[go: up one dir, main page]

WO2025122955A1 - Delivery of oncolytic viruses with engineered salmonella - Google Patents

Delivery of oncolytic viruses with engineered salmonella Download PDF

Info

Publication number
WO2025122955A1
WO2025122955A1 PCT/US2024/059002 US2024059002W WO2025122955A1 WO 2025122955 A1 WO2025122955 A1 WO 2025122955A1 US 2024059002 W US2024059002 W US 2024059002W WO 2025122955 A1 WO2025122955 A1 WO 2025122955A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
vds
salmonella
bacterial cell
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2024/059002
Other languages
French (fr)
Inventor
Shradha KHANDUJA
Neil S. Forbes
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Massachusetts Amherst
Original Assignee
University of Massachusetts Amherst
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 University of Massachusetts Amherst filed Critical University of Massachusetts Amherst
Publication of WO2025122955A1 publication Critical patent/WO2025122955A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766

Definitions

  • VDS virus-delivering Salmonella
  • the VDS strain contains the PsseJ-lysE delivery circuit and has deletions in four homologous recombination genes ( ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF) to preserve hairpins in the viral genome with a role in replication and infectivity.
  • VDS delivered the genome of a virus to multiple cancers, including breast, pancreatic carcinoma, and osteosarcoma. Viral delivery produced functional viral particles that were cytotoxic and infective to neighboring cells. The release of mature virions initiated new rounds of infection and amplified the infection.
  • Salmonella for delivery circumvents the limitations of oncolytic viruses and provides a novel therapy for cancer.
  • FIG.1 Bacterial delivery of viruses into the cancer cells.
  • VDS carrying the genome of MVMp, invades cancer cells.
  • Salmonella lysis releases the viral plasmid into Salmonella-containing vacuoles (SCVs).
  • SCV breakdown releases the plasmid into the cytoplasm.
  • FIGS.2A-2B ID Salmonella delivers encoding plasmids into cells.
  • CMV-GFP eukaryotic-expression plasmid
  • PsseJ-lysE intracellular delivering gene circuit
  • FIGS. 3A-3D Bacteria with PsseJ-lysE (ID-CMV-GFP) induced significantly more GFP expression than control CMV-GFP bacteria (***, P ⁇ 0.001).
  • FIGS. 3A-3D Parental Salmonella deleted the right-end hairpin and disabled viral replication.
  • A) The MVMp plasmid contains the full MVM genome including the left- and right-end hairpins. This plasmid replicates in Salmonella and produces viruses when in transfected into mammalian cells.
  • An SSPI restriction digest of the MVMp plasmid produced three fragments (left), 5078, 1395 and 732 bp, when maintained in SURE 2.
  • Strains CD, BB’CDF and BCDJ maintained the complete right-end hairpin similar to SURE 2. All of the other strains had a 97 bp deletion resulting in a shorter third fragment (635 bp).
  • FIGS.5A-5F Deletion of homologous recombination genes does not impair growth or cell invasion.
  • FIGS. 6A-6I VDS delivers functional virions that kill cancer cells.
  • VDS-B contains both the MVMp and PsseJ-lysE plasmids
  • BB’CDF contains only the PsseJ-lysE plasmid.
  • VDS-C was compared to strain CD, its bacterial control. After two hours, external bacteria were removed with gentamicin (green arrow). In parallel, positive control cells were directly transfected with MVMp using lipofectamine (bottom). After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using crystal violet, ethidium homodimer and MTT.
  • VDS-B invaded cancer cells in culture, the cells produced viral NS1, similar to direct transfection with the MVMp plasmid. Cell invaded with VDS-C did not produce NS1.
  • VDS-B At 60 hours after invasion (or transfection), more dead cells (arrows, ethidium homodimer + ) started to appear in cultures treated with VDS-B or directly transfected with MVMp.
  • F With time, the area of cell death increased for VDS-B and MVMp transfection but remained constant for BB’CDF.
  • G Death caused by VDS-B was greater than BB’CDF (*, P ⁇ 0.05).
  • VDS-C did not increase cell death compared to CD.
  • FIGS. 7A-7C VDS delivers function virions that spread to neighboring cells.
  • conditioned media from VDS-treated cells was applied to cultures of na ⁇ ve cells. Cancer cells (4T1) were incubated (blue arrows) with VDS-B or bacterial controls (BB’CDF) and external bacteria were cleared after two hours with gentamicin (green arrow, top). Positive controls were cells directly transfected with the MVMp plasmid using lipofectamine (bottom). After 72 hours, the culture medium was collected and added to fresh media at a 40:60 ratio. Addition of 50 ⁇ g/mL gentamycin prevented bacterial growth.
  • This conditioned medium was added to na ⁇ ve cultures of (1) HEK- 293T cells to measure virus formation, and (2) 4T1 cancer cells to measure cytotoxicity.
  • VDS-B Treatment with VDS-B produced NS1 when applied to human embryonic kidney cells (HEK- 293T), human osteosarcoma cells (U2OS), and murine pancreatic ductal adenocarcinoma cells (KPCY). No NS1 was expressed in any of the cell lines after being treated with VDS-C.
  • FIGS. 9A-9B Deletion of homologous recombination genes maintains the right-end hairpin of H1PV.
  • FIGS.10A-10I VDS delivers functional virions that kill cancer cells. To measure virus delivery and induced lysis, VDS was applied to cultures of Human Hepatocellular carcinoma (HCC) cells. Human Hepatocellular carcinoma cells (HUH7) were incubated with VDS-B or bacterial controls (BB’CDF).
  • HCC Human Hepatocellular carcinoma
  • BB’CDF Human Hepatocellular carcinoma cells
  • VDS-B contains both the H1PV or MH1PV and PsseJ- lysE plasmids; BB’CDF contains only the PsseJ-lysE plasmid.
  • VDS-C was compared to strain CDS, its bacterial control and VDS-BS was compared to its bacterial control BB’CDFS. After two hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using MTT.
  • VDS virus delivery and induced lysis
  • HCC Murine Hepatocellular carcinoma
  • Hepa1-6 Murine Hepatocellular carcinoma cells
  • BB bacterial controls
  • VDS- C was compared to strain CDS, its bacterial control. After three hours, external bacteria were removed with gentamicin.
  • positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using MTT.
  • FIGS. 12A-12E To measure virus delivery and induced lysis, VDS was applied to cultures of pancreatic ductal adenocarcinoma cells (KPCY). Murine pancreatic ductal adenocarcinoma cells (KPCY) were incubated with VDS-B or bacterial controls (BB’CDF).
  • VDS-C was compared to strain CDS, its bacterial control. After three hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using MTT.
  • FIGS. 13A-13K To measure virus delivery and induced lysis, VDS was applied to cultures of human embryonic kidney cells (HEK-293T) and human Mammary Adenocarcinoma (MCF 7). KPCY and MCF 7 were incubated with VDS-B or bacterial controls (BB’CDF).
  • VDS-C was compared to strain CDS, its bacterial control and VDS-BS was compared to its bacterial control BB’CDFS. After two hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After a further 72 hours, cell viability and cell death were measured using MTT. For HEK-293T cells A) Cell viability, measured by MTT assay, decreased for H1PV VDS-B compared to BB’CDF (****, P ⁇ 0.0001). B) H1PV VDS-C did not increase cell death compared to CDS.
  • H1PV VDS-BS did not increase cell death compared to BB’CDFS.
  • H) H1PV VDS-BS did not increase cell death compared to BB’CDFS.
  • FIGS. 14A-14H Salmonella delivers functional H1PV virions that are cytotoxic to liver cancer cells: A) Similar to the SURE 2 strain of E. coli, the engineered BB’CDF and CDS strains preserved H1PV genome integrity as shown by both DNA bands after restriction digestion.
  • VDS-B and VDS-C delivered functional virus into Hepa 1-6 mouse hepatoma cells as shown by expression of NS1.
  • H Delivery of H1PV with VDS-B induced significant cellular cytotoxicity compared to the bacterial control (*, P ⁇ 0.05).
  • FIGS.16A-16E Figure 3: VDS-B inhibits the growth of liver cancer in vivo.
  • VDS-B mice injected with VDS-B exhibited minimal tumor growth as compared to mice injected with the BB’CDF control strain.
  • FIGS. 17A-17F. Intravenous injection is similarly efficacious compared to intratumoral injection of VDS-B.
  • VDS-B generates an innate immunostimulatory response against tumors.
  • VDS-B generates an anti-tumor adaptive immune response.
  • D) The ratio of CD8+ T cells to regulatory T cells increased 12-fold after treatment with VDS-B as compared BB’CDF (*, P ⁇ 0.05).
  • mice were challenged with hepa1-6 tumor cells two weeks after splenocyte transfer and monitored for tumor growth for two 66 days.
  • J 75% of na ⁇ ve mice grafted hepa1-6 tumor cells as compared to 0% of mice with adoptively transferred splenocytes from VDS-B treated, tumor bearing mice.
  • K After tumor cell injection, mice with adoptively transferred splenocytes from VDS-B treated mice exhibited no tumor growth, in contrast to the control mice.
  • Engineered Salmonella deliver functional H1PV in liver tumors in vivo.
  • A) C57BL/6 mice with subcutaneous Hepa1-6 tumors were intravenously injected with BB’CDF.
  • OVs Oncolytic viruses
  • OVs When injected systemically, OVs are cleared from the blood and do not effectively reach tumors (1, 5, 7-20). This problem could be overcome with a carrier with tumor specific tropism. It has been shown that bacterial therapies predominantly accumulate in tumors after intravenous injections (21-30). Developing a bacterial system to deliver viruses into cancer cells would couple the benefits of these two microbial therapies and focus treatment specifically to cancers.
  • the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.”
  • the terms “including,” “includes,” “having,” “has,” “with,” or variants thereof, are intended to be inclusive similar to the term “comprising.”
  • the term “about” means plus or minus 10% of the indicated value. For example, about 100 means from 90 to 110.
  • Numerical ranges recited herein by endpoints include all numbers and fractions subsumed within that range (e.g., 1 to 5 includes 1, 1.5, 2, 2.75, 3, 3.90, 4, and 5). It is also to be understood that all numbers and fractions thereof are presumed to be modified by the term “about.”
  • the terms "individual,” “subject,” and “patient,” are used interchangeably herein and refer to any subject for whom diagnosis, treatment, or therapy is desired, including a mammal. Mammals include, but are not limited to, humans, farm animals, sport animals and pets. A “subject” is a vertebrate, such as a mammal, including a human. Mammals include, but are not limited to, humans, farm animals, sport animals and companion animals.
  • animal is dog, cat, fish, gerbil, guinea pig, hamster, horse, rabbit, swine, mouse, monkey (e.g., ape, gorilla, chimpanzee, orangutan) rat, sheep, goat, cow and bird.
  • treatment e.g., ape, gorilla, chimpanzee, orangutan
  • rat sheep, goat, cow and bird.
  • treatment e.g., ape, gorilla, chimpanzee, orangutan
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • various clinical and scientific methodologies and assays may be used to assess the development or progression of a disorder, and similarly, various clinical and scientific methodologies and assays may be used to assess the reduction, regression, or remission of a disorder or its symptoms.
  • treatment can be applied to a subject or to a cell culture (in vivo or in vitro).
  • inhibitor refers to the slowing, halting, or reversing the growth or progression of a disease, infection, condition, group of cells, protein or its expression.
  • the inhibition can be greater than about 20%, 40%, 60%, 80%, 90%, 95%, or 99%, for example, compared to the growth or progression that occurs in the absence of the treatment or contacting.
  • “Expression” refers to the production of RNA from DNA and/or the production of protein directed by genetic material (e.g., RNA (mRNA)).
  • mRNA RNA
  • Inducible expression is expression which only occurs under certain conditions, such as in the presence of specific molecule (e.g., arabinose) or an environmental que.
  • nucleic acid or a protein
  • a host refers to a nucleic acid that does not occur in (and cannot be obtained from) a cell of that particular type as it is found in nature or a protein encoded by such a nucleic acid.
  • a non-naturally occurring nucleic acid is considered to be exogenous to a host once in the host. It is important to note that non-naturally occurring nucleic acids can contain nucleic acid subsequences or fragments of nucleic acid sequences that are found in nature provided the nucleic acid as a whole does not exist in nature.
  • a nucleic acid molecule containing a genomic DNA sequence within an expression vector is non-naturally occurring nucleic acid, and thus is exogenous to a host cell once introduced into the host, since that nucleic acid molecule as a whole (genomic DNA plus vector DNA) does not exist in nature.
  • any vector, autonomously replicating plasmid, or virus e.g., retrovirus, adenovirus, or herpes virus
  • genomic DNA fragments produced by PCR or restriction endonuclease treatment as well as cDNAs are considered to be non-naturally occurring nucleic acid since they exist as separate molecules not found in nature.
  • an exogenous sequence may therefore be integrated into the genome of the host. It also follows that any nucleic acid containing a promoter sequence and polypeptide-encoding sequence (e.g., cDNA or genomic DNA) in an arrangement not found in nature is non-naturally occurring nucleic acid.
  • a nucleic acid that is naturally occurring can be exogenous to a particular host microorganism. For example, an entire chromosome isolated from a cell of yeast x is an exogenous nucleic acid with respect to a cell of yeast y once that chromosome is introduced into a cell of yeast y.
  • endogenous as used herein with reference to a nucleic acid (e.g., a gene) (or a protein) and a host refers to a nucleic acid (or protein) that does occur in (and can be obtained from) that particular host as it is found in nature.
  • a cell “endogenously expressing” a nucleic acid (or protein) expresses that nucleic acid (or protein) as does a host of the same particular type as it is found in nature.
  • a host “endogenously producing” or that "endogenously produces” a nucleic acid, protein, or other compound produces that nucleic acid, protein, or compound as does a host of the same particular type as it is found in nature.
  • Engineered Salmonella could be any strain of Salmonella designed to lyse and deliver protein intracellularly.
  • the engineered Salmonella is non-pathogenic.
  • the term "contacting” refers to the act of touching, making contact, or of bringing to immediate or close proximity, including at the cellular or molecular level, for example, to bring about a physiological reaction, a chemical reaction, or a physical change, e.g., in a solution, in a reaction mixture, in vitro, or in vivo.
  • An "effective amount” is an amount sufficient to effect beneficial or desired result, such as a preclinical or clinical result. An effective amount can be administered in one or more administrations.
  • an effective amount means the quantity necessary to render the desired therapeutic result.
  • an effective amount is a level effective to treat, cure, or alleviate the symptoms of a disorder and/or disease for which the therapeutic compound, biologic or composition is being administered.
  • Amounts effective for the particular therapeutic goal sought will depend upon a variety of factors including the disorder being treated and its severity and/or stage of development/progression; the bioavailability, and activity of the specific compound, biologic or pharmaceutical composition used; the route or method of administration and introduction site on the subject; the rate of clearance of the specific compound or biologic and other pharmacokinetic properties; the duration of treatment; inoculation regimen; drugs used in combination or coincident with the specific compound, biologic or composition; the age, body weight, sex, diet, physiology and general health of the subject being treated; and like factors well known to one of skill in the relevant scientific art.
  • dosage can occur depending upon the condition of the subject being treated, and the physician or other individual administering treatment will, in any event, determine the appropriate dose for an individual patient.
