezing temperature.
  • the culture can be frozen in a more gradual manner (e.g., by placing in a vial of the culture in the final storage temperature).
  • the culture can also be frozen by any other known method for freezing a bacterial culture.
  • the storage temperature of the culture can be, for example, between -20 and - 80°C.
  • the temperature can be significantly below -20°C or not warmer than -70°C.
  • the temperature can be about -70°C, -20°C, -30°C, -40°C, -50°C, - 60°C, -80°C, -30 to -70°C, -40 to -70°C, -50 to -70°C, -60 to -70°C, -30 to -80°C, -40 to - 80°C, -50 to -80°C, -60 to -80°C, or -70 to -80°C.
  • the temperature can be colder than 70°C or colder than -80°C.
  • immunogenic compositions comprising a recombinant fusion polypeptide as disclosed herein, a nucleic acid encoding a recombinant fusion polypeptide as disclosed herein, or a recombinant bacteria or Listeria strain as disclosed herein.
  • An immunogenic composition comprising a Listeria strain can be inherently immunogenic by virtue of its comprising a Listeria strain and/or the composition can also further comprise an adjuvant.
  • Other immunogenic compositions comprise DNA immunotherapy or peptide immunotherapy compositions.
  • immunogenic composition refers to any composition containing an antigen that elicits an immune response against the antigen in a subject upon exposure to the composition.
  • the immune response elicited by an immunogenic composition can be to a particular antigen or to a particular epitope on the antigen.
  • An immunogenic composition can comprise a single recombinant fusion polypeptide as disclosed herein, nucleic acid encoding a recombinant fusion polypeptide as disclosed herein, or recombinant bacteria or Listeria strain as disclosed herein, or it can comprise multiple different recombinant fusion polypeptides as disclosed herein, nucleic acids encoding recombinant fusion polypeptides as disclosed herein, or recombinant bacteria or Listeria strains as disclosed herein.
  • a first recombinant fusion polypeptide is different from a second recombinant fusion polypeptide, for example, if it includes one antigenic peptide that the second recombinant fusion polypeptide does not.
  • the two recombinant fusion polypeptides can include some of the same antigenic peptides and still be considered different.
  • Such different recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, or recombinant bacteria or Listeria strains can be administered concomitantly to a subject or sequentially to a subject.
  • Sequential administration can be particularly useful when a drug substance comprising a recombinant Listeria strain (or recombinant fusion polypeptide or nucleic acid) disclosed herein is in different dosage forms (e.g., one agent is a tablet or capsule and another agent is a sterile liquid) and/or is administered on different dosing schedules (e.g., one composition from the mixture is administered at least daily and another is administered less frequently, such as once weekly, once every two weeks, or once every three weeks).
  • the multiple recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, or recombinant bacteria or Listeria strains can each comprise a different set of antigenic peptides.
  • two or more of the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, or recombinant bacteria or Listeria strains can comprise the same set of antigenic peptides (e.g., the same set of antigenic peptides in a different order).
  • An immunogenic composition can additionally comprise an adjuvant (e.g., two or more adjuvants), a cytokine, a chemokine, or combination thereof.
  • an immunogenic composition can additionally comprises antigen presenting cells (APCs), which can be autologous or can be allogeneic to the subject.
  • APCs antigen presenting cells
  • an adjuvant includes compounds or mixtures that enhance the immune response to an antigen.
  • an adjuvant can be a non-specific stimulator of an immune response or substances that allow generation of a depot in a subject which when combined with an immunogenic composition disclosed herein provides for an even more enhanced and/or prolonged immune response.
  • An adjuvant can favor, for example, a predominantly Thl-mediated immune response, a Thl-type immune response, or a Thl- mediated immune response.
  • an adjuvant can favor a cell- mediated immune response over an antibody-mediated response.
  • an adjuvant can favor an antibody-mediated response.
  • Some adjuvants can enhance the immune response by slowly releasing the antigen, while other adjuvants can mediate their effects by any of the following mechanisms: increasing cellular infiltration, inflammation, and trafficking to the injection site, particularly for antigen-presenting cells (APC); promoting the activation state of APCs by upregulating costimulatory signals or major histocompatibility complex (MHC) expression; enhancing antigen presentation; or inducing cytokine release for indirect effect.
  • APC antigen-presenting cells
  • MHC major histocompatibility complex
  • adjuvants include saponin QS21, CpG oligonucleotides, unmethylated CpG-containing oligonucleotides, MPL, TLR agonists, TLR4 agonists,
  • TLR9 agonists Resiquimod ® , imiquimod, cytokines or nucleic acids encoding the same, chemokines or nucleic acids encoding same, IL-12 or a nucleic acid encoding the same, IL-6 or a nucleic acid encoding the same, and lipopolysaccharides.
  • a suitable adjuvant is Montanide ISA 51. Montanide ISA 51 contains a natural
  • dtLLO listeriolysin O
  • SEQ ID NO: 115 A dtLLO encoded by a sequence at least 90%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 115 is also suitable for use as an adjuvant.
  • a suitable adjuvant examples include granulocyte/macrophage colony- stimulating factor (GM-CSF) or a nucleic acid encoding the same and keyhole limpet hemocyanin (KLH) proteins or nucleic acids encoding the same.
  • the GM-CSF can be, for example, a human protein grown in a yeast (S. cerevisiae) vector.
  • GM-CSF promotes clonal expansion and differentiation of hematopoietic progenitor cells, antigen presenting cells (APCs), dendritic cells, and T cells.
  • adjuvants include growth factors or nucleic acids encoding the same, cell populations, Freund's incomplete adjuvant, aluminum phosphate, aluminum hydroxide, BCG (bacille Calmette-Guerin), alum, interleukins or nucleic acids encoding the same, quill glycosides, monophosphoryl lipid A, liposomes, bacterial mitogens, bacterial toxins, or any other type of known adjuvant (see, e.g., Fundamental Immunology, 5th ed. (March 2003): William E. Paul (Editor); Lippincott Williams & Wilkins Publishers; Chapter 43: Vaccines, GJV Nossal, which is herein incorporated by reference in its entirety for all purposes).
  • An immunogenic composition can further comprise one or more
  • immunomodulatory molecules examples include interferon gamma, a cytokine, a chemokine, and a T bell stimulant.
  • An immunogenic composition can be in the form of a vaccine or
  • a vaccine may be, for example, a peptide vaccine (e.g., comprising a recombinant fusion polypeptide as disclosed herein), a DNA vaccine (e.g., comprising a nucleic acid encoding a recombinant fusion
  • polypeptide as disclosed herein may prevent a subject from contracting or developing a disease or condition and/or a vaccine may be therapeutic to a subject having a disease or condition.
  • Methods for preparing peptide vaccines are well known and are described, for example, in EP 1408048, US 2007/0154953, and Ogasawara et al. (1992) Proc. Natl Acad Sci USA 89:8995-8999, each of which is herein incorporated by reference in its entirety for all purposes.
  • peptide evolution techniques can be used to create an antigen with higher immunogenicity. Techniques for peptide evolution are well known and are described, for example, in US 6,773,900, herein incorporated by reference in its entirety for all purposes.