  • disorder refers to a disorder, disease or condition, or other departure from healthy or normal biological activity, and the terms can be used interchangeably. The terms would refer to any condition that impairs normal function.
  • the condition may be caused by sporadic or heritable genetic abnormalities.
  • the condition may also be caused by non- genetic abnormalities.
  • the condition may also be caused by injuries to a subject from environmental factors, such as, but not limited to, cutting, crushing, burning, piercing, stretching, shearing, injecting, or otherwise modifying a subject's cell(s), tissue(s), organ(s), system(s), or the like.
  • the terms “cell,” “cell line,” and “cell culture” as used herein may be used interchangeably. All of these terms also include their progeny, which are any and all subsequent generations. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations.
  • a “coding region” of a gene consists of the nucleotide residues of the coding strand of the gene and the nucleotides of the non-coding strand of the gene which are homologous with or complementary to, respectively, the coding region of an mRNA molecule which is produced by transcription of the gene.
  • “Complementary” as used herein refers to the broad concept of subunit sequence complementarity between two nucleic acids, e.g., two DNA molecules. When a nucleotide position in both of the molecules is occupied by nucleotides normally capable of base pairing with each other, then the nucleic acids are considered to be complementary to each other at this position.
  • nucleic acids are complementary to each other when a substantial number (at least 50%) of corresponding positions in each of the molecules are occupied by nucleotides which normally base pair with each other (e.g., A:T and G:C nucleotide pairs).
  • nucleotides which normally base pair with each other (e.g., A:T and G:C nucleotide pairs).
  • base pairing specific hydrogen bonds
  • a cytosine residue of a first nucleic acid strand is capable of base pairing with a residue of a second nucleic acid strand which is antiparallel to the first strand if the residue is guanine.
  • a first region of a nucleic acid is complementary to a second region of the same or a different nucleic acid if, when the two regions are arranged in an antiparallel fashion, at least one nucleotide residue of the first region is capable of base pairing with a residue of the second region.
  • the first region comprises a first portion and the second region comprises a second portion, whereby, when the first and second portions are arranged in an antiparallel fashion, at least about 50%, and preferably at least about 75%, at least about 90%, or at least about 95% of the nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. More preferably, all nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion.
  • Encoding refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
  • both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA.
  • an “essentially pure” preparation of a particular protein or peptide is a preparation wherein at least about 95%, and preferably at least about 99%, by weight, of the protein or peptide in the preparation is the particular protein or peptide.
  • a “fragment” or “segment” is a portion of an amino acid sequence, comprising at least one amino acid, or a portion of a nucleic acid sequence comprising at least one nucleotide.
  • the terms “fragment” and “segment” are used interchangeably herein.
  • a “functional” biological molecule is a biological molecule in a form in which it exhibits a property by which it is characterized.
  • a functional enzyme for example, is one which exhibits the characteristic catalytic activity by which the enzyme is characterized.
  • “Homologous” as used herein refers to the subunit sequence similarity between two polymeric molecules, e.g., between two nucleic acid molecules, e.g., two DNA molecules or two RNA molecules, or between two polypeptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position.
  • the homology between two sequences is a direct function of the number of matching or homologous positions, e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two compound sequences are homologous then the two sequences are 50% homologous, if 90% of the positions, e.g., 9 of 10, are matched or homologous, the two sequences share 90% homology.
  • the DNA sequences 3’ATTGCC5’ and 3’TATGGC share 50% homology.
  • “homology” is used synonymously with “identity.” The determination of percent identity between two nucleotide or amino acid sequences can be accomplished using a mathematical algorithm.
  • a mathematical algorithm useful for comparing two sequences is the algorithm of Karlin and Altschul (1990, Proc. Natl. Acad. Sci. USA 87:2264-2268), modified as in Karlin and Altschul (1993, Proc. Natl. Acad. Sci. USA 90:5873-5877).
  • This algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990, J. Mol. Biol. 215:403-410), and can be accessed, for example at the National Center for Biotechnology Information (NCBI) world wide web site having the universal resource locator using the BLAST tool at the NCBI website.
  • NCBI National Center for Biotechnology Information
  • BLAST protein searches can be performed with the XBLAST program (designated “blastn” at the NCBI web site) or the NCBI “blastp” program, using the following parameters: expectation value 10.0, BLOSUM62 scoring matrix to obtain amino acid sequences homologous to a protein molecule described herein.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997, Nucleic Acids Res. 25:3389-3402).
  • PSI-Blast or PHI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.) and relationships between molecules which share a common pattern.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically exact matches are counted.
  • hybridization is used in reference to the pairing of complementary nucleic acids. Hybridization and the strength of hybridization (i.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementarity between the nucleic acids, stringency of the conditions involved, the length of the formed hybrid, and the G:C ratio within the nucleic acids.
  • an “instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the peptide of the invention in the kit for effecting alleviation of the various diseases or disorders recited herein.
  • the instructional material may describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a mammal.
  • the instructional material of the kit of the invention may, for example, be affixed to a container which contains the identified compound invention or be shipped together with a container which contains the identified compound.
  • the instructional material may be shipped separately from the container with the intention that the instructional material and the compound be used cooperatively by the recipient.
  • nucleic acid typically refers to large polynucleotides.
  • nucleic acid is meant any nucleic acid, whether composed of deoxyribonucleosides or ribonucleosides, and whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sulfone linkages, and combinations of such linkages.
  • phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridge
  • nucleic acid also specifically includes nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine and uracil).
  • nucleic acid encompasses RNA as well as single and double stranded DNA and cDNA.
  • nucleic acid encompasses RNA as well as single and double stranded DNA and cDNA.
  • nucleic acid encompasses RNA as well as single and double stranded DNA and cDNA.
  • DNA DNA
  • RNA and similar terms also include nucleic acid analogs, i.e., analogs having other than a phosphodiester backbone.
  • peptide nucleic acids which are known in the art and have peptide bonds instead of phosphodiester bonds in the backbone, are considered within the scope of the present invention.
  • nucleic acid is meant any nucleic acid, whether composed of deoxyribonucleosides or ribonucleosides, and whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sulfone linkages, and combinations of such linkages.
  • phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridge
  • nucleic acid also specifically includes nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine, and uracil).
  • bases other than the five biologically occurring bases
  • Conventional notation is used herein to describe polynucleotide sequences: the left-hand end of a single-stranded polynucleotide sequence is the 5’-end; the left-hand direction of a double-stranded polynucleotide sequence is referred to as the 5’-direction.
  • the direction of 5’ to 3’ addition of nucleotides to nascent RNA transcripts is referred to as the transcription direction.
  • nucleic acid construct encompasses DNA and RNA sequences encoding the particular gene or gene fragment desired, whether obtained by genomic or synthetic methods. Unless otherwise specified, a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence.
  • Nucleotide sequences that encode proteins and RNA may include introns.
  • the term “oligonucleotide” typically refers to short polynucleotides, generally, no greater than about 50 nucleotides. It will be understood that when a nucleotide sequence is represented by a DNA sequence (i.e., A, T, G, C), this also includes an RNA sequence (i.e., A, U, G, C) in which “U” replaces “T.”
  • “Substantially homologous nucleic acid sequence” means a nucleic acid sequence corresponding to a reference nucleic acid sequence wherein the corresponding sequence encodes a peptide having substantially the same structure and function as the peptide encoded by the reference nucleic acid sequence; e.g., where only changes in amino acids not significantly affecting the peptide function occur.
  • the substantially identical nucleic acid sequence encodes the peptide encoded by the reference nucleic acid sequence.
  • the percentage of identity between the substantially similar nucleic acid sequence and the reference nucleic acid sequence is at least about 50%, 65%, 75%, 85%, 95%, 99% or more.
  • Substantial identity of nucleic acid sequences can be determined by comparing the sequence identity of two sequences, for example by physical/chemical methods (i.e., hybridization) or by sequence alignment via computer algorithm.
  • Suitable nucleic acid hybridization conditions to determine if a nucleotide sequence is substantially similar to a reference nucleotide sequence are: 7% sodium dodecyl sulfate SDS, 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 2X standard saline citrate (SSC), 0.1% SDS at 50°C; preferably in 7% (SDS), 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 1X SSC, 0.1% SDS at 50°C; preferably 7% SDS, 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 0.5X SSC, 0.1% SDS at 50°C; and more preferably in 7% SDS, 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 0.1X SSC, 0.1% SDS at 65°C.
  • Suitable computer algorithms to determine substantial similarity between two nucleic acid sequences include, GCS program package (Devereux et al., 1984 Nucl. Acids Res. 12:387), and the BLASTN or FASTA programs (Altschul et al., 1990 Proc. Natl. Acad. Sci. USA.1990 87:14:5509-13; Altschul et al., J. Mol. Biol. 1990215:3:403-10; Altschul et al., 1997 Nucleic Acids Res.25:3389-3402). The default settings provided with these programs are suitable for determining substantial similarity of nucleic acid sequences for purposes of the present invention.
  • two polynucleotides as “operably linked” is meant that a single-stranded or double-stranded nucleic acid moiety comprises the two polynucleotides arranged within the nucleic acid moiety in such a manner that at least one of the two polynucleotides is able to exert a physiological effect by which it is characterized upon the other.
  • a promoter operably linked to the coding region of a gene is able to promote transcription of the coding region.
  • pharmaceutically acceptable carrier means a chemical composition with which an appropriate compound or derivative can be combined and which, following the combination, can be used to administer the appropriate compound to a subject.
  • “Pharmaceutically acceptable” means physiologically tolerable, for either human or veterinary application.
  • “pharmaceutical compositions” include formulations for human and veterinary use.
  • the term “purified” and like terms relate to an enrichment of a molecule or compound relative to other components normally associated with the molecule or compound in a native environment. The term “purified” does not necessarily indicate that complete purity of the particular molecule has been achieved during the process.
  • a “highly purified” compound as used herein refers to a compound that is greater than 90% pure.
  • purified sperm cell DNA refers to DNA that does not produce significant detectable levels of non-sperm cell DNA upon PCR amplification of the purified sperm cell DNA and subsequent analysis of that amplified DNA.
  • a “significant detectable level” is an amount of contaminate that would be visible in the presented data and would need to be addressed/explained during analysis of the forensic evidence.
  • “Recombinant polynucleotide” refers to a polynucleotide having sequences that are not naturally joined together. An amplified or assembled recombinant polynucleotide may be included in a suitable vector, and the vector can be used to transform a suitable host cell.
  • a recombinant polynucleotide may serve a non-coding function (e.g., promoter, origin of replication, ribosome-binding site, etc.) as well.
  • a host cell that comprises a recombinant polynucleotide is referred to as a “recombinant host cell.”
  • a gene which is expressed in a recombinant host cell wherein the gene comprises a recombinant polynucleotide produces a “recombinant polypeptide.”
  • a “recombinant polypeptide” is one which is produced upon expression of a recombinant polynucleotide.
  • a “recombinant cell” is a cell that comprises a transgene.
  • Such a cell may be a eukaryotic or a prokaryotic cell.
  • the transgenic cell encompasses, but is not limited to, an embryonic stem cell comprising the transgene, a cell obtained from a chimeric mammal derived from a transgenic embryonic stem cell where the cell comprises the transgene, a cell obtained from a transgenic mammal, or fetal or placental tissue thereof, and a prokaryotic cell comprising the transgene.
  • the term “regulate” refers to either stimulating or inhibiting a function or activity of interest.
  • binds to when a compound or ligand functions in a binding reaction or assay conditions which is determinative of the presence of the compound in a sample of heterogeneous compounds, or it means that one molecule, such as a binding moiety, e.g., an oligonucleotide or antibody, binds preferentially to another molecule, such as a target molecule, e.g., a nucleic acid or a protein, in the presence of other molecules in a sample.
  • a binding moiety e.g., an oligonucleotide or antibody
  • telomere binding domain a structure allowing recognition and binding to a specific protein structure within a binding partner rather than to molecules in general.
  • a ligand is specific for binding pocket "A,” in a reaction containing labeled peptide ligand "A” (such as an isolated phage displayed peptide or isolated synthetic peptide) and unlabeled "A" in the presence of a protein comprising a binding pocket A the unlabeled peptide ligand will reduce the amount of labeled peptide ligand bound to the binding partner, in other words a competitive binding assay.
  • labeled peptide ligand "A” such as an isolated phage displayed peptide or isolated synthetic peptide
  • unlabeled peptide ligand will reduce the amount of labeled peptide ligand bound to the binding partner, in other words a competitive binding assay.
  • standard refers to something used for comparison.
  • Standard can be a known standard agent or compound which is administered and used for comparing results when administering a test compound, or it can be a standard parameter or function which is measured to obtain a control value when measuring an effect of an agent or compound on a parameter or function.
  • Standard can also refer to an “internal standard”, such as an agent or compound which is added at known amounts to a sample and is useful in determining such things as purification or recovery rates when a sample is processed or subjected to purification or extraction procedures before a marker of interest is measured.
  • Internal standards are often a purified marker of interest which has been labeled, such as with a radioactive isotope, allowing it to be distinguished from an endogenous marker. Methods involving conventional molecular biology techniques are described herein.
  • Bacteria Bacteria useful in the invention include, but are not limited to, Clostridium, Bifidus, Escherichia coli or Salmonella, T3SS-dependent bacteria, such as shigella, salmonella and Yersinia Pestis. Further, E.
  • Salmonella coli can be used if the T3SS system is place in E. Coli.
  • Salmonella examples of Salmonella strains which can be employed in the present invention include Salmonella typhi (ATCC No. 7251) and S. typhimurium (ATCC No. 13311). Attenuated Salmonella strains include S. typhi-aroC-aroD (Hone et al. Vacc.9:810 (1991) S. typhimurium- aroA mutant (Mastroeni et al. Micro. Pathol. 13:477 (1992)) and Salmonella typhimurium 7207.
  • Additional attenuated Salmonella strains that can be used in the invention include one or more other attenuating mutations such as (i) auxotrophic mutations, such as aro (Hoiseth et al. Nature, 291:238-239 (1981)), gua (McFarland et al Microbiol. Path., 3:129-141 (1987)), nad (Park et al. J. Bact, 170:3725-3730 (1988), thy (Nnalue et al. Infect.
  • auxotrophic mutations such as aro (Hoiseth et al. Nature, 291:238-239 (1981)), gua (McFarland et al Microbiol. Path., 3:129-141 (1987)), nad (Park et al. J. Bact, 170:3725-3730 (1988), thy (Nnalue et al. Infect.
  • the attenuating mutations can be either constitutively expressed or under the control of inducible promoters, such as the temperature sensitive heat shock family of promoters (Neidhardt et al. supra), or the anaerobically induced nirB promoter (Harbome et al. Mol. Micro., 6:2805-2813 (1992)) or repressible promoters, such as uapA (Gorfinkiel et al. J. Biol. Chem., 268:23376-23381 (1993)) or gcv (Stauffer et al. J. Bact, 176:6159-6164 (1994)).