  • a "pharmaceutically acceptable carrier” refers to a vehicle for containing an immunogenic composition that can be introduced into a subject without significant adverse effects and without having deleterious effects on the immunogenic composition. That is, “pharmaceutically acceptable” refers to any formulation which is safe, and provides the appropriate delivery for the desired route of administration of an effective amount of at least one immunogenic composition for use in the methods disclosed herein. Pharmaceutically acceptable carriers or vehicles or excipients are well known.
  • Such carriers can be suitable for any route of administration (e.g., parenteral, enteral (e.g., oral), or topical application).
  • Such pharmaceutical compositions can be buffered, for example, wherein the pH is maintained at a particular desired value, ranging from pH 4.0 to pH 9.0, in accordance with the stability of the immunogenic compositions and route of administration.
  • Suitable pharmaceutically acceptable carriers include, for example, sterile water, salt solutions such as saline, glucose, buffered solutions such as phosphate buffered solutions or bicarbonate buffered solutions, alcohols, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatine, carbohydrates (e.g., lactose, amylose or starch), magnesium stearate, talc, silicic acid, viscous paraffin, white paraffin, glycerol, alginates, hyaluronic acid, collagen, perfume oil, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, hydroxy methylcellulose, polyvinyl pyrrolidone, and the like.
  • compositions or vaccines may also include auxiliary agents including, for example, diluents, stabilizers (e.g., sugars and amino acids), preservatives, wetting agents, emulsifiers, pH buffering agents, viscosity enhancing additives, lubricants, salts for influencing osmotic pressure, buffers, vitamins, coloring, flavoring, aromatic substances, and the like which do not deleteriously react with the immunogenic composition.
  • auxiliary agents including, for example, diluents, stabilizers (e.g., sugars and amino acids), preservatives, wetting agents, emulsifiers, pH buffering agents, viscosity enhancing additives, lubricants, salts for influencing osmotic pressure, buffers, vitamins, coloring, flavoring, aromatic substances, and the like which do not deleteriously react with the immunogenic composition.
  • pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, emulsions, or oils.
  • Non-aqueous solvents include, for example, propylene glycol, polyethylene glycol, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include, for example, water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • oils include those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, olive oil, sunflower oil, and fish- liver oil.
  • Solid carriers/diluents include, for example, a gum, a starch (e.g., corn starch, pregeletanized starch), a sugar (e.g., lactose, mannitol, sucrose, or dextrose), a cellulosic material (e.g., microcrystalline cellulose), an acrylate (e.g., polymethylacrylate), calcium carbonate, magnesium oxide, talc, or mixtures thereof.
  • a gum e.g., corn starch, pregeletanized starch
  • a sugar e.g., lactose, mannitol, sucrose, or dextrose
  • a cellulosic material e.g., microcrystalline cellulose
  • an acrylate e.g., polymethylacrylate
  • calcium carbonate e.g., magnesium oxide, talc, or mixtures thereof.
  • sustained or directed release pharmaceutical compositions or vaccines can be formulated. This can be accomplished, for example, through use of liposomes or compositions wherein the active compound is protected with differentially degradable coatings (e.g., by microencapsulation, multiple coatings, and so forth). Such compositions may be formulated for immediate or slow release. It is also possible to freeze-dry the compositions and use the lyophilisates obtained (e.g., for the preparation of products for injection).
  • An immunogenic composition, pharmaceutical composition, or vaccine disclosed herein may also comprise one or more additional compounds effective in preventing or treating cancer.
  • the additional compound may comprise a compound useful in chemotherapy, such as amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clofarabine, crisantaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, docetaxel, doxorubicin, epirubicin, etoposide, fludarabine, fluorouracil (5-FU), gemcitabine, gliadelimplants, hydro xycarbamide, idarubicin, ifosfamide, irinotecan, leucovorin, liposomaldoxorubicin, liposomaldaunorubicin, lomustine,
  • the additional compound can also comprise other biologies, including Herceptin ® (trastuzumab) against the HER2 antigen, Avastin ® (bevacizumab) against VEGF, or antibodies to the EGF receptor, such as Erbitux ® (cetuximab), durvalumab (Medi4736), and Vectibix ® (panitumumab).
  • Herceptin ® tacuzumab
  • Avastin ® bevacizumab
  • VEGF vascular endothelial growth factor receptor
  • EGF receptor such as Erbitux ® (cetuximab), durvalumab (Medi4736), and Vectibix ® (panitumumab).
  • the additional compound can also comprise, for example, an additional immunotherapy.
  • An additional compound can also comprise an immune checkpoint inhibitor antagonist, such as a PD-1 signaling pathway inhibitor, a CD-80/86 and CTLA-4 signaling pathway inhibitor, a T cell membrane protein 3 (TIM3) signaling pathway inhibitor, an adenosine A2a receptor (A2aR) signaling pathway inhibitor, a lymphocyte activation gene 3 (LAG3) signaling pathway inhibitor, a killer immunoglobulin receptor (KIR) signaling pathway inhibitor, a CD40 signaling pathway inhibitor, or any other antigen-presenting cell/T cell signaling pathway inhibitor.
  • an immune checkpoint inhibitor antagonists include an anti-PD-Ll/PD-L2 antibody or fragment thereof, an anti-PD-1 antibody or fragment thereof, an anti-CTLA-4 antibody or fragment thereof, or an anti-B7- H4 antibody or fragment thereof.
  • an anti-PD-1 antibody is Opdivo (nivolumab) an anti-PD-1 monoclonal antibody.
  • a combination therapy comprising a recombinant Listeria strain comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST- containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem, wherein the HPV16 antigenic peptide in an HPV16 E6 antigenic peptide or an HPV16 E7 antigenic peptide, and wherein the HPV18 antigenic peptide is an HPV18 E6 antigenic peptide or an HPV18 E7 antigenic peptide and Opdivo (nivolumab) is used to treat a subject with metastatic cervical cancer.
  • the Advaxis comprising a re
  • Nivolumab to Treat Recurrent or Metastatic Cervical Cancer (ADVANCE) study is to develop a second line treatment for cervical cancer who failed first line treatment in women with recurrent or metastatic cervical cancer.
  • the ADVANCE study is a randomized phase global study with over 500 patients randomized to evaluate the safety and efficacy of ADXS-602 (ADXS-DUAL) in combination with nivolumab compared with investigator's choice of single-agent chemotherapy in patients with recurrent or metastatic cervical cancer who have failed or were ineligible to receive first-line therapy.
  • An additional compound can also comprise a T cell stimulator, such as an antibody or functional fragment thereof binding to a T-cell receptor co-stimulatory molecule, an antigen presenting cell receptor binding co-stimulatory molecule, or a member of the TNF receptor superfamily.
  • the T-cell receptor co-stimulatory molecule can comprise, for example, CD28 or ICOS.
  • the antigen presenting cell receptor binding co-stimulatory molecule can comprise, for example, a CD80 receptor, a CD86 receptor, or a CD46 receptor.
  • the TNF receptor superfamily member can comprise, for example, glucocorticoid-induced TNF receptor (GITR), OX40 (CD134 receptor), 4-1BB (CD137 receptor), or TNFR25. See, e.g., WO2016100929, WO2016011362, and WO2016011357, each of which is incorporated by reference in its entirety for all purposes.
  • the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, and vaccines disclosed herein can be used in various methods. For example, they can be used in methods of inducing an anti-tumor-associated- antigen immune response in a subject, in methods of inducing an anti-tumor or anti-cancer immune response in a subject, in methods of treating a tumor or cancer in a subject, in methods of preventing a tumor or cancer in a subject, or in methods of protecting a subject against a tumor or cancer.
  • T effector cells can also be used in methods of increasing the ratio of T effector cells to regulatory T cells (Tregs) in the spleen and tumor of a subject, wherein the T effector cells are targeted to a tumor-associated antigen. They can also be used in methods for increasing tumor-associated-antigen T cells in a subject, increasing survival time of a subject having a tumor or cancer, delaying the onset of cancer in a subject, or reducing tumor or metastasis size in a subject.
  • the tumor or cancer in any of the above methods can be, for example, an HPV-associated cancer such as a cervical tumor or cancer, an anal tumor or cancer, a head and neck tumor or cancer, or an oropharyngeal tumor or cancer.
  • the cancer in any of the methods described herein can be metastatic cervical cancer.
  • a method of inducing an anti-HPV16 and/or anti-HPV18 immune response in a subject can comprise, for example, administering to the subject a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion polypeptide, a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein (e.g., that comprises a recombinant fusion polypeptide comprising the HPV16 and HPV18 antigenic peptides or a nucleic acid encoding the recombinant fusion polypeptide).
  • An anti-HPV16 and/or anti-HPV18 immune response can thereby be induced in the subject.
  • the Listeria strain in the case of a recombinant Listeria strain, can express the fusion polypeptide, thereby eliciting an immune response in the subject.
  • the immune response can comprise, for example, a T-cell response, such as a CD4+FoxP3- T cell response, a CD8+ T cell response, or a
  • CD4+FoxP3- and CD8+ T cell response can also increase the ratio of T effector cells to regulatory T cells (Tregs) in the spleen and tumor microenvironments of the subject, allowing for a more profound anti-tumor response in the subject.
  • Tregs regulatory T cells
  • a method of inducing an anti-tumor or anti-cancer immune response in a subject can comprise, for example, administering to the subject a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion polypeptide, a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein.
  • An anti-tumor or anti-cancer immune response can thereby be induced in the subject.
  • the Listeria strain can express the fusion polypeptide, thereby eliciting an anti-tumor or anticancer response in the subject.
  • a method of treating a tumor or cancer in a subject can comprise, for example, administering to the subject a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion polypeptide, a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein.
  • the subject can then mount an immune response against the tumor or cancer expressing the HPV16 and/or HPV18 antigenic peptides, thereby treating the tumor or cancer in the subject.
  • a method of preventing a tumor or cancer in a subject or protecting a subject against developing a tumor or cancer can comprise, for example, administering to the subject a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion polypeptide, a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein.
  • the subject can then mount an immune response against the HPV16 and/or HPV18 antigenic peptides, thereby preventing a tumor or cancer or protecting the subject against developing a tumor or cancer.
  • two or more recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines are administered.
  • the multiple recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines can be administered sequentially in any order or combination, or can be administered simultaneously in any combination.
  • four different Listeria strains are being administered, they can be administered sequentially, they can be administered simultaneously, or they can be administered in any combination (e.g., administering the first and second strains simultaneously and subsequently administering the third and fourth strains
  • compositions can be administered during the same immune response, preferably within 0-10 or 3-7 days of each other.
  • the multiple recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines can each comprise a different set of antigenic peptides.
  • two or more can comprise the same set of antigenic peptides (e.g., the same set of antigenic peptides in a different order).
  • HPV-associated cancers include, for example, cancers of the vagina, vulva, penis, anus, rectum, and others.
  • HPV-associated cancers include, but are not limited to, cervical cancer, anal cancer, head and neck cancer, and oropharyngeal cancer.
  • HPV is thought to be responsible for more than 90% of anal and cervical cancers and more than 50% of vaginal, vulvar, and penile cancers.
  • Cancers of the head and neck are mostly caused by tobacco and alcohol, but recent studies show that about 60% to 70% of cancers of the oropharynx may be linked to HPV.
  • treat refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or lessen the targeted tumor or cancer.
  • target tumors or cancers include, but are not limited to, a cervical tumor or cancer, an anal tumor or cancer, a head and neck tumor or cancer, or an oropharyngeal tumor or cancer. Treating may include one or more of directly affecting or curing, suppressing, inhibiting, preventing, reducing the severity of, delaying the onset of, slowing the progression of, stabilizing the progression of, inducing remission of, preventing or delaying the metastasis of, reducing/ameliorating symptoms associated with the tumor or cancer, or a combination thereof.
  • treating may include increasing expected survival time or decreasing tumor or metastasis size.
  • the effect e.g., suppressing, inhibiting, preventing, reducing the severity of, delaying the onset of, slowing the progression of, stabilizing the progression of, inducing remission of, preventing or delaying the metastasis of, reducing/ameliorating symptoms of, and so forth, can be relative to a control subject not receiving a treatment or receiving a placebo treatment.
  • the term “treat” or “treating” can also refer to increasing percent chance of survival or increasing expected time of survival for a subject with the tumor or cancer (e.g., relative to a control subject not receiving a treatment or receiving a placebo treatment).
  • treating refers to delaying progression, expediting remission, inducing remission, augmenting remission, speeding recovery, increasing efficacy of alternative therapeutics, decreasing resistance to alternative therapeutics, or a combination thereof (e.g., relative to a control subject not receiving a treatment or receiving a placebo treatment).
  • preventing or “impeding” can refer, for example to delaying the onset of symptoms, preventing relapse of a tumor or cancer, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, preventing metastasis of a tumor or cancer, or a combination thereof.
  • suppressing can refer, for example, to 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.
  • subject refers to a mammal (e.g., a human) in need of therapy for, or susceptible to developing, a tumor or a cancer.
  • subject also refers to a mammal (e.g., a human) that receives either prophylactic or therapeutic treatment.