  • inducible promoters such as the temperature sensitive heat shock family of promoters (Neidhardt et al. supra), or the anaerobically induced nirB promoter (Harbome et al. Mol. Micro., 6:2805-2813 (1992)
  • repressible promoters
  • the bacterial delivery system is safe and based on a non-toxic, attenuated Salmonella strain that has a partial deletion of the msbB gene. This deletion diminishes the TNF immune response to bacterial lipopolysaccharides and prevents septic shock. In another embodiment, it also has a partial deletion of the purI gene. This deletion makes the bacteria dependent on external sources of purines and speeds clearance from non-cancerous tissues (13). In mice, the virulence (LD 50 ) of the therapeutic strain is 10,000-fold less than wild-type Salmonella (72, 73). In pre-clinical trials, attenuated Salmonella has been administered systemically into mice and dogs without toxic side effects (17, 27).
  • the strain of bacteria is VNP20009, a derivative strain of Salmonella typhimurium. Deletion of two of its genes - msbB and purI -resulted in its complete attenuation (by preventing toxic shock in animal hosts) and dependence on external sources of purine for survival. This dependence renders the organism incapable of replicating in normal tissue such as the liver or spleen, but still capable of growing in tumors where purine is available. Further, insertion of a failsafe circuit into the bacterial vector prevents unwanted infection and defines the end of therapy without the need for antibiotics to remove the bacteria (e.g., salmonella). II.
  • DNA, RNA (e.g., a nucleic acid-based gene interfering agent) or protein may be produced by recombinant methods.
  • the nucleic acid is inserted into a replicable vector for expression.
  • the vector components generally include, but are not limited to, one or more of the following: an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence and coding sequence.
  • the gene and/or promoter may be integrated into the host cell chromosome or may be presented on, for example, a plasmid/vector.
  • Expression vectors usually contain a selection gene, also termed a selectable marker. This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will not survive in the culture medium.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media.
  • Expression vectors can contain a promoter that is recognized by the host organism and is operably linked to the nucleic acid sequence, such as a nucleic acid sequence coding for an open reading frame. Promoters are untranslated sequences located upstream (5') to the start codon of a structural gene (generally within about 100 to 1000 bp) that control the transcription of particular nucleic acid sequence to which they are operably linked. In bacterial cells, the region controlling overall regulation can be referred to as the operator.
  • Promoters typically fall into two classes, inducible and constitutive. Inducible promoters are promoters that initiate increased levels of transcription from DNA under their control in response to some change in culture conditions, e.g., the presence or absence of a nutrient or a change in temperature. A large number of promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the ⁇ -lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, hybrid promoters such as the tac promoter, and starvation promoters (Matin, A.
  • bacterial promoters are also suitable. Such nucleotide sequences have been published, thereby enabling a skilled worker to operably ligate them to a DNA coding sequence. Promoters for use in bacterial systems also can contain a Shine-Dalgarno (S.D.) sequence operably linked to the coding sequence. Construction of suitable vectors containing one or more of the above-listed components employs standard ligation techniques. Isolated plasmids or DNA fragments are cleaved, tailored, and re-ligated in the form desired to generate the plasmids required.
  • the expression vector is a plasmid or bacteriophage vector suitable for use in Salmonella, and the DNA, RNA and/or protein is provided to a subject through expression by an engineered Salmonella (in one aspect attenuated) administered to the patient.
  • plasmid refers to any nucleic acid encoding an expressible gene and includes linear or circular nucleic acids and double or single stranded nucleic acids.
  • the nucleic acid can be DNA or RNA and may comprise modified nucleotides or ribonucleotides and may be chemically modified by such means as methylation or the inclusion of protecting groups or cap- or tail structures.
  • One embodiment provides a Salmonella strain comprising a lysis gene or cassette operably linked to an intracellularly induced Salmonella promoter.
  • One embodiment provides an attenuated Salmonella strain comprising a lysis gene or cassette operably linked to an intracellularly induced Salmonella promoter.
  • the lysis cassette is Lysin E from phage phiX174, phage iEPS5, or lambda phage.
  • the promoter is a promoter for one of the genes in Salmonella pathogenicity island 2 type III secretion system (SPI2-T3SS) selected from the group SpiC/SsaB (accession no. CBW17423.1), SseF (accession no.
  • SpiC/SsaB accession no. CBW17423.1: 1 MSEEGFMLAV LKGIPLIQDI RAEGNSRSWI MTIDGHPARG EIFSEAFSIS LFLNDLESLP 61 KPCLAYVTLL LAAHPDVHDY AIQLTADGGW LNGYYTTSSS SELIAIEIEK HLALTCILKN 121 VIRNHHKLYS GGV (SEQ ID NO: 1) SseF (accession no.
  • Oncolytic Virus Oncolytic viruses are an emerging class of cancer therapeutics that offer the benefits of selective replication in tumor cells, delivery of multiple eukaryotic transgene payloads, induction of immunogenic cell death and promotion of antitumor immunity, and a tolerable safety profile that largely does not overlap with that of other cancer therapeutics, such as immune checkpoint inhibitors (ICIs) or chimeric antigen receptors (CARs).
  • ICIs immune checkpoint inhibitors
  • CARs chimeric antigen receptors
  • the first oncolytic virus immunotherapy to be approved by the FDA was T-VEC, which is used to treat metastatic melanoma.
  • T-VEC is a herpes virus that has been engineered to be less likely to infect healthy cells. OVs directly kill cancer cells.
  • OVs replicate inside cancer cells, causing them to burst and release materials that the immune system can recognize.
  • OVs can be modified to carry genes that boost the treatment's effectiveness.
  • OVs can be engineered to express immune regulators that enhance antitumor immunity.
  • Oncolytic viruses include, but are not limited to, DNA (ssDNA) viruses, double-stranded DNA (dsDNA) viruses, single-stranded RNA (ssRNA) viruses, double-stranded RNA (dsRNA) viruses or a combination thereof.
  • viruses for use in the invention can include, but are not limited to, adenoviruses, coxsackievirus, herpes simplex virus (HSV), maraba viruses, measles, Newcastle disease virus, picornavirus, reovirus, respiratory syncytial virus, vesicular stomatitis virus, parvoviruses, poxviruses, such as vaccinia virus (VACV) and myxoma virus (MYXV), minute virus of mice (MVM), talimogene laherparepvec (T-VEC, or Imlygic®), and/or poliovirus (such as PVS- RIPO). Any of these viruses can be used in the methods herein.
  • HSV herpes simplex virus
  • MYXV myxoma virus
  • MYXV minute virus of mice
  • T-VEC talimogene laherparepvec
  • Imlygic® Imlygic®
  • poliovirus such as PVS
  • Bacteria such as Salmonella, Clostridium and Bifidobacterium have a natural tropism for cancers, such as solid tumors.
  • Types of cancer that can be treated using the methods of the invention include, but are not limited to, solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, osteosarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, pancreatic ductal adenocarcinoma, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cyst
  • the subject is treated with radiation, immunotherapy and/or chemotherapy before, after or during administration of the bacterial cells described herein.
  • Administration includes administration of the attenuated bacteria, such as Salmonella, strains described herein and methods for preparing pharmaceutical compositions and administering such as well. Such methods comprise formulating a pharmaceutically acceptable carrier with one or more of the attenuated Salmonella strains described herein.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents such as ethylenediaminetetraacetic acid
  • buffers such as
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL TM (BASF; Parsippany, N.J.) or phosphate buffered saline (PBS). It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of other (undesired) microorganisms.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • a coating such as lecithin
  • surfactants for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients discussed above.
  • dispersions are prepared by incorporating the active compound into a vehicle which contains a basic dispersion medium and various other ingredients discussed above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously.
  • Oral compositions generally include an inert diluent or an edible carrier. For example, they can be enclosed in gelatin capsules.
  • the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules.
  • Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the bacteria are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • the attenuated Salmonella When administered to a patient the attenuated Salmonella can be used alone or may be combined with any physiological carrier.
  • the dosage ranges from about 1.0 c.f.u./kg to about 1x10 12 c.f.u./kg; optionally from about 1.0 c.f.u./kg to about 1x10 10 c.f.u./kg; optionally from about 1.0 c.f.u./kg to about 1x10 8 c.f.u./kg; optionally from about 1x10 2 c.f.u./kg to about 1x10 8 c.f.u./kg; optionally from about 1x10 4 c.f.u./kg to about 1x10 8 c.f.u./kg; optionally from about 1x10 5 c.f.u./kg to about 1x10 12 c.f.u./kg; optionally from about 1x10 5 c.f.u./kg to about 1x10 10 c
  • EXAMPLE The following examples are provided in order to demonstrate and further illustrate certain embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
  • Example I Introduction Many of the current problems associated with cancer treatment, e.g., metastatic disease and refractory tumors, could be overcome with microbial therapies.
  • Oncolytic viruses (OVs) have the potential to treat many tumors by directly lysing cancer cells and stimulating immune responses that eliminate cancer cells regardless of their location in the body (1-6). Despite this potential, oncolytic viruses have not been effective at treating internal solid tumors (5, 7-10). When injected systemically, OVs are cleared from the blood and do not effectively reach tumors (1, 5, 7-20). This problem could be overcome with a carrier with tumor specific tropism.
  • ID Salmonella intracellular delivering
  • ID Salmonella is a bacterial system recently developed to deliver macromolecules into cancer cells in tumors (31).
  • ID Salmonella contain a genetic circuit (PsseJ-lysE) that controls the release of plasmids and macromolecules (31). In tumors, Salmonella naturally invades cancer cells (31).
  • MMM Minute virus of mice
  • the prototype strain (MVMp) does not integrate into the genome, is non-infectious, and is non-pathogenic in humans (36, 38, 39).
  • NS1 is the initiator protein that creates a site-specific nick in the single-stranded DNA and serves as a helicase to initiate DNA replication (34, 38, 40, 42).
  • This DNA template is used to produce more single-stranded viral genomes, which are assembled in the nucleus into new virus particles (36, 43, 44).
  • Cell lysis kills cells and releases new viral particles that infect neighboring cells and initiates new rounds of infection (5, 10, 20, 45, 46). Because of its small size, the MVMp genome can be included in a standard cloning plasmid (47).
  • MVMp When mammalian cells are transfected with an MVMp-containing plasmid, they transcribe NS1 and produce viral particles, similar to natural infection (47, 48). Bacterial delivery of viral DNA is complicated by hairpins in the genomes of many viruses. These hairpins have a role in DNA replication, packaging, and viral infectivity. Palindromic sequences are not well tolerated in Salmonella and are subject to partial or complete deletion (49, 50). MVMp has two imperfect palindromes at the 3’ and 5’ termini that form hairpins in the DNA of the viral genome (32, 34, 41, 43, 47, 51-53). These palindromic regions are primers for replication and are used in virus formation and function (33, 41, 43, 47, 51-53).
  • homologous recombination When transformed into cloning strains of E. coli, homologous recombination forms site-specific deletions in the right-end hairpin of plasmids containing the MVMp genome (51). This deletion is prevented in SURE 2 (‘stop unwanted rearrangement events’ 2) E. Coli ( ⁇ recB, ⁇ recJ, and ⁇ sbcC), which is restriction minus, endonuclease deficient, and recombination deficient.
  • Salmonella contains two systems for repairing DNA damage and regulating homologous recombination: one for double-stranded breaks (DSBs) and another for single- stranded gap (SSG) repairs (54-56). These systems maintain genetic stability but also modify foreign DNA (54).
  • RecBCD A key component of the DSB repair system is RecBCD, which is a helicase- nuclease complex that initiates repair by unwinding double-stranded DNA (dsDNA) and creating a single-stranded tail for RecA to bind (54, 57-61).
  • RecA is a central protein in homologous recombination that compares and exchanges complementary DNA sequences (54, 60).
  • SbcB and SbcCD Two additional components of the DSB repair system are SbcB and SbcCD (62).
  • SbcCD is a nuclease that cleaves double stranded hairpins (59-61).
  • the primary component of SSG repair system is RecFOR (54, 57).
  • the bacterial vector must (1) preserve the viral genome and (2) deposit it intact into cancer cells. It was hypothesized that 1) DNA delivered by ID Salmonella is expressed by cancer cells, 2) removal of homologous recombination genes from Salmonella prevents hairpin deletion and stabilize the MVMp genome, 3) Salmonella delivery of the MVMp genome produces functional viral particles, and 4) the produced viral particles are infective and kill cancer cells. To test these hypotheses, seven strains of Salmonella were generated with deletions in six genes: recA, recB, recF, sbcB, sbcCD, and recJ. For H1PV and MH1PV sseJ knockouts were created in the strains which could stabilize the hairpins.
  • VDS Virus- delivering Salmonella
  • HRDS homologous-recombination- deficient Salmonella
  • HRDS homologous-recombination- deficient Salmonella
  • Fluorescence microscopy was used to measure the delivery of plasmid DNA to cancer cells with ID Salmonella.
  • Gentamicin invasion assay was used to determine the viability and invasiveness of the HRDS strains.
  • Cancer cells were treated with VDS to measure the production of NS1, the initiation of virus formation, and the cytotoxicity of the delivered virus. Finally, cells were retreated with conditioned media to quantify the infectiveness of released virions.
  • VDS By delivering OVs, VDS can expand the effectiveness of microbial therapy to include more solid tumors and be effective for a broader range of cancer patients.
  • Materials and Methods Bacterial culture All bacterial cultures (SURE 2, Salmonella) were grown in LB (10 g/L sodium chloride, 10 g/L tryptone, and 5 g/L yeast extract). Resistant strains of bacteria were grown in the presence of 100 ⁇ g/mL of carbenicillin, 33 ⁇ g/mL chloramphenicol, or 50 ⁇ g/mL of kanamycin.
  • Bacterial strains and plasmids The MVMp plasmid was provided by Dr. Peter Tattersall (Yale School of Medicine, New Haven, CT).
  • the plasmid is ampicillin resistant and has a ColE1 origin of replication.
  • the ptac-GFP plasmid contains green fluorescent protein (GFP) under control of the constitutive bacterial promoter ptac and a Lysin gene E from ⁇ X174 bacteriophage under the control of intracellular responsive Salmonella promoter PsseJ (31).
  • This plasmid is chloramphenicol resistant and has a p15A origin of replication.
  • HEK-293T Human embryonic kidney cells
  • RRID:CVCL_0063 human osteosarcoma cells
  • U2OS RRID:CVCL_0042
  • KPCY murine pancreatic ductal adenocarcinoma cells
  • DMEM low glucose Dulbecco’s modified Eagle medium
  • FBS fetal bovine serum
  • Murine mammary carcinoma cells (4T1, RRID:CVCL 0125) were maintained in Roswell Park Memorial Institute (RPMI 1640; Sigma Aldrich, St. Louis, MO) medium supplemented with 2 g/l of sodium bicarbonate and 10% FBS at 37 °C and 5% CO2.
  • RPMI 1640 Roswell Park Memorial Institute
  • FBS FBS
  • DMEM Roswell Park Memorial Institute
  • FBS FBS
  • GFP expression from a eukaryote promoter after delivery with Salmonella To measure plasmid and gene delivery, two plasmids for delivery and eukaryotic GFP expression were transformed into parental Salmonella ( ⁇ msbB, ⁇ purI, ⁇ xyl) and termed as ID- CMV-Sal.
  • the GFP expression plasmid contained the gene for green fluorescent protein (GFP) under control of the cytomegalovirus (CMV) mammalian expression promoter.
  • the delivery plasmid contained the PsseJ-lysE circuit.
  • Control bacteria were parental Salmonella transformed with CMV-GFP but not the PsseJ-lysE lysis plasmid. Salmonella strains were grown to an OD 600 between 0.8-1.0 and added to cultures of 4T1 cells for 2 hours. During this period, the bacteria invaded into the cancer cells.
  • strains A: ⁇ recA; B: ⁇ recB; F: ⁇ recF; CD: ⁇ sbcCD; BB’F: ⁇ recB, ⁇ sbcB and ⁇ recF; BCDJ: ⁇ recB, ⁇ sbcCD and ⁇ recJ; and BB’CDF: ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF, CDS: ⁇ sbcCD ⁇ sseJ, BB’CDFS: ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF ⁇ sseJ).
  • Salmonella was transformed with pkd46 (Yale CGSC E. Coli stock center), grown to OD6000.1, and induced with 10 mM arabinose. Once the OD600 reached 0.6-0.8, the bacteria were centrifuged at 3000 ⁇ g for 15 minutes. The pellet was washed twice with ice-cold water. A PCR product for the in-frame deletion of the gene using specific primers (Supplemental Table S1) was amplified from the pkd4 plasmid (accession AY048743.1) containing the FRT- KAN-FRT sequence. The PCR product, which contains 50 bp homology for the gene, was transformed into Salmonella using electroporation.
  • the recovery was plated on kanamycin plates (50 ⁇ g/ml) and grown overnight.
  • the colonies were screened for deletion by performing a colony PCR of the junction sites of the inserted PCR amplified products. Colonies with a successful knockout were grown overnight at 43 °C to eliminate pkd46.
  • the antibiotic resistance was removed by transforming the pcp20 plasmid into the deleted Salmonella. The colonies were grown overnight at 43 °C to eliminate pcp20. After the elimination of the plasmid, the overnight culture was diluted to approximately 100 CFU/ml and plated on hektoen plates. The colonies which were dark green were confirmed as Salmonella.
  • the colonies were sequentially streaked on hektoen, kanamycin, and chloramphenicol agar to verify the removal of the antibiotics. Hairpin stability and bacterial growth To test hairpin stability, the MVMp plasmid was transformed into each knockout strain by electroporation. Electroporation was performed in 1 mm cuvettes at 1800 V and 25 ⁇ F with a time constant of 5 msec.
  • Plasmids were extracted using the ZymoPURE plasmid miniprep kit per the manufacturer’s instructions (Irvine, CA) and sent for sequencing (Massachusetts General Hospital CCIB DNA core facility) To further test hairpin stability, isolated plasmids were digested with SSPI restriction enzyme (New England Biolabs, Ipswich, MA). Strains BB’CDF and CD strains were transformed with a plasmid containing the PsseJ-lysE cassette to deliver the MVMp plasmid and were termed virus-delivering Salmonella (VDS-B and VDS- C, respectively).
  • Salmonella strains were transformed with a plasmid containing EGFP under a constitutive promoter and grown to an OD600 between 0.8-1.0. Salmonella were added to the cell cultures for 2 hours. Following this invasion period, cultures were rinsed with 1X PBS five times and incubated in RPMI with 2 g/L sodium bicarbonate, 10% FBS, and 50 ⁇ g/mL gentamicin. Cells were stained with 10 ⁇ g/mL Hoechst 33342 (Invitrogen, Waltham, MA). Multiple regions in a well were randomly selected, and 30 cells/regions were randomly selected and scored based on EGFP expression to quantify the percentage of the cells that were invaded.
  • VDS-B and VDS-C were added to the HEK- 293T cells at an MOI of 20. After two hours of invasion, extracellular bacteria were removed with gentamicin.
  • the positive control was the MVMp plasmid transfected into na ⁇ ve cells using Lipofectamine 3000 per manufacturer’s protocol (Thermo Fisher, Waltham, MA). Thirty-six hours after infection or transfection, cells were scraped from the flasks. Cells were collected and pelleted by centrifugation at 1000g for 5 minutes. Cells were suspended in Laemmli SDS 6x sample buffer (Alfa Aesar, Haverhill, MA).
  • VDS-B or VDS- C Bacteria (VDS-B or VDS- C) were added to cultures of 4T1 cells at an MOI of 100. Strains without the MVMp plasmid (CD or BB’CDF) were negative controls. The positive control was 4T1 cells transfected with the MVMp plasmid using Lipofectamine 3000. After two hours of invasion, bacteria were cleared with gentamicin. Seventy-two hours after infection/transfection, the cells were fixed with 10% formaldehyde in water for 1 hour at room temperature. Dead cell plaques were stained with crystal violet staining, dead cells were stained with ethidium homodimer and cell viability was determined by MTT assay.
  • crystal violet staining the formaldehyde was removed and the cells were stained with 1% crystal violet stain (Acros Organics, Waltham, MA). Cells were incubated in crystal violet for 15 minutes and rinsed with water. Cells were imaged on a Zeiss Axio Observer Z.1 microscope using a 5x objective lens. Images were tiled to image the entire surface. The area of unstained cells was analyzed using MATLAB. To quantify cell death, 1 ⁇ g/mL ethidium homodimer (Invitrogen, Waltham, MA) was added to cells 72 hours after infection or transfection. Live cell images were acquired every 10 minutes. The experiment was performed in triplicate, and death was determined in three areas per well.
  • the extent of cell death was determined in ImageJ by measuring the area with red fluorescence compared to the area covered by cells at the time of invasion/transfection.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; Themo Fisher, Waltham, MA
  • VDS VDS-B
  • BB BB
  • CDF bacterial negative control
  • ID Salmonella was transformed with a plasmid containing GFP under control of the cytomegalovirus (CMV) promoter. This promoter only initiates transcription in eukaryotic cells (65). Salmonella with CMV-GFP did not express GFP. In addition to CMV-GFP, ID-CMV-GFP Salmonella contained the PsseJ-lysE circuit, which induces lysis after cell invasion (31). Control Salmonella contained CMV-GFP, but not PsseJ- lysE. When incubated with murine 4T1 mammary cancer cells, ID-CMV-GFP produced GFP (arrows, FIG 2A).
  • CMV cytomegalovirus
  • ID-CMV-GFP produced significantly more GFP (P ⁇ 0.001, FIG. 2B). This difference indicates that the lysis circuit is needed for delivering plasmids into cancer cells. GFP expression also indicates that after bacterial lysis, released plasmids were transported to the nucleus and delivered genes were translated by recipient cells.
  • Parental Salmonella disables MVMp replication When not modified, parental Salmonella ( ⁇ msbB, ⁇ purI, ⁇ xyl) deletes 97 base pairs from the right-end hairpin of MVMp (FIG. 3). The genome of MVMp contains two palindromes at either end that are primers for replication and are needed for virus formation.
  • the genome for MVMp is on a plasmid that also contains a ColE1 prokaryotic origin of replication, and ampicillin resistance (FIG.3A). Transcription of MVMp is initiated by the P4 promoter within its genome in a mammalian nucleus (44). Replicating this plasmid in SURE 2 recombination-deficient E. coli ( ⁇ recB, ⁇ recJ, and ⁇ sbcC), maintained both hairpins (FIG. 3B). SSPI digestion of the MVMp plasmid produced three fragments: 5078, 1395 and 732 bp (FIG. 3B, left). The smallest fragment contains the right-end hairpin.
  • CDS For H1PV and MH1PV plasmids two more strains with sseJ knockout were created in CD and BB’CDF and were termed as CDS: ( ⁇ sbcCD ⁇ sseJ), BB’CDFS ( ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF ⁇ sseJ).
  • BB CDFS
  • ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF ⁇ sseJ When the MVMp plasmid was replicated in each of these seven bacterial strains, four of the strains deleted 97 bp from the right-end hairpin (A, B, F and BB’F), as shown by SSPI restriction digest (FIG. 4A) and DNA sequencing (FIG. 4B).
  • FIGS.4A, B Three of the knockout stains maintained the hairpin (CD, BCDJ and BB’CDF; FIGS.4A, B).
  • the MVMp plasmid was replicated in CD and BB’CDF, purified, and transfected into HEK-293T cells. After transfection, all of these cells formed NS1 (FIG. 4C), demonstrating the initiation of virus formation. To show the durability of virus replication, the MVMp plasmid was maintained in CD, BB’CDF and SURE 2 for three passages.
  • both CD and BB’CDF maintained the full-length plasmid and the right- end hairpin (similar to SURE 2, FIG. 4D).
  • Deletion of homologous recombination genes does not impair growth or invasion
  • deletion of homologous recombination genes did not affect bacterial growth or invasiveness into cancer cells (FIG.5).
  • the growth of strain CD and BB’CDF were comparable with parental Salmonella (FIG. 5A).
  • the growth of strain BCDJ was impaired.
  • the deletions in BCDJ delayed growth by more than six hours (FIG. 5A).
  • the density of BCDJ was 4.3 times lower than parental Salmonella (P ⁇ 0.0001, FIG. 5B).
  • the densities of CD and BB’CDF were comparable to parental Salmonella at nine hours (FIG.5B).
  • the CD and BB’CDF strains invaded into cancer cells at comparable rates to parental Salmonella (FIG.5C-F).
  • the Salmonella strains (CD, BB’CDF, BCDJ and parental) were co-cultured with 4T1 murine mammary cancer cells for two hours and extracellular bacteria were cleared with gentamicin. After four hours, bacteria were inside cells (arrows) for the CD, BB’CDF and parental strains (FIG. 5C).
  • VDS-B BB’CDF
  • VDS-C VDS-C
  • both strains of VDS were applied to cultures of 4T1 cells (FIG. 6A). After two hours of co-culture, extracellular bacteria were removed with gentamicin (FIG.6A). After a further 36 hours to allow for plasmid release, transport to the nucleus and gene expression, the cells cultured with VDS-B produced the viral NS1 protein (FIG. 6B). NS1 production was comparable to positive control cells that were directly transfected with the MVMp plasmid using lipofectamine (FIG. 6B).
  • the bacterial control was the BB’CDF strain, which did not contain the MVMp plasmid but was transformed with the delivery (PsseJ-lysE) plasmid, and had the same knockouts ( ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF) as VDS-B. Seventy-two hours after bacterial invasion, cells treated with VDS-B formed virus-infected, dead cell plaques (FIG. 6C). The area of the plaques was significantly more than bacterial controls (P ⁇ 0.05, FIG. 6D). Over time, VDS-B caused death similarly to transfection controls (arrows in FIGS. 6E-F).
  • VDS-C VDS-C
  • strain HRDS-CD its bacterial control
  • FIG.6I Direct transfection of MVMp caused more cell death than VDS-C (P ⁇ 0.001, FIG. 6I).
  • Lack of virus formation and undetectable cell death shows limited utility for VDS-C (compared to VDS-B).
  • VDS creates functional virus particles that infect new cells Delivery of MVMp by VDS forms functional and infective virions (FIG.7).
  • VDS was applied to cultures of 4T1 cancer cells for two hours (FIG. 7A). After this time for invasion, the cells were washed and fed with complete media containing gentamicin.
  • VDS-mediated cell death is independent of cancer type When applied to multiple cancer cell types, VDS induced the formation of cytotoxic viral particles that kill cells and infect neighboring cells (FIG.8).
  • VDS-B was applied to HEK-293T human embryonic kidney cells, U2OS human osteosarcoma cells, and KPCY murine pancreatic ductal adenocarcinoma cells (FIG. 8A). A similar procedure was used as FIG. 6A.
  • NS1 was produced by cells that were directly transfected with the MVMp plasmid using lipofectamine (FIG. 8A). When VDS-B was applied for two hours, these cells also produce NS1 (FIG. 8A). VDS-C did not produce NS1 in any of the cell types.
  • the delivery of MVMp with VDS killed osteosarcoma and pancreatic ductal adenocarcinoma cells (FIG. 8B). At 72 h after infection of KPCY cells, VDS-B significantly reduced viability compared to bacterial controls (strain BB’CDF; P ⁇ 0.01, FIG.8B).
  • VDS-B In U2OS cell, MVMp delivered by VDS-B also significantly reduced viability compared to bacteria controls (P ⁇ 0.0001, FIG.8B). As with results in FIG 6, VDS-C did not induce death in either of these cell lines.
  • VDS-C When applied to osteosarcoma and pancreatic ductal adenocarcinoma cells, VDS produced infective viral particles (re-infection, FIG.8C).
  • FIG. 7A VDS was added to cell cultures for 2 hours, extracellular bacterial were killed with gentamicin, and conditioned media was added to cultures of HEK-293T cells. After 48h in conditioned media, the HEK-293T cells that received media from VDS-B cultures produced NS1 (FIG. 8C).
  • Virus was also present in the culture media from cells transfected with the MVMp plasmid (FIG. 8C). No virus was produced by cultures treated with VDS-C. Viewed together, these data demonstrate that MVMp delivery by VDS resulted in the production of viable virus particles that kill infected cancer cells, independent of cell type. Discussion A bacterial vector was created that delivers oncolytic viruses into cancer cells. The genome for the virus (MVMp) was contained on a plasmid that the bacteria propagated without deleting palindromic sections that are essential for virus infection and replication (FIGS. 3-5). When these virus-delivering Salmonella (VDS) encounter cancer cells, they invade, and deposit the plasmid into the cellular cytoplasm.
  • the cell expresses the viral genes (FIGS. 2, 6).
  • Production of initiating proteins begins the process of forming and assembling viral particles in the cells (FIG.6). Once formed, these virions kill the invaded cancer cells (FIG.6) and spread into the environment. There the viruses invade and kill na ⁇ ve cells (FIG.7).
  • the release of virions by infected cells perpetuates and amplifies the infection.
  • the results show that deletion of homologous recombination genes enables Salmonella to carry and maintain the MVMp genome (FIGS. 3-5). This parvovirus genome contains a right-end hairpin that is unstable in Salmonella (FIG. 3). VDS was designed to prevent this deletion.
  • VDS delivers functional viruses to multiple cancer types, including breast carcinoma, pancreatic carcinoma, and osteosarcoma (FIG. 8).
  • VDS was used to deliver MVMp, which is a single-stranded DNA (ssDNA) virus.
  • VDS could also deliver double- stranded DNA (dsDNA), single-stranded RNA (ssRNA) and double-stranded RNA (dsRNA) viruses.
  • ssDNA single-stranded DNA
  • ssRNA single-stranded RNA
  • dsRNA double-stranded RNA
  • VDS enables oncolytic virus treatment of solid tumors.
  • VDS After systemic injection, VDS would preferentially accumulate in tumors over other organs, similar to other Salmonella vectors (31, 67). Delivery of the viral genome would produce infective virus particles that can initiate successive rounds of infection (FIG. 7) and mediate a potent bystander effect. Salmonella delivery would increase the delivery of viruses to the tumor microenvironment. Combining the benefits of Salmonella delivery and oncolytic virus therapy can provide an effective therapy for many cancers.