  • the subject may include dogs, cats, pigs, cows, sheep, goats, horses, rats, mice, non-human mammals, and humans.
  • subject does not necessarily exclude an individual that is healthy in all respects and does not have or show signs of cancer or a tumor.
  • An individual is at increased risk of developing a tumor or a cancer if the subject has at least one known risk-factor (e.g., genetic, biochemical, family history, and situational exposure) placing individuals with that risk factor at a statistically significant greater risk of developing the tumor or cancer than individuals without the risk factor.
  • risk-factor e.g., genetic, biochemical, family history, and situational exposure
  • a "symptom” or “sign” refers to objective evidence of a disease as observed by a physician or subjective evidence of a disease, such as altered gait, as perceived by the subject.
  • a symptom or sign may be any manifestation of a disease.
  • Symptoms can be primary or secondary.
  • the term "primary” refers to a symptom that is a direct result of a particular disease or disorder (e.g., a tumor or cancer), while the term “secondary” refers to a symptom that is derived from or consequent to a primary cause.
  • the recombinant fusion polypeptides, nucleic acids encoding the recombinant fusion polypeptides, the immunogenic compositions, the pharmaceutical compositions, and the vaccines disclosed herein can treat primary or secondary symptoms or secondary complications.
  • the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines are administered in an effective regime, meaning a dosage, route of administration, and frequency of administration that delays the onset, reduces the severity, inhibits further deterioration, and/or ameliorates at least one sign or symptom of the tumor or cancer.
  • the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines are administered in an effective regime, meaning a dosage, route of
  • the regime can be referred to as a therapeutically effective regime. If the subject is at elevated risk of developing the tumor or cancer relative to the general population but is not yet experiencing symptoms, the regime can be referred to as a prophylactically effective regime.
  • therapeutic or prophylactic efficacy can be observed in an individual patient relative to historical controls or past experience in the same patient. In other instances, therapeutic or prophylactic efficacy can be demonstrated in a preclinical or clinical trial in a population of treated patients relative to a control population of untreated patients.
  • a regime can be considered therapeutically or prophylactically effective if an individual treated patient achieves an outcome more favorable than the mean outcome in a control population of comparable patients not treated by methods described herein, or if a more favorable outcome is demonstrated in treated patients versus control patients in a controlled clinical trial (e.g., a phase II, phase II/III or phase III trial) at the p ⁇ 0.05 or 0.01 or even 0.001 level.
  • a controlled clinical trial e.g., a phase II, phase II/III or phase III trial
  • Exemplary dosages for a recombinant Listeria strain are, for example, 1 x 10 6 - 1 x 10 7 CFU, 1 x 10 7 - 1 x 10 s CFU, 1 x 10 s - 3.31 x 10 10 CFU, 1 x 10 9 - 3.31 x 10 10 CFU, 5-500 x 10 8 CFU, 7-500 x 10 8 CFU, 10-500 x 10 8 CFU, 20-500 x 10 8 CFU, 30-500 x 10 8 CFU, 50-500 x 10 8 CFU, 70-500 x 10 8 CFU, 100-500 x 10 8 CFU, 150-500 x 10 8 CFU, 5- 300 x 10 8 CFU, 5-200 x 10 8 CFU, 5-15 x 10 8 CFU, 5-100 x 10 8 CFU, 5-70 x 10 8 CFU, 5- 50 x 10 8 CFU, 5-30 x 10 8 CFU, 5-20 x 10 8 CFU, 1-30 x 10 9 CFU
  • exemplary dosages for a recombinant Listeria strain are, for example, 1 x 10 7 organisms, 1.5 x 10 7 organisms, 2 x 10 8 organisms, 3 x 10 7 organisms, 4 x 10 7 organisms, 5 x 10 7 organisms, 6 x 10 7 organisms, 7 x 10 7 organisms, 8 x 10 7 organisms, 10 x 10 7 organisms, 1.5 x 10 8 organisms, 2 x 10 8 organisms, 2.5 x 10 8 organisms, 3 x 10 8 organisms, 3.3 x 10 8 organisms, 4 x 10 8 organisms, 5 x 10 8 organisms, 1 x 10 9 organisms, 1.5 x 10 9 organisms, 2 x 10 9 organisms, 3 x 10 9 organisms, 4 x 10 9 organisms, 5 x 10 9 organisms, 6 x 10 9 organisms, 7 x 10 9 organisms, 8 x 10 9 organisms, 10 x 10 organisms, 1.5 x 10 organisms, 2 x 10 organisms, 2.5 x 10
  • Administration can be by any suitable means.
  • administration can be parenteral, intravenous, oral, subcutaneous, intra- arterial, intracranial, intrathecal, intracerebroventricular, intraperitoneal, topical, intranasal, intramuscular, intra-ocular, intrarectal, conjunctival, transdermal, intradermal, vaginal, rectal, intratumoral, parcanceral, transmucosal, intravascular, intraventricular, inhalation (aerosol), nasal aspiration (spray), sublingual, aerosol, suppository, or a combination thereof.
  • solutions or suspensions of the recombinant fusion polypeptides for intranasal administration or application by inhalation, solutions or suspensions of the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines mixed and aerosolized or nebulized in the presence of the appropriate carrier are suitable.
  • Such an aerosol may comprise any recombinant fusion polypeptide, nucleic acids encoding a recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine described herein.
  • Administration may also be in the form of a suppository (e.g., rectal suppository or urethral suppository), in the form of a pellet for subcutaneous implantation (e.g., providing for controlled release over a period of time), or in the form of a capsule. Administration may also be via injection into a tumor site or into a tumor. Regimens of administration can be readily determined based on factors such as exact nature and type of the tumor or cancer being treated, the severity of the tumor or cancer, the age and general physical condition of the subject, body weight of the subject, response of the individual subject, and the like.
  • the frequency of administration can depend on the half-life of the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines in the subject, the condition of the subject, and the route of administration, among other factors.
  • the frequency can be, for example, daily, weekly, monthly, quarterly, or at irregular intervals in response to changes in the subject's condition or progression of the tumor or cancer being treated.
  • the course of treatment can depend on the condition of the subject and other factors. For example, the course of treatment can be several weeks, several months, or several years (e.g., up to 2 years).
  • repeat administrations may be undertaken immediately following the first course of treatment or after an interval of days, weeks or months to achieve tumor regression or suppression of tumor growth.
  • Assessment may be determined by any known technique, including diagnostic methods such as imaging techniques, analysis of serum tumor markers, biopsy, or the presence, absence, or amelioration of tumor-associated symptoms.
  • the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines can be administered every 3 weeks for up to 2 years.
  • a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion polypeptide, a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein is administered in increasing doses in order to increase the T-effector cell to regulatory T cell ratio and generate a more potent anti-tumor immune response.
  • Anti-tumor immune responses can be further strengthened by providing the subject with cytokines including, for example, IFN- ⁇ , TNF-a, and other cytokines known to enhance cellular immune response. See, e.g., US 6,991,785, herein incorporated by reference in its entirety for all purposes.
  • Some methods may further comprise "boosting" the subject with additional recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines or administering the recombinant fusion polypeptides, nucleic acids encoding recombinant fusion polypeptides, recombinant bacteria or Listeria strains, immunogenic compositions, pharmaceutical compositions, or vaccines multiple times.
  • Boosting refers to administering an additional dose to a subject. For example, in some methods, 2 boosts (or a total of 3 inoculations) are administered, 3 boosts are
  • boosts are administered, 4 boosts are administered, 5 boosts are administered, or 6 or more boosts are administered.
  • the number of dosages administered can depend on, for example, the response of the tumor or cancer to the treatment.
  • the recombinant fusion polypeptide, nucleic acids encoding a recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine used in the booster inoculation is the same as the recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine used in the initial "priming" inoculation.
  • the booster recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine is different from the priming recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine.
  • the same dosages are used in the priming and boosting inoculations.
  • a larger dosage is used in the booster, or a smaller dosage is used in the booster.
  • the period between priming and boosting inoculations can be experimentally determined.
  • the period between priming and boosting inoculations can be 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6-8 weeks, or 8-10 weeks.
  • DNA vaccine priming followed by boosting with protein in adjuvant or by viral vector delivery of DNA encoding antigen is one effective way of improving antigen- specific antibody and CD4 + T-cell responses or CD8 + T-cell responses.
  • Shiver et al. (2002) Nature 415: 331-335; Gilbert et al. (2002) Vaccine 20: 1039- 1045; Billaut-Mulot et al. (2000) Vaccine 19:95-102; and Sin et al. (1999) DNA Cell Biol. 18:771-779, each of which is herein incorporated by reference in its entirety for all purposes.
  • adding CRL1005 poloxamer (12 kDa, 5% POE) to DNA encoding an antigen can enhance T-cell responses when subjects are vaccinated with a DNA prime followed by a boost with an adenoviral vector expressing the antigen.
  • immunogenic portion of an antigen and a protein comprising the immunogenic portion of the antigen can be administered. See, e.g., US 2002/0165172, herein incorporated by reference in its entirety for all purposes.
  • an immune response of nucleic acid vaccination can be enhanced by simultaneous administration of (e.g., during the same immune response, preferably within 0-10 or 3-7 days of each other) a polynucleotide and polypeptide of interest. See, e.g., US 6,500,432, herein incorporated by reference in its entirety for all purposes.
  • the therapeutic methods disclosed herein can also comprise administering one or more additional compounds effective in preventing or treating cancer.
  • an additional compound may comprise a compound useful in chemotherapy, such as amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clofarabine, crisantaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, docetaxel, doxorubicin, epirubicin, etoposide, fludarabine, fluorouracil (5-FU), gemcitabine, gliadelimplants, hydroxycarbamide, idarubicin, ifosfamide, irinotecan, leucovorin, liposomaldoxorubicin,
  • a compound useful in chemotherapy such as amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cla
  • an additional compound can also comprise other biologies, including Herceptin ® (trastuzumab) against the HER2 antigen, Avastin ® (bevacizumab) against VEGF, or antibodies to the EGF receptor, such as Erbitux ® (cetuximab), and Vectibix ® (panitumumab).
  • an additional compound can comprise other immunotherapies.
  • the additional compound can be an indoleamine 2,3-dioxygenase (IDO) pathway inhibitor, such as 1- methyltryptophan (1MT), 1-methyltryptophan (1MT), Necro statin- 1, Pyridoxal
  • IDO inhibition can enhance the efficacy of chemotherapeutic agents.
  • the therapeutic methods disclosed herein can also be combined with radiation (e.g., intensity-modulated radiation therapy (IMRT)), stem cell treatment, surgery, or any other treatment.
  • radiation e.g., intensity-modulated radiation therapy (IMRT)
  • stem cell treatment e.g., stem cell treatment, surgery, or any other treatment.
  • IMRT intensity-modulated radiation therapy
  • Such additional compounds or treatments can precede the administration of a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion
  • polypeptide a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein, follow the administration of a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion
  • polypeptide a recombinant bacteria or Listeria strain, an immunogenic composition, a pharmaceutical composition, or a vaccine disclosed herein, or be simultaneous to the administration of a recombinant fusion polypeptide, a nucleic acid encoding a recombinant fusion polypeptide, a recombinant bacteria or Listeria strain, an immunogenic
  • Targeted immunomodulatory therapy is focused primarily on the activation of costimulatory receptors, for example by using agonist antibodies that target members of the tumor necrosis factor receptor superfamily, including 4- IBB, OX40, and GITR (glucocorticoid-induced TNF receptor-related).
  • the modulation of GITR has
  • Another target for agonist antibodies are co-stimulatory signal molecules for T cell activation.
  • costimulatory signal molecules may lead to enhanced activation of T cells and facilitation of a more potent immune response.
  • Co- stimulation may also help prevent inhibitory influences from checkpoint inhibition and increase antigen- specific T cell proliferation.
  • Liste na-based immunotherapy acts by inducing the de novo generation of tumor antigen- specific T cells that infiltrate and destroy the tumor and by reducing the numbers and activities of immunosuppressive regulatory T cells (Tregs) and myeloid- derived suppressor cells (MDSCs) in the tumor microenvironment.
  • Antibodies (or functional fragments thereof) for T cell co-inhibitory or co-stimulatory receptors e.g., checkpoint inhibitors CTLA-4, PD-1, TIM-3, LAG3 and co-stimulators CD137, OX40, GITR, and CD40
  • T cell co-inhibitory or co-stimulatory receptors e.g., checkpoint inhibitors CTLA-4, PD-1, TIM-3, LAG3 and co-stimulators CD137, OX40, GITR, and CD40
  • some methods can comprise further administering a composition comprising an immune checkpoint inhibitor antagonist, such as a PD-1 signaling pathway inhibitor, a CD-80/86 and CTLA-4 signaling pathway inhibitor, a T cell membrane protein 3 (TIM3) signaling pathway inhibitor, an adenosine A2a receptor (A2aR) signaling pathway inhibitor, a lymphocyte activation gene 3 (LAG3) signaling pathway inhibitor, a killer immunoglobulin receptor (KIR) signaling pathway inhibitor, a CD40 signaling pathway inhibitor, or any other antigen-presenting cell/T cell signaling pathway inhibitor.
  • an immune checkpoint inhibitor antagonist such as a PD-1 signaling pathway inhibitor, a CD-80/86 and CTLA-4 signaling pathway inhibitor, a T cell membrane protein 3 (TIM3) signaling pathway inhibitor, an adenosine A2a receptor (A2aR) signaling pathway inhibitor, a lymphocyte activation gene 3 (LAG3) signaling pathway inhibitor, a killer immunoglobulin receptor (KIR) signaling pathway inhibitor, a CD40
  • immune checkpoint inhibitor antagonists include an anti-PD-Ll/PD-L2 antibody or fragment thereof, an anti-PD- 1 antibody or fragment thereof, an anti-CTLA-4 antibody or fragment thereof, or an anti-B7-H4 antibody or fragment thereof.
  • an anti PD-1 antibody can be administered to a subject at 5-10 mg/kg every 2 weeks, 5-10 mg/kg every 3 weeks, 1-2 mg/kg every 3 weeks, 1-10 mg/kg every week, 1- 10 mg/kg every 2 weeks, 1-10 mg/kg every 3 weeks, or 1-10 mg/kg every 4 weeks.