  • Salmonella delivers functional H1PV virions that are cytotoxic to liver cancer cells: Two strains of homologous recombination deficient Salmonella, BB’CDF ( ⁇ recB, ⁇ sbcB, ⁇ sbcCD and ⁇ recF) and CDS ( ⁇ sbcCD and ⁇ sseJ) maintained the H1PV right-end hairpin. Two test the stability of the plasmid, SSPI and EcoRI restriction digest was performed and the size of the two bands were same as from the plasmid maintained in the SURE 2 strain (FIG. 14A). To test whether, the plasmid from these strains could generate functional virus, plasmid was transfected into HEK-293T cells.
  • BB’CDF and CDS strains of Salmonella was transformed with the H1PV plasmid and a plasmid containing the intracellular delivery circuit.
  • the strains containing both the plasmid were termed as H1PV VDS-B when transformed in BB’CDF and H1PV VDS-C when transformed into CDS.
  • H1PV VDS-B and VDS-C were co-cultured with HUH7 (human liver cancer cells) at a Multiplicity of infection (MOI) of 100.2 hours post infection, the cells were washed 3X with PBS and added new media with gentamycin.36 hours post cells were scraped and looked for NS1 protein. Both the strains of Salmonella were able to deliver the viral plasmid in human liver cancer cell similar to the transfection of the viral plasmid. (FIG. 14C).
  • H1PV plasmid delivery by VDS-B at a MOI of 100 resulted in the generation of NS1 protein (FIG. 14D).
  • BB’CDF was better at delivering the viral plasmid in the cancer cells than the CDS. Therefore, BB’CDF strain was carried forward for all the in vivo experiments. This delivery resulted in a significant decrease in the cell viability in all the above cell lines.
  • Engineered Salmonella delivers H1PV in tumors that creates functional virions: 1*10 7 CFU of H1PV VDS-B was injected intravenously (IV) into tumor free mice. The mice injected regained their weights in three days after injection (FIG.15A). After 7 days blood was collected from mice injected with H1PV VDS-B and saline and the serum was analyzed for complete cytokine panel.
  • mice injected with H1PV VDS-B and saline There was no difference in the cytokine levels between mice injected with H1PV VDS-B and saline (FIG.15B).
  • 4*10 7 CFU of bacteria was injected in mice intratumorally (IT) in subcutaneous liver hepatoma (Hepa1-6) tumors between 250-450mm 3 .
  • 5 days post injection tumor was harvested, processed, and stained for viral protein, NS1 on a western blot (FIG.15C).
  • mice with tumors injected with 4*10 7 CFU of H1PV VDS-B were harvested and processed.
  • Functional virions were harvested from the tumors and the supernatant containing the virus was put on HEK-293T cells. 72 hours post infection, the HEK-293T cells were analyzed for the NS1 viral protein (FIG. 15E).
  • H1PV VDS-B had NS1 band when the supernatant from these tumors were put on HEK-293T cells whereas the supernatant from tumors injected with saline and H1PV VDS-C did not show up a NS1 band on the immunoblot (FIG.15F).
  • BB’CDF was used as a control and the same amount was injected.
  • the tumors were measured every three days until the maximum tumor burden was reached (1000mm 3 ). Injections were given until the last control died, a total of six injections were given every six days to both control and experimental group (FIG.16A).
  • Intravenous (IV) delivery of H1PV VDS-B is as efficacious as intratumoral (IT) delivery: 4*10 7 CFU of H1PV VDS-B or BB’CDF was injected IV in Hepa1-6 tumors between the size of 50-180mm 3 .6 injections were given, six days apart and tumors were measured every three days until maximum tumor burden was reached (FIG.17A).
  • the efficacy was stopped when the first control mice died.
  • the mice in experimental also survived significantly longer than the mice in the control group (P ⁇ 0.05) (FIG.17E).
  • IV injected mice were compared with IT injected mice of H1PV VDS-B there was no difference in the tumor volumes (FIG.
  • H1PV delivery by Salmonella causes reduction in tumor weights, cancer cells and increases immunostimulatory innate immune cells: 4*10 7 CFU of H1PV VDS-B was injected IT in Hepa1-6 tumor model with BB’CDF as a control. 3 injections were given, six days apart and five days after the last injection mice were euthanized and tumor were harvested (FIG.18A).
  • the tumor was then stained for Epcam positive cells which is pan cancer cell marker. There was over a 2-fold reduction in the Epcam positive cells in mice injected with H1PV VDS-B as compared to mice injected with BB’CDF (P ⁇ 0.05) (FIG.18D). The tumor was also stained for all the innate cells; Natural killer cells (Npk46 marker), Macrophages (F4/80 marker). There was a significant increase in the NK cells (P ⁇ 0.05) (FIG. 17E) and Macrophages (P ⁇ 0.05) (FIG. 18F) in the experimental group as compared to the control group.
  • the H1PV VDS-B tumors also have a significant decrease in the CD45 + cells as compared to the control tumors which is a pan immune cell marker (P ⁇ 0.05) (FIG.18G). Since all the other cells (NK cells, Macrophages and CD3 + cells) had a significant increase in mice injected with H1PV VDS-B. This reduction could be attributed to the reduction in neutrophils. So, there was a significant reduction in the neutrophil population in the experimental group as compared to the control group (P ⁇ 0.01) (FIG. 18H). The increase in neutrophils is associated with tissue damage and poor prognosis of the tumor.
  • H1PV delivery be engineered Salmonella activates adaptive immunity and generates tumor memory: 4*10 7 CFU of H1PV VDS-B was injected IT with Hepa1-6 tumor model with BB’CDF as a control.
  • FIG.19A 3 injections were given, six days apart and five days after the last injection mice were euthanized and tumor were harvested (FIG.19A).
  • the tumor was processed and stained for pan T cells (CD3 marker), cytotoxic T cells (CD8 marker), Helper T cells (CD4 marker), Regulatory T cells (CD25 marker).
  • Mice injected with H1PV VDS-B had over a 4-fold increase in the CD3 + cells as compared to the bacterial control group (P ⁇ 0.05) (FIG. 19B).
  • the cells were then stained for CD8 T cells and regulatory T cells (FIG. 19C). There was a 12-fold increase in the ratio of CD8 + to CD4 + CD25 + cells (P ⁇ 0.05) (FIG. 19D).
  • the virus delivery can recruit both virus specific T cells and cancer specific T cells. To check whether the T cells recruited in the tumor microenvironment were cancer specific.
  • the mice were injected with H1PV VDS-B three times, six days apart with 4*10 7 CFU. Five days post the last injection, spleens were then extracted from the mice. The spleen was processed and the splenocytes were then injected intraperitonially (IP) in the na ⁇ ve mice. Two weeks post the injection of splenocytes na ⁇ ve mice were rechallenged with 2.5 million Hepa1-6 cells (FIG. 19I). To determine the take rate of the Hepa1-6 cells, a million cells were injected in the na ⁇ ve mice.
  • mice not injected with splenocytes derived from H1PV VDS-B mice tumors took in 75% mice of the mice whereas the mice injected with the splenocytes no mice grafted tumors (FIG.19J).
  • the tumors injected were monitored for 66 days. All the tumors in the mice injected with saline grew while the mice injected with the splenocytes had pin pricks but then regressed and the mice remained tumor free for 66 days (FIG.19K).
  • Engineered Salmonella colonize tumor and is safe 1*10 7 CFU BB’CDF strain of Salmonella with the intracellular delivery plasmid was injected intravenously in the mice having a Hepa1-6. 72 hours post injection tumor along with liver, spleen, lungs, kidney, heart, and brain were harvested to determine the colonization of bacteria in different organs (FIG.20A).
  • Engineered Salmonella colonized tumors significantly higher than any other healthy organs (FIG. 20B). The colonization in the tumors was over 50 million-fold higher than in liver and spleen (FIG.20B).
  • BB’CDF and saline There was no difference in the cytokine levels between BB’CDF and saline (FIG.20F). There was a significant difference in the levels of IL-6 (P ⁇ 0.05) (FIG.20G) and TNF ⁇ (P ⁇ 0.01) (FIG. 20H) between BB’CDF and parental Salmonella, the two most important cytokines responsible for cytokine storm.
  • MTD maximum tolerable dose
  • BB’CDF and parental Salmonella strain were injected IV in mice. 5*10 7 CFU of BB’CDF bacteria was very well tolerated in mice with weight loss of around 10% but with the parental strain the weight loss was around 15% (FIG. 20I). After 7 days the mice were rechallenged with another same dose of the bacteria.
  • mice After the second dose of parental Salmonella, mice died whereas mice injected with BB’CDF regained their weights after 3 days of injection (FIG. 20I). Mice were then injected with 1*10 8 CFU of BB’CDF strain. After the first injection, mice lost around 12% of their body weights but after second injection they lost about 18% of their body weights (FIG.20I). Taken together, all these results indicate that the engineered Salmonella strain colonizes tumors and clears out of healthy organs better than the parental strain. The strain is also significantly safer than the parental strain.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Provided herein are methods and compositions for treating cancer. One composition includes an engineered bacterial cell comprising: a) a lysis gene or lysis cassette operably linked to an intracellularly induced Salmonella promoter; b) one or more of the bacterial cell genes selected from the group consisting of recA, recB, recF, sbcB, sbcCD, red, sseJ or any combination thereof are knocked out; and c) a nucleic acid sequence coding for an oncolytic virus genome.

Description

DELIVERY OF ONCOLYTIC VIRUSES WITH ENGINEERED SALMONELLA PRIORITY This application claims the benefit of the filing date of U.S. provisional application No. 63/607,690, filed on December 8, 2023, the disclosures of which is incorporated by reference herein in its entirety. GOVERNMENT SUPPORT This invention was made with government support under grant nos. R01CA188382 and R21CA293765 awarded by The National Institutes of Health and grant numbers W81XWH1910602 and HT94252310067 awarded by the Department of Defense. The government has certain rights in the invention. BACKGROUND Cancer is generally characterized by an uncontrolled and invasive growth of cells. These cells may spread to other parts of the body (metastasis). Conventional anticancer therapies, such as surgical resection, radiotherapy and chemotherapy, can be effective for some cancers/patients; however, they are not effective for many cancer sufferers. Thus, further medical treatments are needed. The role of bacteria as an anticancer agent has been recognized for over 100 years, and many genera of bacteria, including Clostridium, Bifidus, and Salmonella, have been shown to accumulate in tumor tissue and cause regression. The use of Salmonella typhimurium to treat solid tumors began with the development of a nonpathogenic strain, VNP20009. Well-tolerated in mice and humans, this strain has been shown to accumulate (>2000-fold) in tumors over the liver, spleen, lung, heart and skin, retarding tumor growth between 38-79%, and prolonging survival of tumor-bearing mice. In initial clinical trials, S. typhimurium was found to be tolerated at high dose and able to effectively colonize human tumors. SUMMARY As therapies, oncolytic viruses regress tumors and have the potential to treat hard-to- treat and late-stage cancers. Despite the promise of this approach, poor delivery limits their efficacy treating internal solid tumors. To address this issue, virus-delivering Salmonella (VDS) was developed to carry oncolytic viruses into cancer cells. In one embodiment, the VDS strain contains the PsseJ-lysE delivery circuit and has deletions in four homologous recombination genes (ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF) to preserve hairpins in the viral genome with a role in replication and infectivity. VDS delivered the genome of a virus to multiple cancers, including breast, pancreatic carcinoma, and osteosarcoma. Viral delivery produced functional viral particles that were cytotoxic and infective to neighboring cells. The release of mature virions initiated new rounds of infection and amplified the infection. Using Salmonella for delivery circumvents the limitations of oncolytic viruses and provides a novel therapy for cancer. BRIEF DESCRIPTION OF THE DRAWINGS The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which: FIG.1. Bacterial delivery of viruses into the cancer cells. (1) VDS, carrying the genome of MVMp, invades cancer cells. (2) Once intracellular, Salmonella lysis releases the viral plasmid into Salmonella-containing vacuoles (SCVs). SCV breakdown releases the plasmid into the cytoplasm. (3) After transport into the nucleus, cellular machinery converts the viral DNA into double-stranded DNA that is transcribed and translated to form structural (i.e., capsid) and non-structural proteins. (4) The double-stranded DNA is also a template for synthesizing new viral genomes. (5) The virus progeny is assembled in the nucleus. (6) The viral infection causes cell lysis, releasing new virions that infect the neighboring cells and initiate new rounds of infection. FIGS.2A-2B. ID Salmonella delivers encoding plasmids into cells. A) Administration of ID Salmonella with a eukaryotic-expression plasmid (CMV-GFP) and the intracellular delivering gene circuit (PsseJ-lysE) induced GFP expression (left, arrows). Bacteria (ID-CMV- GFP) were co-cultured with 4T1 cancer cells for two hours. Extracellular bacteria were cleared with 50 μg/mL gentamicin. Live images were acquired 24 hours after invasion. There was no GFP expression in parental Salmonella containing only the CMV-GFP plasmid but not the lysis circuit (right). B) Bacteria with PsseJ-lysE (ID-CMV-GFP) induced significantly more GFP expression than control CMV-GFP bacteria (***, P< 0.001). FIGS. 3A-3D. Parental Salmonella deleted the right-end hairpin and disabled viral replication. A) The MVMp plasmid contains the full MVM genome including the left- and right-end hairpins. This plasmid replicates in Salmonella and produces viruses when in transfected into mammalian cells. B) An SSPI restriction digest of the MVMp plasmid produced three fragments (left), 5078, 1395 and 732 bp, when maintained in SURE 2. Propagation in parental Salmonella deleted 97 bp from the right hairpin, reducing the third fragment to 635 bp. C) After sequencing, the MVMp plasmid that was propagated in SURE 2 (top) contained the full MVM genome. Propagation in parental Salmonella deleted 97 bp from the center of the palindrome in the right-end hairpin (bottom). D) The NS1 protein (83 kDa) was produced when HEK-293T cells were transfected with MVMp after propagation in SURE 2. No NS1 was produced in cells transfected with MVMp propagated in parental Salmonella. FIGS. 4A-4D. Deletion of homologous recombination genes maintains the right-end hairpin of MVMp. A) SSPI restriction digest of the MVMp plasmid, when maintained in (left to right) SURE 2 (lane 1), parental Salmonella (lane 2), A-HRDS (lane 3), B-HRDS (lane 4), F-HRDS (lane 5), BB’F (lane 6), CD (lane 7), BB’CDF (lane 8), BCDJ (lane 9). Strains CD, BB’CDF and BCDJ maintained the complete right-end hairpin similar to SURE 2. All of the other strains had a 97 bp deletion resulting in a shorter third fragment (635 bp). B) The sequence of plasmids maintained in parental Salmonella, or strains A, B, F, and BB’F had a 97 bp deletion in the center of the palindrome of the right-end hairpin. The sequence of plasmids maintained in strains CD, BB’CDF and BCDJ had intact right-end hairpins. C) After HEK- 293T cells were transfected with MVMp plasmids that were maintained in SURE 2, BB’CDF and CD, the cells produced NS1 protein (83KDa). Uninfected cells did not produce NS1. D) The MVMp plasmid was maintained in SURE 2, BB’CDF and CD for three generations. For each passage, the plasmid maintained the hairpin for all Salmonella strains. FIGS.5A-5F. Deletion of homologous recombination genes does not impair growth or cell invasion. A) Bacterial growth was measured for 16 hours for strains BB’CDF, CD, BCDJ and parental Salmonella. Two strains (BB’CDF and CD) had similar growth patterns compared to parental Salmonella. The growth of BCDJ was delayed by six hours. B) At nine hours, the bacterial density of BCDJ was significantly lower compared to parental Salmonella (****, P < 0.0001). Strains BB’CDF and CD had similar densities to parental Salmonella. C) After two hours of invasion, the number of intracellular bacteria (green, arrows) for BB’CDF (top right) and CD (bottom left) was similar to parental Salmonella (top left). Incubation with strain BCDJ (bottom right) produced less intracellular bacteria. D-F) There was no significant difference in the invasion levels of BB’CDF (D) or CD (E) compared to parental Salmonella. Invasion levels were significantly compromised for the BCDJ strain (F) compared to parental Salmonella (P < 0.05). FIGS. 6A-6I. VDS delivers functional virions that kill cancer cells. A) To measure virus delivery and induced lysis, VDS was applied to cultures of cancer cells (top). Murine mammary carcinoma cells (4T1) were incubated (blue arrows) with VDS-B or bacterial controls (BB’CDF). VDS-B contains both the MVMp and PsseJ-lysE plasmids; BB’CDF contains only the PsseJ-lysE plasmid. Similarly, VDS-C was compared to strain CD, its bacterial control. After two hours, external bacteria were removed with gentamicin (green arrow). In parallel, positive control cells were directly transfected with MVMp using lipofectamine (bottom). After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using crystal violet, ethidium homodimer and MTT. B) After VDS-B invaded cancer cells in culture, the cells produced viral NS1, similar to direct transfection with the MVMp plasmid. Cell invaded with VDS-C did not produce NS1. C) Cells delivered MVMp with VDS-B formed dead-cell plaques (arrow). Cells were stained with crystal violet. Bacterial infection alone formed few plaques. D) The area of cell death area was significantly higher in cells invaded with VDS-B than in cells invaded with BB’CDF (*, P < 0.05). E) Over time, treatment with VDS-B killed more cancer cells than bacterial controls. At 60 hours after invasion (or transfection), more dead cells (arrows, ethidium homodimer+) started to appear in cultures treated with VDS-B or directly transfected with MVMp. F) With time, the area of cell death increased for VDS-B and MVMp transfection but remained constant for BB’CDF. G) Death caused by VDS-B was greater than BB’CDF (*, P < 0.05). H) Cell viability, measured by MTT assay, decreased for VDS-B and MVMp transfection compared to BB’CDF. I) VDS-C did not increase cell death compared to CD. FIGS. 7A-7C. VDS delivers function virions that spread to neighboring cells. A) To measure the infectivity of the produced virus particles, conditioned media from VDS-treated cells was applied to cultures of naïve cells. Cancer cells (4T1) were incubated (blue arrows) with VDS-B or bacterial controls (BB’CDF) and external bacteria were cleared after two hours with gentamicin (green arrow, top). Positive controls were cells directly transfected with the MVMp plasmid using lipofectamine (bottom). After 72 hours, the culture medium was collected and added to fresh media at a 40:60 ratio. Addition of 50 μg/mL gentamycin prevented bacterial growth. This conditioned medium was added to naïve cultures of (1) HEK- 293T cells to measure virus formation, and (2) 4T1 cancer cells to measure cytotoxicity. B) Secondary cultures of HEK-293T cells that received media (and virus) from primary cultures treated with VDS-B (or transfected with MVMp) produced NS1. Uninfected controls and primary cultures treated with VDS-C did not produce NS1. C) Virions in conditioned media from VDS-B cultures killed more 4T1 cells than bacterial controls (BB’CDF) that did not produce virus (***, P <0.001). Death was equivalent to viruses from directly transfected cells. FIGS.8A-8C. Salmonella mediated delivery of MVMp is independent of cell type. A) Treatment with VDS-B produced NS1 when applied to human embryonic kidney cells (HEK- 293T), human osteosarcoma cells (U2OS), and murine pancreatic ductal adenocarcinoma cells (KPCY). No NS1 was expressed in any of the cell lines after being treated with VDS-C. B) Treatment with VDS-B significantly decreased viability in KPCY (**, P < 0.01) and U2OS (****, P < 0.0001) cells, compared to bacterial controls (BB’CDF). C) Re-infection of HEK- 293T cells with conditioned media from primary of cultures of KPCY and U2OS cells that were treated with VDS-B produced NS1. Conditioned media from cells directly transfected with the MVMp plasmid also produced NS1. Untreated controls and cultures treated with VDS- C did not produce NS1. FIGS. 9A-9B. Deletion of homologous recombination genes maintains the right-end hairpin of H1PV. A) ECORI, SSPI restriction digest of the H1PV and Modified H1PV plasmid, when maintained in (left to right) SURE 2 (lane 2), BB’CDF (lane 3), CDS (lane 4) and BB’CDFS (lane 5) for H1PV and SURE 2 (lane 7), BB’CDF (lane 8), CDS (lane 9) and BB’CDFS (lane 10) for MH1PV. Strains CDS, BB’CDF and BB’CDFS maintained the complete right-end hairpin similar to SURE 2. B) After HEK-293T cells were transfected with H1PV and MH1PV plasmids that were maintained in SURE 2, BB’CDF, CDS and BB’CDFS, the cells produced NS1 protein (83KDa). Uninfected cells did not produce NS1. FIGS.10A-10I. VDS delivers functional virions that kill cancer cells. To measure virus delivery and induced lysis, VDS was applied to cultures of Human Hepatocellular carcinoma (HCC) cells. Human Hepatocellular carcinoma cells (HUH7) were incubated with VDS-B or bacterial controls (BB’CDF). VDS-B contains both the H1PV or MH1PV and PsseJ- lysE plasmids; BB’CDF contains only the PsseJ-lysE plasmid. Similarly, VDS-C was compared to strain CDS, its bacterial control and VDS-BS was compared to its bacterial control BB’CDFS. After two hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using MTT. A) After VDS-B and VDS- C invaded cancer cells in culture, the cells produced viral NS1, similar to direct transfection with the H1PV plasmid. Cell invaded with VDS-BS did not produce NS1. B) Cell viability, measured by MTT assay, decreased for H1PV VDS-B and H1PV transfection compared to BB’CDF (**, P < 0.01). C) H1PV VDS-C did not increase cell death compared to CDS. D) H1PV VDS-BS did not increase cell death compared to BB’CDFS. E) Cell viability decreased for MH1PV VDS-B and MH1PV transfection compared to BB’CDF (***, P < 0.001). F) MH1PV VDS-C did not increase cell death compared to CDS. G) Cell viability decreased for MH1PV VDS-BS and MH1PV transfection compared to BB’CDFS (****, P < 0.0001. H) Cell viability for H1PV and MH1PV VDS-B deceased significantly as compared to BB’CDF at MOIs of 30, 60, 100, and 120. I) Cell viability for H1PV and MH1PV VDS-C deceased significantly as compared to CDS at MOIs of 60, 100, and 120 for H1PV and MOI 120 for MH1PV. FIGS. 11A-11G. To measure virus delivery and induced lysis, VDS was applied to cultures of Murine Hepatocellular carcinoma (HCC) cells. Murine Hepatocellular carcinoma cells (Hepa1-6) were incubated with VDS-B or bacterial controls (BB’CDF). Similarly, VDS- C was compared to strain CDS, its bacterial control. After three hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using MTT. A) After VDS-B and VDS- C invaded cancer cells in culture, the cells produced viral NS1, similar to direct transfection with the H1PV plasmid. B) Cell viability for H1PV and MH1PV VDS-B deceased significantly as compared to BB’CDF at MOI 100 for H1PV and MOIs 50 and 100 for MH1PV. C) Cell viability for H1PV and MH1PV VDS-C deceased significantly as compared to CDS at MOI 50 for H1PV and MOIs 50 and 100 for MH1PV. D) Cell viability, measured by MTT assay, decreased for H1PV VDS-B compared to BB’CDF (*, P < 0.05). E) H1PV VDS-C did not increase cell death compared to CDS. F) Cell viability decreased for MH1PV VDS-B compared to BB’CDF (*, P < 0.05). G) Cell viability decreased for MH1PV VDS-C compared to BB’CDF (*, P < 0.05). FIGS. 12A-12E. To measure virus delivery and induced lysis, VDS was applied to cultures of pancreatic ductal adenocarcinoma cells (KPCY). Murine pancreatic ductal adenocarcinoma cells (KPCY) were incubated with VDS-B or bacterial controls (BB’CDF). Similarly, VDS-C was compared to strain CDS, its bacterial control. After three hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After 36 hours, NS1 expression was measured by immunoblot. After a further 36 hours, cell viability and cell death were measured using MTT. A) After VDS-B invaded cancer cells in culture, the cells produced viral NS1, similar to direct transfection with the H1PV plasmid. No NS1 was produced by the cells invaded by VDS-C. B) Cell viability, measured by MTT assay, decreased for H1PV VDS-B compared to BB’CDF (*, P < 0.05). C) Cell viability decreased for H1PV VDS-C compared to BB’CDF (*, P < 0.05). D) MH1PV VDS-B did not increase cell death compared to BB’CDF. E) MH1PV VDS-C did not increase cell death compared to CDS. FIGS. 13A-13K. To measure virus delivery and induced lysis, VDS was applied to cultures of human embryonic kidney cells (HEK-293T) and human Mammary Adenocarcinoma (MCF 7). KPCY and MCF 7 were incubated with VDS-B or bacterial controls (BB’CDF). Similarly, VDS-C was compared to strain CDS, its bacterial control and VDS-BS was compared to its bacterial control BB’CDFS. After two hours, external bacteria were removed with gentamicin. In parallel, positive control cells were directly transfected with H1PV or MH1PV using lipofectamine. After a further 72 hours, cell viability and cell death were measured using MTT. For HEK-293T cells A) Cell viability, measured by MTT assay, decreased for H1PV VDS-B compared to BB’CDF (****, P < 0.0001). B) H1PV VDS-C did not increase cell death compared to CDS. C) H1PV VDS-BS did not increase cell death compared to BB’CDFS. D) MH1PV VDS-B did not increase cell death compared to BB’CDF. E) Cell viability decreased for MH1PV VDS-C compared to CDS (***, P < 0.001). For MCF 7 cells F) Cell viability decreased for H1PV VDS-B compared to BB’CDF (***, P < 0.001). G) Cell viability decreased for H1PV VDS-C compared to CDS (***, P < 0.001). H) H1PV VDS-BS did not increase cell death compared to BB’CDFS. I) Cell viability decreased for MH1PV VDS-B compared to BB’CDF (*, P < 0.05). J) MH1PV VDS-C did not increase cell death compared to CDS. K) MH1PV VDS-BS did not increase cell death compared to BB’CDFS. FIGS. 14A-14H. Salmonella delivers functional H1PV virions that are cytotoxic to liver cancer cells: A) Similar to the SURE 2 strain of E. coli, the engineered BB’CDF and CDS strains preserved H1PV genome integrity as shown by both DNA bands after restriction digestion. B) DNA from SURE 2 and the engineered Salmonella strains (BB’CDF and CDS) was transfected into HEK-293T cells and produced functional virus as demonstrated by NS1 expression (red arrow). C) Infection of HUH7 liver cancer cells with either CDS or BB’CDF caused DNA delivery and produced functional H1PV virus particles as shown by expression of NS1 by the infected cancer cells. Viral delivery with VDS-B and VDS-C induced cancer cell death in D) HUH7 (**, P<0.01), E) Hep3B (**, P<0.01; *, P<0.05) and F) Sk-Hep-1 (****, P<0.0001) liver cancer cells compared to bacterial controls (BB’CDF or CDS). G) VDS-B and VDS-C delivered functional virus into Hepa 1-6 mouse hepatoma cells as shown by expression of NS1. H) Delivery of H1PV with VDS-B induced significant cellular cytotoxicity compared to the bacterial control (*, P<0.05). FIGS. 15A-15F. VDS-B delivery induces functional H1PV production within liver tumors in vivo. A) Similar to BB’CDF, mice injected with VDS-B rapidly recovered three days after bacterial injection. B) There was no difference in serum proinflammatory cytokines seven days after VDS-B injection as compared to saline injected mice. C) To assess virus delivery in vivo, C57BL/6 mice with hepa1-6 tumors were intratumorally injected with VDS-B. Three days after injection, tumors were harvested and D) evaluated for NS1 expression on western blot. NS1 expression indicated that VDS-B delivered and launched an H1PV infection in tumors. E) To test for fully functional virion production in vivo, C57BL/6 mice with hepa1-6 tumors were intratumorally injected with VDS-B or saline as a control. Five days after intratumoral injection, tumors were harvested, homogenized in PBS, and centrifuged. The supernatant, which contains infective virus, was filtered and gentamycin was added to eliminate any bacteria. The virus containing supernatant, or saline (control) was added to HEK- 293T cells. 48 hours after incubation, HEK293T cells were harvested and evaluated for NS1 expression using a western blot. F) NS1 was expressed in HEK293T cells infected with tumor homogenates, indicating that VDS-B delivery produced functional and re-infective viral particles. FIGS.16A-16E. Figure 3: VDS-B inhibits the growth of liver cancer in vivo. A) Mice with subcutaneous hepa1-6 tumors were injected intratumorally with VDS-B or the BB’CDF control strain a total of six times and tumors were measured every 3 days. B) Mice injected with VDS-B exhibited minimal tumor growth as compared to mice injected with the BB’CDF control strain. C) Mice injected with the BB’CDF control strain exhibited steady tumor growth while mice injected with VDS-B did not exhibit any tumor growth 36 days after the first bacterial injection. D) VDS-B injection reduced tumor volumes by 84% compared to treatment with the BB’CDF delivery strain (**, P<0.01). E) 100% of VDS-B mice continued to survive after injection while 0% of control mice survived 33 days after injection (**, P<0.01). FIGS. 17A-17F. Intravenous injection is similarly efficacious compared to intratumoral injection of VDS-B. A) C57BL/6 mice with hepa1-6 tumors were intravenously injected with VDS-B six times every six days. B) After intravenous injection, VDS-B injected mice exhibited minimal tumor growth compared to control injected mice. C) Intravenous injection resulted in a 79% reduction in tumor growth and D) a 100% survival compared to control treated mice (*, P<0.05). E) Intravenous administration resulted in equivalent tumor volume reduction as compared to intratumoral injection of VDS-B (**, P<0.01) which, F) resulted in similar overall survival compared to the bacterially injected control (*, P<0.05; **, P<0.01). FIGS. 18A-18J. VDS-B generates an innate immunostimulatory response against tumors. A) Mice with subcutaneous hepa1-6 tumors were injected three times with VDS-B at 6-day intervals and sacrificed five days after the last injection. B) The same tumors were measured for the duration of the study. VDS-B treated tumors exhibited a 72% reduction in tumor volume as compared to control tumors (*, P<0.05). C) VDS-B reduced the mass of viable tumor by 35% compared to bacterial control treated tumors (*, P<0.05). D) The total fraction of cancer cells reduced by 50% compared to control tumors (*, P<0.05). E) The number of natural killer cells increased by 66% compared to the bacterial control treated tumors (*, P<0.01). F) The fraction of intratumoral macrophages increased 2.2-fold as compared to controls (*, P<0.05). G) The fraction of immune cells in tumors significantly decreased after VDS-B treatment (*, P<0.05). H) The reduction in (G) was likely because of a significant reduction in neutrophil infiltration after treatment with VDS-B (**, P<0.01). I) VDS-B treatment increased the percentage of M1 macrophages while decreasing the percentage of M2 macrophages as compared to the BB’CDF control. J) The ratio of M1/M2 macrophages in tumors increased 2.5-fold after treatment with VDS-B as compared to BB’CDF (*, P<0.05). FIGS.19A-19K. VDS-B generates an anti-tumor adaptive immune response. A) Mice with subcutaneous hepa1-6 tumors were injected three times with VDS-B at 6-day intervals and sacrificed five days after the last injection. B) VDS-B induced a 4-fold increase in T cell infiltration within tumors (*, P<0.05). C) A significant portion of the infiltrating lymphocytes in VDS-B treated tumors were CD8 T cells. D) The ratio of CD8+ T cells to regulatory T cells increased 12-fold after treatment with VDS-B as compared BB’CDF (*, P<0.05). E) CD4+ T cells (likely Th1) increased 3-fold after treatment with VDS-B as compared to BB’CDF (*, P<0.05). F) More activated CD8+ T cells infiltrated tumors after administration of VDS-B as compared with BB’CDF. G) There were 6-fold more activated CD8+ T cells present in tumors after VDS-B administration as compared to BB’CDF (*, P<0.05). H) The fraction of granulated CD8+ T cells in tumors increased 4-fold after administration of VDS-B as compared to BB’CDF (*, P<0.05). I) Splenocytes were harvested from Mice with hepa1-6 tumors that were treated with VDS-B. The splenocytes were adoptively transferred into C57BL/6 mice. The mice were challenged with hepa1-6 tumor cells two weeks after splenocyte transfer and monitored for tumor growth for two 66 days. J) 75% of naïve mice grafted hepa1-6 tumor cells as compared to 0% of mice with adoptively transferred splenocytes from VDS-B treated, tumor bearing mice. K) After tumor cell injection, mice with adoptively transferred splenocytes from VDS-B treated mice exhibited no tumor growth, in contrast to the control mice. FIGS. 20A-20I. Engineered Salmonella deliver functional H1PV in liver tumors in vivo. A) C57BL/6 mice with subcutaneous Hepa1-6 tumors were intravenously injected with BB’CDF. 3 days after injection, organs were harvested, and bacteria were enumerated. B) BB’CDF colonized tumors at least 50 million-fold more than any other organ (****, P<0.0001). C) BB’CDF colonized tumors to a similar level as