  • some methods can further comprise administering a T cell stimulator, such as an antibody or functional fragment thereof binding to a T-cell receptor co-stimulatory molecule, an antigen presenting cell receptor binding co-stimulatory molecule, or a member of the TNF receptor superfamily.
  • a T cell stimulator such as an antibody or functional fragment thereof binding to a T-cell receptor co-stimulatory molecule, an antigen presenting cell receptor binding co-stimulatory molecule, or a member of the TNF receptor superfamily.
  • the T-cell receptor costimulatory molecule can comprise, for example, CD28 or ICOS.
  • the antigen presenting cell receptor binding co-stimulatory molecule can comprise, for example, a CD80 receptor, a CD86 receptor, or a CD46 receptor.
  • the TNF receptor superfamily member can comprise, for example, glucocorticoid-induced TNF receptor (GITR), OX40 (CD 134 receptor), 4-1BB (CD137 receptor), or TNFR25.
  • some methods can further comprise administering an effective amount of a composition comprising an antibody or functional fragment thereof binding to a T-cell receptor co-stimulatory molecule or an antibody or functional fragment thereof binding to an antigen presenting cell receptor binding a co-stimulatory molecule.
  • the antibody can be, for example, an anti-TNF receptor antibody or antigen-binding fragment thereof (e.g., TNF receptor superfamily member glucocorticoid-induced TNF receptor (GITR), OX40 (CD134 receptor), 4-1BB (CD137 receptor), or TNFR25), an anti-OX40 antibody or antigen-binding fragment thereof, or an anti-GITR antibody or antigen binding fragment thereof.
  • agonistic molecules can be administered (e.g., GITRL, an active fragment of GITRL, a fusion protein containing GITRL, a fusion protein containing an active fragment of GITRL, an antigen presenting cell (APC)/T cell agonist, CD 134 or a ligand or fragment thereof, CD 137 or a ligand or fragment thereof, or an inducible T cell co stimulatory (ICOS) or a ligand or fragment thereof, or an agonistic small molecule).
  • GITRL an active fragment of GITRL
  • a fusion protein containing GITRL e.g., a fusion protein containing GITRL, a fusion protein containing an active fragment of GITRL, an antigen presenting cell (APC)/T cell agonist, CD 134 or a ligand or fragment thereof, CD 137 or a ligand or fragment thereof, or an inducible T cell co stimulatory (ICOS) or a ligand or fragment thereof, or an agonistic small
  • some methods can further comprise administering an anti-CTLA-4 antibody or a functional fragment thereof and/or an anti-CD 137 antibody or functional fragment thereof.
  • the anti-CTLA-4 antibody or a functional fragment thereof or the anti-CD 137 antibody or functional fragment thereof can be administered about 72 hours after the first dose of recombinant fusion polypeptide, nucleic acids encoding a recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine, or about 48 hours after the first dose of recombinant fusion polypeptide, nucleic acids encoding a recombinant fusion polypeptide, recombinant bacteria or Listeria strain, immunogenic composition, pharmaceutical composition, or vaccine.
  • the anti-CTLA-4 antibody or a functional fragment thereof or anti-CD 137 antibody or functional fragment thereof can be administered at a dose, for example, of about 0.05 mg/kg and about 5 mg/kg.
  • a recombinant Listeria strain or immunogenic composition comprising a recombinant Listeria strain can be administered at a dose, for example, of about 1 x 10 9 CFU.
  • Some such methods can further comprise administering an effective amount of an anti-PD- 1 antibody or functional fragment thereof.
  • a prostate cancer model can be to test methods and compositions disclosed herein, such as a TRAMP-C2 mouse model, a 178-2 BMA cell model, a PAIII
  • adenocarcinoma cells model a PC-3M model, or any other prostate cancer model.
  • the immunotherapy can be tested in human subjects, and efficacy can be monitored using known.
  • Such methods can include, for example, directly measuring CD4+ and CD8+ T cell responses, or measuring disease progression (e.g., by determining the number or size of tumor metastases, or monitoring disease symptoms such as cough, chest pain, weight loss, and so forth).
  • Methods for assessing the efficacy of a cancer immunotherapy in human subjects are well known and are described, for example, in Uenaka et al. (2007) Cancer Immun. 7:9 and Thomas-
  • kits comprising a reagent utilized in performing a method disclosed herein or kits comprising a composition, tool, or instrument disclosed herein.
  • kits can comprise a recombinant fusion polypeptide disclosed herein, a nucleic acid encoding a recombinant fusion polypeptide disclosed herein, a recombinant bacteria or Listeria strain disclosed herein, an immunogenic composition disclosed herein, a pharmaceutical composition disclosed herein, or a vaccine disclosed herein.
  • kits can additionally comprise an instructional material which describes use of the recombinant fusion polypeptide, the nucleic acid encoding the recombinant fusion polypeptide, the recombinant Listeria strain, the immunogenic composition, the pharmaceutical composition, or the vaccine to perform the methods disclosed herein.
  • kits can optionally further comprise an applicator.
  • a recombinant Listeria strain comprising a nucleic acid comprising a first open reading frame encoding a fusion polypeptide, wherein the fusion polypeptide comprises a PEST-containing peptide fused to an HPV16 antigenic peptide and an HPV18 antigenic peptide, wherein the HPV16 antigenic peptide and the HPV18 antigenic peptide are operably linked in tandem.2.
  • HPV16-E7 antigenic peptide comprises a sequence at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 96; and/or wherein the HPV18-E7 antigenic peptide comprises a sequence at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 98.
  • segment of the open reading frame encoding the segment of the fusion polypeptide comprising the HPV16 antigenic peptide and the HPV18 antigenic peptide comprises a sequence at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 99, 101, 103, 105, 107, 109, 111, or 113 and encodes the sequence set forth in SEQ ID NO: 100, 102, 104, 106, 108, 110, 112, or 114, respectively.
  • the fusion polypeptide further comprises one or more peptide tags N-terminal and/or C-terminal to the HPV16 antigenic peptide and the HPV18 antigenic peptide operably linked in tandem.
  • the one or more peptide tags comprise one or more of the following: 3xFLAG tag; 2xFLAG tag, 6xHis tag; and SIINFEKL tag.
  • the fusion polypeptide comprises a SIINFEKL tag N-terminal to and a 3xFLAG tag C-terminal to the HPV16 antigenic peptide and the HPV18 antigenic peptide operably linked in tandem.
  • LLO listeriolysin O
  • LLO mutation comprises one of the following: (1) a substitution of residues C484, W491, or W492 of SEQ ID NO: 55 or corresponding substitutions when the LLO protein is optimally aligned with SEQ ID NO: 55; or (2) a deletion of 1-11 amino acids within the residues 483-493 of SEQ ID NO: 55 or a corresponding deletion when the LLO protein is optimally aligned with SEQ ID NO: 55.
  • nucleic acid comprises a second open reading frame encoding a metabolic enzyme.
  • recombinant Listeria strain of any one of embodiments 1-19 wherein the recombinant Listeria strain is an attenuated Listeria monocytogenes strain comprising a deletion of or inactivating mutation in prfA, wherein the nucleic acid is in an episomal plasmid and comprises a second open reading frame encoding a D133V PrfA mutant protein.
  • recombinant Listeria strain of any one of embodiments 1-19 wherein the recombinant Listeria strain is an attenuated Listeria monocytogenes strain comprising a deletion of or inactivating mutation in actA, dal, and dat, wherein the nucleic acid is in an episomal plasmid and comprises a second open reading frame encoding an alanine racemase enzyme or a D-amino acid aminotransferase enzyme, and wherein the PEST-containing peptide is an N-terminal fragment of LLO. 42.
  • recombinant Listeria strain of any one of embodiments 1-19 wherein recombinant Listeria strain is an attenuated Listeria monocytogenes strain comprising a deletion of or inactivating mutation in actA and inlB, wherein the nucleic acid is genomically integrated, and wherein the PEST-containing peptide is an ActA protein or a fragment thereof.
  • recombinant Listeria strain of any preceding embodiment wherein the recombinant Listeria strain has been passaged through an animal host.
  • An immunogenic composition comprising the recombinant Listeria strain of any preceding embodiment.
  • immunogenic composition further comprises an adjuvant.
  • GM-CSF granulocyte/macrophage colony- stimulating factor
  • a method of inducing an immune response against a tumor or cancer in a subject comprising administering to the subject the recombinant Listeria strain of any one of embodiments 1-44 or the immunogenic composition of any one of embodiments 45-47.
  • a method of preventing or treating a tumor or cancer in a subject comprising administering to the subject the recombinant Listeria strain of any one of embodiments 1-44 or the immunogenic composition of any one of embodiments 45-47.
  • the immune checkpoint inhibitor comprises an anti-PD-1 antibody or an antigen-binding fragment thereof and/or an anti-CTLA-4 antibody or an antigen-binding fragment thereof.
  • T cell stimulator comprises an anti-OX40 antibody or an antigen-binding fragment thereof or an anti-GITR antibody or an antigen-binding fragment thereof.
  • tumor or cancer is a cervical tumor or cancer, an anal tumor or cancer, a head and neck tumor or cancer, or an oropharyngeal tumor or cancer.
  • a cell bank comprising one or more recombinant Listeria strains as in any one of embodiments 1-44.
  • 59. The cell bank of embodiment 58, wherein the cell bank is a frozen cell bank or a lyophilized cell bank.
  • nucleotide and amino acid sequences listed in the accompanying sequence listing are shown using standard letter abbreviations for nucleotide bases, and three-letter code for amino acids.
  • the nucleotide sequences follow the standard convention of beginning at the 5' end of the sequence and proceeding forward (i.e., from left to right in each line) to the 3' end. Only one strand of each nucleotide sequence is shown, but the complementary strand is understood to be included by any reference to the displayed strand.
  • the amino acid sequences follow the standard convention of beginning at the amino terminus of the sequence and proceeding forward (i.e., from left to right in each line) to the carboxy terminus.
  • N-Terminal Truncated LLO Protein v.2 (SEQ ID NO: 58):
  • N-Terminal Truncated LLO Protein v.3 (SEQ ID NO: 59):
  • HLA-A2 restricted Epitope from NY-ESO-1 SEQ ID NO: 75
  • HPV16 E7-HPV18E7 insert (SEQ ID NO: 100)
  • GOG-0265 is a single-arm, open- label Phase 2 multicenter study
  • AXAL axalimogene filolisbac
  • PRmCC persistent or recurrent metastatic (squamous or non-squamous cell) carcinoma of the cervix (PRmCC) in a standard Simon two-stage design.
  • AXAL is a live attenuated Listeria monocytogenes (Lrri) bioengineered to secrete an HPV 16 E7 protein fused with a truncated fragment of listeriolysin O (tLLO).
  • tLLO listeriolysin O
  • AXAL targets HPV- transformed cells, inducing antitumor T-cell immunity and breaking immune tolerance in the tumor microenvironment.
  • the first stage of the study included a six-patient safety run- in and enrolled 26 patients. The second stage enrolled 24 patients.
  • AXAL is bioengineered to secrete an E7 protein from HPV16. Patients positive for HPV16 had a 12-month overall survival rate of 57%, which is significantly higher than the expected 25% overall survival rate, consistent with AXAL inducing an anti-tumor immune response against HPV16-transformed cells. Surprisingly, however, the patients who did not test positive for HPV16 but tested positive for HPV18 or
  • HPV45 types of HPV that are not HPV16 or even in the same HPV family as HPV16— had a 38% 12-month overall survival rate, which is a 52% increase in the expected 25% 12-month overall survival rate, despite the fact that the HPV16 E7 protein is only 42% identical to the HPV 18 E7 protein.
  • the resultant plasmid will be electroporated into a suitable Listeria strain.
  • One suitable Listeria strain is strain XFL-7, which lacks the Lm transcriptional activator PrfA, an XFL-7-based Listeria strain, or a similar Listeria strain deficient in prfA.
  • the small size of the HPV 18 E7 protein (105 amino acids; SEQ ID NO: 96) and the small size of the HPV16 E7 protein (97 amino acids; SEQ ID NO: 98) make it possible to generate a live attenuated Listeria monocytogenes ⁇ Lm) bioengineered to secrete a tLLO protein fused to HPV 16 E7 and HPV 18 E7 in tandem even with the size limitations of the platform.
  • TC-1 tumor cells are derived from a C57BL/6 lung epithelial cell line that was immortalized with E6 and E7 of HPV 16 and transformed with an activated ras oncogene. To establish primary tumors, 1 x 105 TC-1 cells were injected subcutaneously in the hind flank of C57BL/6 mice and allowed to grow for 5 days prior to the start of treatment.
  • Tumor- bearing mice received 1 x 108 CFU ADXS-602, 1 x 108 CFU XFL7 (parental strain of ADXS-602 that lacks a tumor-associated antigen), or PBS at weekly intervals for a total of 3 doses (see Figure 1). Tumor volume [(length x width x width)/ 2] was measured twice per week. Mice whose tumor volume approached 2000 mm3 were sacrificed.
  • ADXS-DUAL showed significant tumor control ( Figure 2) and survival ( Figure 3) response when compared to PBS and XFL-7 (empty Lm) vector.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne des polypeptides de fusion recombinés comprenant un peptide antigénique de HPV16 et un peptide antigénique de HPV18, le peptide antigénique de HPV16 et le peptide antigénique de HPV18 étant liés fonctionnellement en tandem (par exemple, fusionnés à un peptide contenant du PEST). La présente invention concerne également des acides nucléiques codant pour de tels polypeptides de fusion, des bactéries ou des souches de Listeria recombinées comprenant de tels polypeptides de fusion ou de tels acides nucléiques, et des banques cellulaires comprenant de telles bactéries ou souches de Listeria recombinées. La présente invention concerne également des méthodes de production de tels polypeptides de fusion, de tels acides nucléiques, et de telles bactéries ou souches de Listeria recombinées. La présente invention concerne également des compositions immunogènes, des compositions pharmaceutiques et des vaccins comprenant de tels polypeptides de fusion, de tels acides nucléiques, ou de telles bactéries ou souches de Listeria recombinées. La présente invention concerne aussi des méthodes d'induction d'une réponse immunitaire anti-antigène associé à une tumeur chez un sujet, des méthodes d'induction d'une réponse immunitaire anti-tumorale ou anticancéreuse chez un sujet, des méthodes de traitement d'une tumeur ou d'un cancer chez un sujet, des méthodes de prévention d'une tumeur ou d'un cancer chez un sujet, et des méthodes de protection d'un sujet contre une tumeur ou un cancer au moyen de tels polypeptides de fusion, acides nucléiques, bactéries ou souches de Listeria recombinés, de compositions immunogènes, de compositions pharmaceutiques, ou de vaccins.
PCT/US2018/012570 2017-01-05 2018-01-05 Souches vaccinales de listeria recombinées et leurs méthodes d'utilisation en immunothérapie anticancéreuse WO2018129306A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762442940P 2017-01-05 2017-01-05
US62/442,940 2017-01-05

Publications (1)

Publication Number Publication Date
WO2018129306A1 true WO2018129306A1 (fr) 2018-07-12

Family

ID=62790883

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/012570 WO2018129306A1 (fr) 2017-01-05 2018-01-05 Souches vaccinales de listeria recombinées et leurs méthodes d'utilisation en immunothérapie anticancéreuse

Country Status (3)

Country Link
AR (1) AR110730A1 (fr)
TW (1) TW201833323A (fr)
WO (1) WO2018129306A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10258679B2 (en) 2014-04-24 2019-04-16 Advaxis, Inc. Recombinant Listeria vaccine strains and methods of producing the same
US20210003558A1 (en) * 2018-03-09 2021-01-07 Advaxis, Inc. Compositions and methods for evaluating attenuation and infectivity of listeria strains
US10900044B2 (en) 2015-03-03 2021-01-26 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11179339B2 (en) 2017-09-19 2021-11-23 Advaxis, Inc. Compositions and methods for lyophilization of bacteria or listeria strains
US11446369B2 (en) 2007-05-10 2022-09-20 Advaxis, Inc. Compositions and methods comprising KLK3 or FOLH1 antigen
US11897927B2 (en) 2016-11-30 2024-02-13 Advaxis, Inc. Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111269868A (zh) * 2019-12-10 2020-06-12 浙江农林大学 一种减毒单核细胞增多性李斯特菌的构建方法和用途

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160228530A1 (en) * 2014-10-14 2016-08-11 The Trustees Of The University Of Pennsylvania Combination therapy for use in cancer therapy
WO2016183361A1 (fr) * 2015-05-13 2016-11-17 Advaxis, Inc. Compositions immunogènes à base de listeria comprenant des protéines hybrides d'acta tronqué-antigène et procédés d'utilisation de ces compositions
WO2017066706A1 (fr) * 2015-10-14 2017-04-20 Advaxis, Inc. Souches recombinées vaccinales de listeria et leurs méthodes d'utilisation en immunothérapie anticancéreuse

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160228530A1 (en) * 2014-10-14 2016-08-11 The Trustees Of The University Of Pennsylvania Combination therapy for use in cancer therapy
WO2016183361A1 (fr) * 2015-05-13 2016-11-17 Advaxis, Inc. Compositions immunogènes à base de listeria comprenant des protéines hybrides d'acta tronqué-antigène et procédés d'utilisation de ces compositions
WO2017066706A1 (fr) * 2015-10-14 2017-04-20 Advaxis, Inc. Souches recombinées vaccinales de listeria et leurs méthodes d'utilisation en immunothérapie anticancéreuse

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KANODIA, SHREYA ET AL.: "Recent advances in strategies for immunotherapy of human papillomavirus-induced lesions : Immunotherapy of HPV-Induced", INTERNATIONAL JOURNAL OF CANCER, vol. 122, no. 2, 15 January 2008 (2008-01-15), pages 247 - 259, XP055512372 *
KHALLOUF, HADEEL ET AL.: "Therapeutic vaccine strategies against human papillomavirus", VACCINES, vol. 2, no. 2, 13 June 2014 (2014-06-13), pages 422 - 462, XP055425140 *
YANG, ANDREW ET AL.: "Perspectives for therapeutic HPV vaccine development", JOURNAL OF BIOMEDICAL SCIENCE, vol. 23, no. 1, 4 November 2016 (2016-11-04), pages 75, XP055512363 *

Cited By (7)

* 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
US10258679B2 (en) 2014-04-24 2019-04-16 Advaxis, Inc. Recombinant Listeria vaccine strains and methods of producing the same
US10900044B2 (en) 2015-03-03 2021-01-26 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11702664B2 (en) 2015-03-03 2023-07-18 Advaxis, Inc. Listeria-based compositions comprising a peptide minigene expression system and methods of use thereof
US11897927B2 (en) 2016-11-30 2024-02-13 Advaxis, Inc. Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof
US11179339B2 (en) 2017-09-19 2021-11-23 Advaxis, Inc. Compositions and methods for lyophilization of bacteria or listeria strains
US20210003558A1 (en) * 2018-03-09 2021-01-07 Advaxis, Inc. Compositions and methods for evaluating attenuation and infectivity of listeria strains

Also Published As

Publication number Publication date
TW201833323A (zh) 2018-09-16
AR110730A1 (es) 2019-04-24

Similar Documents

Publication Publication Date Title
WO2019157098A1 (fr) Compositions comprenant une souche de listeria recombinante et un anticorps anti-ccr8 et méthodes d'utilisation
US20240124540A1 (en) Immunogenic compositions targeting recurrent cancer mutations and methods of use thereof
WO2018129306A1 (fr) Souches vaccinales de listeria recombinées et leurs méthodes d'utilisation en immunothérapie anticancéreuse
AU2018336988B2 (en) Compositions and methods for lyophilization of bacteria or Listeria strains
US20210177955A1 (en) Immunogenic heteroclitic peptides from cancer-associated proteins and methods of use thereof
US20190248856A1 (en) Listeria-Based Immunogenic Compositions Comprising Wilms Tumor Protein Antigens And Methods Of Use Thereof
WO2019006401A2 (fr) Compositions immunogènes à base de listeria comprenant des antigènes de protéine tumorale de wilms hétéroclitique et procédés d'utilisation correspondants
US20180360940A1 (en) Listeria-based immunotherapy and methods of use thereof
WO2018170313A1 (fr) Procédés et compositions destinés à augmenter l'efficacité de vaccins

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: 18736111

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18736111

Country of ref document: EP

Kind code of ref document: A1