the parental delivery strain. Although not statistically significant, BB’CDF also colonized health organ tissue at least 100- fold less than the parental delivery strain of Salmonella. D) Similar to (A) healthy mice were injected with either parental or BB’CDF and body weights were measured for 7 days. E) Mice injected with BB’CDF fully recovered from bacterial challenge 3 days post injection unlike mice injected with the parental strain, which took 5 days to fully recover. F) Seven days after bacterial injection, serum cytokine profiling was evaluated and BB’CDF demonstrated lower levels of all inflammatory cytokines compared to the parental strain, although not statistically significant. Two of the most critical cytokines associated with septic shock, (G) IL-6 (*, P<0.05) and (H) TNFα (**, P<0.01), exhibited significantly lower serum levels seven days after BB’CDF injection as compared to parental Salmonella injection. I) Mice injected with parental Salmonella lost more body weight and took longer to recover from administration as compared to mice injected with BB’CDF. DESCRIPTION The majority of proteins are intracellular. Specifically targeting intracellular pathways in cancer cells using macromolecular therapies increases the potential treatment options for any patient. However, macromolecular therapies that target intracellular pathways face significant barriers associated with tumor targeting, distribution, internalization and endosomal release. Engineered, non-pathogenic Salmonella selectively colonize tumors one thousand-fold more than any other organ, invade and deliver therapies cytosolically into cancer cells making the bacteria ideal delivery vehicles for cancer therapy. Many of the current problems associated with cancer treatment, e.g., metastatic disease and refractory tumors, could be overcome with microbial therapies. Oncolytic viruses (OVs) have the potential to treat many tumors by directly lysing cancer cells and stimulating immune responses that eliminate cancer cells regardless of their location in the body (1-6). Despite this potential, oncolytic viruses have not been effective at treating internal solid tumors (5, 7-10). When injected systemically, OVs are cleared from the blood and do not effectively reach tumors (1, 5, 7-20). This problem could be overcome with a carrier with tumor specific tropism. It has been shown that bacterial therapies predominantly accumulate in tumors after intravenous injections (21-30). Developing a bacterial system to deliver viruses into cancer cells would couple the benefits of these two microbial therapies and focus treatment specifically to cancers. Abbreviations OV - oncolytic virus VDS - Virus-delivering Salmonella MVM - minute virus of mice MVMp - prototype strain of MVM H1PV: H1 parvovirus MH1PV: Modified H1 parvovirus ID - intracellular delivering HRDS - homologous-recombination-deficient Salmonella CMV - Cytomegalovirus GFP - Green fluorescent protein SURE 2 - stop unwanted rearrangement events’ 2 Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, several embodiments with regards to methods and materials are described herein. As used herein, each of the following terms has the meaning associated with it in this section. For the purposes of clarity and a concise description, features can be described herein as part of the same or separate embodiments; however, it will be appreciated that the scope of the invention may include embodiments having combinations of all or some of the features described. References in the specification to "one embodiment", "an embodiment", etc., indicate that the embodiment described may include a particular aspect, feature, structure, moiety, or characteristic, but not every embodiment necessarily includes that aspect, feature, structure, moiety, or characteristic. Moreover, such phrases may, but do not necessarily, refer to the same embodiment referred to in other portions of the specification. Further, when a particular aspect, feature, structure, moiety, or characteristic is described in connection with an embodiment, it is within the knowledge of one skilled in the art to affect or connect such aspect, feature, structure, moiety, or characteristic with other embodiments, whether or not explicitly described. As used herein, the indefinite articles “a”, “an” and “the” should be understood to include plural reference unless the context clearly indicates otherwise. The phrase “and/or,” as used herein, should be understood to mean “either or both” of the elements so conjoined, e.g., elements that are conjunctively present in some cases and disjunctively present in other cases. As used herein, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating a listing of items, “and/or” or “or” shall be interpreted as being inclusive, e.g., the inclusion of at least one, but also including more than one, of a number of items, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of” or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” As used herein, the terms “including,” “includes,” “having,” “has,” “with,” or variants thereof, are intended to be inclusive similar to the term “comprising.” As used herein, the term “about” means plus or minus 10% of the indicated value. For example, about 100 means from 90 to 110. Numerical ranges recited herein by endpoints include all numbers and fractions subsumed within that range (e.g., 1 to 5 includes 1, 1.5, 2, 2.75, 3, 3.90, 4, and 5). It is also to be understood that all numbers and fractions thereof are presumed to be modified by the term “about.” The terms "individual," "subject," and "patient," are used interchangeably herein and refer to any subject for whom diagnosis, treatment, or therapy is desired, including a mammal. Mammals include, but are not limited to, humans, farm animals, sport animals and pets. A “subject” is a vertebrate, such as a mammal, including a human. Mammals include, but are not limited to, humans, farm animals, sport animals and companion animals. Included in the term “animal” is dog, cat, fish, gerbil, guinea pig, hamster, horse, rabbit, swine, mouse, monkey (e.g., ape, gorilla, chimpanzee, orangutan) rat, sheep, goat, cow and bird. The terms "treatment", "treating" and the like are used herein to generally mean obtaining a desired pharmacologic and/or physiologic effect, such as arresting or inhibiting, or attempting to arrest or inhibit, the development or progression of a disorder and/or causing, or attempting to cause, the reduction, suppression, regression, or remission of a disorder and/or a symptom thereof. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. As would be understood by those skilled in the art, various clinical and scientific methodologies and assays may be used to assess the development or progression of a disorder, and similarly, various clinical and scientific methodologies and assays may be used to assess the reduction, regression, or remission of a disorder or its symptoms. Additionally, treatment can be applied to a subject or to a cell culture (in vivo or in vitro). The terms "inhibit", "inhibiting", and "inhibition" refer to the slowing, halting, or reversing the growth or progression of a disease, infection, condition, group of cells, protein or its expression. The inhibition can be greater than about 20%, 40%, 60%, 80%, 90%, 95%, or 99%, for example, compared to the growth or progression that occurs in the absence of the treatment or contacting. “Expression” refers to the production of RNA from DNA and/or the production of protein directed by genetic material (e.g., RNA (mRNA)). Inducible expression, as opposed to constitutive expression (expressed all the time), is expression which only occurs under certain conditions, such as in the presence of specific molecule (e.g., arabinose) or an environmental que. The term ''exogenous" as used herein with reference to a nucleic acid (or a protein) and a host refers to a nucleic acid that does not occur in (and cannot be obtained from) a cell of that particular type as it is found in nature or a protein encoded by such a nucleic acid. Thus, a non- naturally occurring nucleic acid is considered to be exogenous to a host once in the host. It is important to note that non-naturally occurring nucleic acids can contain nucleic acid subsequences or fragments of nucleic acid sequences that are found in nature provided the nucleic acid as a whole does not exist in nature. For example, a nucleic acid molecule containing a genomic DNA sequence within an expression vector is non-naturally occurring nucleic acid, and thus is exogenous to a host cell once introduced into the host, since that nucleic acid molecule as a whole (genomic DNA plus vector DNA) does not exist in nature. Thus, any vector, autonomously replicating plasmid, or virus (e.g., retrovirus, adenovirus, or herpes virus) that as a whole does not exist in nature is considered to be non-naturally occurring nucleic acid. It follows that genomic DNA fragments produced by PCR or restriction endonuclease treatment as well as cDNAs are considered to be non-naturally occurring nucleic acid since they exist as separate molecules not found in nature. An exogenous sequence may therefore be integrated into the genome of the host. It also follows that any nucleic acid containing a promoter sequence and polypeptide-encoding sequence (e.g., cDNA or genomic DNA) in an arrangement not found in nature is non-naturally occurring nucleic acid. A nucleic acid that is naturally occurring can be exogenous to a particular host microorganism. For example, an entire chromosome isolated from a cell of yeast x is an exogenous nucleic acid with respect to a cell of yeast y once that chromosome is introduced into a cell of yeast y. In contrast, the term "endogenous" as used herein with reference to a nucleic acid (e.g., a gene) (or a protein) and a host refers to a nucleic acid (or protein) that does occur in (and can be obtained from) that particular host as it is found in nature. Moreover, a cell "endogenously expressing" a nucleic acid (or protein) expresses that nucleic acid (or protein) as does a host of the same particular type as it is found in nature. Moreover, a host "endogenously producing" or that "endogenously produces" a nucleic acid, protein, or other compound produces that nucleic acid, protein, or compound as does a host of the same particular type as it is found in nature. Engineered Salmonella could be any strain of Salmonella designed to lyse and deliver protein intracellularly. In some embodiments, the engineered Salmonella is non-pathogenic. The term "contacting" refers to the act of touching, making contact, or of bringing to immediate or close proximity, including at the cellular or molecular level, for example, to bring about a physiological reaction, a chemical reaction, or a physical change, e.g., in a solution, in a reaction mixture, in vitro, or in vivo. An "effective amount" is an amount sufficient to effect beneficial or desired result, such as a preclinical or clinical result. An effective amount can be administered in one or more administrations. The term “effective amount,” as applied to the compound(s), biologics and pharmaceutical compositions described herein, means the quantity necessary to render the desired therapeutic result. For example, an effective amount is a level effective to treat, cure, or alleviate the symptoms of a disorder and/or disease for which the therapeutic compound, biologic or composition is being administered. Amounts effective for the particular therapeutic goal sought will depend upon a variety of factors including the disorder being treated and its severity and/or stage of development/progression; the bioavailability, and activity of the specific compound, biologic or pharmaceutical composition used; the route or method of administration and introduction site on the subject; the rate of clearance of the specific compound or biologic and other pharmacokinetic properties; the duration of treatment; inoculation regimen; drugs used in combination or coincident with the specific compound, biologic or composition; the age, body weight, sex, diet, physiology and general health of the subject being treated; and like factors well known to one of skill in the relevant scientific art. Some variation in dosage can occur depending upon the condition of the subject being treated, and the physician or other individual administering treatment will, in any event, determine the appropriate dose for an individual patient. As used herein, “disorder” refers to a disorder, disease or condition, or other departure from healthy or normal biological activity, and the terms can be used interchangeably. The terms would refer to any condition that impairs normal function. The condition may be caused by sporadic or heritable genetic abnormalities. The condition may also be caused by non- genetic abnormalities. The condition may also be caused by injuries to a subject from environmental factors, such as, but not limited to, cutting, crushing, burning, piercing, stretching, shearing, injecting, or otherwise modifying a subject's cell(s), tissue(s), organ(s), system(s), or the like. The terms “cell,” “cell line,” and “cell culture” as used herein may be used interchangeably. All of these terms also include their progeny, which are any and all subsequent generations. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations. A “coding region” of a gene consists of the nucleotide residues of the coding strand of the gene and the nucleotides of the non-coding strand of the gene which are homologous with or complementary to, respectively, the coding region of an mRNA molecule which is produced by transcription of the gene. “Complementary” as used herein refers to the broad concept of subunit sequence complementarity between two nucleic acids, e.g., two DNA molecules. When a nucleotide position in both of the molecules is occupied by nucleotides normally capable of base pairing with each other, then the nucleic acids are considered to be complementary to each other at this position. Thus, two nucleic acids are complementary to each other when a substantial number (at least 50%) of corresponding positions in each of the molecules are occupied by nucleotides which normally base pair with each other (e.g., A:T and G:C nucleotide pairs). Thus, it is known that an adenine residue of a first nucleic acid region is capable of forming specific hydrogen bonds (“base pairing”) with a residue of a second nucleic acid region which is antiparallel to the first region if the residue is thymine or uracil. Similarly, it is known that a cytosine residue of a first nucleic acid strand is capable of base pairing with a residue of a second nucleic acid strand which is antiparallel to the first strand if the residue is guanine. A first region of a nucleic acid is complementary to a second region of the same or a different nucleic acid if, when the two regions are arranged in an antiparallel fashion, at least one nucleotide residue of the first region is capable of base pairing with a residue of the second region. Preferably, the first region comprises a first portion and the second region comprises a second portion, whereby, when the first and second portions are arranged in an antiparallel fashion, at least about 50%, and preferably at least about 75%, at least about 90%, or at least about 95% of the nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. More preferably, all nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. “Encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA. As used herein, an “essentially pure” preparation of a particular protein or peptide is a preparation wherein at least about 95%, and preferably at least about 99%, by weight, of the protein or peptide in the preparation is the particular protein or peptide. A “fragment” or “segment” is a portion of an amino acid sequence, comprising at least one amino acid, or a portion of a nucleic acid sequence comprising at least one nucleotide. The terms “fragment” and “segment” are used interchangeably herein. As used herein, a “functional” biological molecule is a biological molecule in a form in which it exhibits a property by which it is characterized. A functional enzyme, for example, is one which exhibits the characteristic catalytic activity by which the enzyme is characterized. “Homologous” as used herein, refers to the subunit sequence similarity between two polymeric molecules, e.g., between two nucleic acid molecules, e.g., two DNA molecules or two RNA molecules, or between two polypeptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position. The homology between two sequences is a direct function of the number of matching or homologous positions, e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two compound sequences are homologous then the two sequences are 50% homologous, if 90% of the positions, e.g., 9 of 10, are matched or homologous, the two sequences share 90% homology. By way of example, the DNA sequences 3’ATTGCC5’ and 3’TATGGC share 50% homology. As used herein, “homology” is used synonymously with “identity.” The determination of percent identity between two nucleotide or amino acid sequences can be accomplished using a mathematical algorithm. For example, a mathematical algorithm useful for comparing two sequences is the algorithm of Karlin and Altschul (1990, Proc. Natl. Acad. Sci. USA 87:2264-2268), modified as in Karlin and Altschul (1993, Proc. Natl. Acad. Sci. USA 90:5873-5877). This algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990, J. Mol. Biol. 215:403-410), and can be accessed, for example at the National Center for Biotechnology Information (NCBI) world wide web site having the universal resource locator using the BLAST tool at the NCBI website. BLAST nucleotide searches can be performed with the NBLAST program (designated “blastn” at the NCBI web site), using the following parameters: gap penalty = 5; gap extension penalty = 2; mismatch penalty = 3; match reward = 1; expectation value 10.0; and word size = 11 to obtain nucleotide sequences homologous to a nucleic acid described herein. BLAST protein searches can be performed with the XBLAST program (designated “blastn” at the NCBI web site) or the NCBI “blastp” program, using the following parameters: expectation value 10.0, BLOSUM62 scoring matrix to obtain amino acid sequences homologous to a protein molecule described herein. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. (1997, Nucleic Acids Res. 25:3389-3402). Alternatively, PSI-Blast or PHI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.) and relationships between molecules which share a common pattern. When utilizing BLAST, Gapped BLAST, PSI-Blast, and PHI-Blast programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically exact matches are counted. As used herein, the term “hybridization” is used in reference to the pairing of complementary nucleic acids. Hybridization and the strength of hybridization (i.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementarity between the nucleic acids, stringency of the conditions involved, the length of the formed hybrid, and the G:C ratio within the nucleic acids. As used herein, an “instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the peptide of the invention in the kit for effecting alleviation of the various diseases or disorders recited herein. Optionally, or alternately, the instructional material may describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a mammal. The instructional material of the kit of the invention may, for example, be affixed to a container which contains the identified compound invention or be shipped together with a container which contains the identified compound. Alternatively, the instructional material may be shipped separately from the container with the intention that the instructional material and the compound be used cooperatively by the recipient. The term “nucleic acid” typically refers to large polynucleotides. By “nucleic acid” is meant any nucleic acid, whether composed of deoxyribonucleosides or ribonucleosides, and whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sulfone linkages, and combinations of such linkages. The term nucleic acid also specifically includes nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine and uracil). As used herein, the term “nucleic acid” encompasses RNA as well as single and double stranded DNA and cDNA. Furthermore, the terms, “nucleic acid,” “DNA,” “RNA” and similar terms also include nucleic acid analogs, i.e., analogs having other than a phosphodiester backbone. For example, the so called “peptide nucleic acids,” which are known in the art and have peptide bonds instead of phosphodiester bonds in the backbone, are considered within the scope of the present invention. By “nucleic acid” is meant any nucleic acid, whether composed of deoxyribonucleosides or ribonucleosides, and whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sulfone linkages, and combinations of such linkages. The term nucleic acid also specifically includes nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine, and uracil). Conventional notation is used herein to describe polynucleotide sequences: the left-hand end of a single-stranded polynucleotide sequence is the 5’-end; the left-hand direction of a double-stranded polynucleotide sequence is referred to as the 5’-direction. The direction of 5’ to 3’ addition of nucleotides to nascent RNA transcripts is referred to as the transcription direction. The DNA strand having the same sequence as an mRNA is referred to as the “coding strand”; sequences on the DNA strand which are located 5’ to a reference point on the DNA are referred to as “upstream sequences”; sequences on the DNA strand which are 3’ to a reference point on the DNA are referred to as “downstream sequences.” The term “nucleic acid construct,” as used herein, encompasses DNA and RNA sequences encoding the particular gene or gene fragment desired, whether obtained by genomic or synthetic methods. Unless otherwise specified, a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns. The term “oligonucleotide” typically refers to short polynucleotides, generally, no greater than about 50 nucleotides. It will be understood that when a nucleotide sequence is represented by a DNA sequence (i.e., A, T, G, C), this also includes an RNA sequence (i.e., A, U, G, C) in which “U” replaces “T.” “Substantially homologous nucleic acid sequence” means a nucleic acid sequence corresponding to a reference nucleic acid sequence wherein the corresponding sequence encodes a peptide having substantially the same structure and function as the peptide encoded by the reference nucleic acid sequence; e.g., where only changes in amino acids not significantly affecting the peptide function occur. Preferably, the substantially identical nucleic acid sequence encodes the peptide encoded by the reference nucleic acid sequence. The percentage of identity between the substantially similar nucleic acid sequence and the reference nucleic acid sequence is at least about 50%, 65%, 75%, 85%, 95%, 99% or more. Substantial identity of nucleic acid sequences can be determined by comparing the sequence identity of two sequences, for example by physical/chemical methods (i.e., hybridization) or by sequence alignment via computer algorithm. Suitable nucleic acid hybridization conditions to determine if a nucleotide sequence is substantially similar to a reference nucleotide sequence are: 7% sodium dodecyl sulfate SDS, 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 2X standard saline citrate (SSC), 0.1% SDS at 50°C; preferably in 7% (SDS), 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 1X SSC, 0.1% SDS at 50°C; preferably 7% SDS, 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 0.5X SSC, 0.1% SDS at 50°C; and more preferably in 7% SDS, 0.5 M NaPO4, 1 mM EDTA at 50°C with washing in 0.1X SSC, 0.1% SDS at 65°C. Suitable computer algorithms to determine substantial similarity between two nucleic acid sequences include, GCS program package (Devereux et al., 1984 Nucl. Acids Res. 12:387), and the BLASTN or FASTA programs (Altschul et al., 1990 Proc. Natl. Acad. Sci. USA.1990 87:14:5509-13; Altschul et al., J. Mol. Biol. 1990215:3:403-10; Altschul et al., 1997 Nucleic Acids Res.25:3389-3402). The default settings provided with these programs are suitable for determining substantial similarity of nucleic acid sequences for purposes of the present invention. By describing two polynucleotides as “operably linked” is meant that a single-stranded or double-stranded nucleic acid moiety comprises the two polynucleotides arranged within the nucleic acid moiety in such a manner that at least one of the two polynucleotides is able to exert a physiological effect by which it is characterized upon the other. By way of example, a promoter operably linked to the coding region of a gene is able to promote transcription of the coding region. As used herein, the term “pharmaceutically acceptable carrier” means a chemical composition with which an appropriate compound or derivative can be combined and which, following the combination, can be used to administer the appropriate compound to a subject. “Pharmaceutically acceptable” means physiologically tolerable, for either human or veterinary application. As used herein, “pharmaceutical compositions” include formulations for human and veterinary use. As used herein, the term “purified” and like terms relate to an enrichment of a molecule or compound relative to other components normally associated with the molecule or compound in a native environment. The term “purified” does not necessarily indicate that complete purity of the particular molecule has been achieved during the process. A “highly purified” compound as used herein refers to a compound that is greater than 90% pure. In particular, purified sperm cell DNA refers to DNA that does not produce significant detectable levels of non-sperm cell DNA upon PCR amplification of the purified sperm cell DNA and subsequent analysis of that amplified DNA. A “significant detectable level” is an amount of contaminate that would be visible in the presented data and would need to be addressed/explained during analysis of the forensic evidence. “Recombinant polynucleotide” refers to a polynucleotide having sequences that are not naturally joined together. An amplified or assembled recombinant polynucleotide may be included in a suitable vector, and the vector can be used to transform a suitable host cell. A recombinant polynucleotide may serve a non-coding function (e.g., promoter, origin of replication, ribosome-binding site, etc.) as well. A host cell that comprises a recombinant polynucleotide is referred to as a “recombinant host cell.” A gene which is expressed in a recombinant host cell wherein the gene comprises a recombinant polynucleotide, produces a “recombinant polypeptide.” A “recombinant polypeptide” is one which is produced upon expression of a recombinant polynucleotide. A “recombinant cell” is a cell that comprises a transgene. Such a cell may be a eukaryotic or a prokaryotic cell. Also, the transgenic cell encompasses, but is not limited to, an embryonic stem cell comprising the transgene, a cell obtained from a chimeric mammal derived from a transgenic embryonic stem cell where the cell comprises the transgene, a cell obtained from a transgenic mammal, or fetal or placental tissue thereof, and a prokaryotic cell comprising the transgene. The term “regulate” refers to either stimulating or inhibiting a function or activity of interest. By the term “specifically binds to,” as used herein, is meant when a compound or ligand functions in a binding reaction or assay conditions which is determinative of the presence of the compound in a sample of heterogeneous compounds, or it means that one molecule, such as a binding moiety, e.g., an oligonucleotide or antibody, binds preferentially to another molecule, such as a target molecule, e.g., a nucleic acid or a protein, in the presence of other molecules in a sample. The terms "specific binding" or "specifically binding" when used in reference to the interaction of a peptide (ligand) and a receptor (molecule) also refers to an interaction that is dependent upon the presence of a particular structure (i.e., an amino sequence of a ligand or a ligand binding domain within a protein); in other words the peptide comprises a structure allowing recognition and binding to a specific protein structure within a binding partner rather than to molecules in general. For example, if a ligand is specific for binding pocket "A," in a reaction containing labeled peptide ligand "A" (such as an isolated phage displayed peptide or isolated synthetic peptide) and unlabeled "A" in the presence of a protein comprising a binding pocket A the unlabeled peptide ligand will reduce the amount of labeled peptide ligand bound to the binding partner, in other words a competitive binding assay. The term “standard,” as used herein, refers to something used for comparison. For example, it can be a known standard agent or compound which is administered and used for comparing results when administering a test compound, or it can be a standard parameter or function which is measured to obtain a control value when measuring an effect of an agent or compound on a parameter or function. Standard can also refer to an “internal standard”, such as an agent or compound which is added at known amounts to a sample and is useful in determining such things as purification or recovery rates when a sample is processed or subjected to purification or extraction procedures before a marker of interest is measured. Internal standards are often a purified marker of interest which has been labeled, such as with a radioactive isotope, allowing it to be distinguished from an endogenous marker. Methods involving conventional molecular biology techniques are described herein. Such techniques are generally known in the art and are described in detail in methodology treatises, such as Molecular Cloning: A Laboratory Manual, 2nd ed., vol.1-3, ed. Sambrook et al., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989; and Current Protocols in Molecular Biology, ed. Ausubel et al., Greene Publishing and Wiley-Interscience, New York, 1992 (with periodic updates). Methods for chemical synthesis of nucleic acids are discussed, for example, in Beaucage and Carruthers, Tetra. Letts. 22: 1859-1862, 1981, and Matteucci et al., J. Am. Chem. Soc.103:3185, 1981. As used herein, the terms “including”, “includes”, “having”, “has”, “with”, or variants thereof, are intended to be inclusive similar to the term “comprising.” The terms “comprises,” “comprising,” and the like can have the meaning ascribed to them in U.S. Patent Law and can mean “includes,” “including” and the like. As used herein, “including” or “includes” or the like means including, without limitation. I. Bacteria Bacteria useful in the invention include, but are not limited to, Clostridium, Bifidus, Escherichia coli or Salmonella, T3SS-dependent bacteria, such as shigella, salmonella and Yersinia Pestis. Further, E. coli can be used if the T3SS system is place in E. Coli. Salmonella Examples of Salmonella strains which can be employed in the present invention include Salmonella typhi (ATCC No. 7251) and S. typhimurium (ATCC No. 13311). Attenuated Salmonella strains include S. typhi-aroC-aroD (Hone et al. Vacc.9:810 (1991) S. typhimurium- aroA mutant (Mastroeni et al. Micro. Pathol. 13:477 (1992)) and Salmonella typhimurium 7207. Additional attenuated Salmonella strains that can be used in the invention include one or more other attenuating mutations such as (i) auxotrophic mutations, such as aro (Hoiseth et al. Nature, 291:238-239 (1981)), gua (McFarland et al Microbiol. Path., 3:129-141 (1987)), nad (Park et al. J. Bact, 170:3725-3730 (1988), thy (Nnalue et al. Infect. Immun., 55:955-962 (1987)), and asd (Curtiss, supra) mutations; (ii) mutations that inactivate global regulatory functions, such as cya (Curtiss et al. Infect. Immun., 55:3035-3043 (1987)), crp (Curtiss et al (1987), supra), phoP/phoQ (Groisman et al. Proc. Natl. Acad. Sci., USA, 86:7077-7081 (1989); and Miller et al. Proc. Natl. Acad. Sci., USA, 86:5054-5058 (1989)), phop.sup.c (Miller et al. J. Bact, 172:2485-2490 (1990)) or ompR (Dorman et al. Infect. Immun., 57:2136-2140 (1989)) mutations; (iii) mutations that modify the stress response, such as recA (Buchmeier et al. MoI. Micro., 7:933-936 (1993)), htrA (Johnson et al. MoI. Micro., 5:401-407 (1991)), htpR (Neidhardt et al. Biochem. Biophys. Res. Com., 100:894-900 (1981)), hsp (Neidhardt et al. Ann. Rev. Genet, 18:295-329 (1984)) and groEL (Buchmeier et al. Sci., 248:730-732 (1990)) mutations; mutations in specific virulence factors, such as IsyA (Libby et al. Proc. Natl. Acad. Sci., USA, 91:489-493 (1994)), pag or prg (Miller et al (1990), supra; and Miller et al (1989), supra), iscA or virG (d'Hauteville et al. MoI. Micro., 6:833-841 (1992)), plcA (Mengaud et al. Mol. Microbiol., 5:367-72 (1991); Camilli et al. J. Exp. Med, 173:751-754 (1991)), and act (Brundage et al. Proc. Natl. Acad. Sci., USA, 90:11890-11894 (1993)) mutations; (v) mutations that affect DNA topology, such as top A (Galan et al. Infect. Immun., 58: 1879-1885 (1990)); (vi) mutations that disrupt or modify the cell cycle, such as min (de Boer et al. Cell, 56:641- 649 (1989)); (vii) introduction of a gene encoding a suicide system, such as sacB (Recorbet et al. App. Environ. Micro., 59:1361-1366 (1993); Quandt et al. Gene, 127:15-21 (1993)), nuc (Ahrenholtz et al. App. Environ. Micro., 60:3746-3751 (1994)), hok, gef, kil, or phlA (Molin et al. Ann. Rev. Microbiol., 47:139-166 (1993)); (viii) mutations that alter the biogenesis of lipopolysaccharide and/or lipid A, such as rFb (Raetz in Esherishia coli and Salmonella typhimurium, Neidhardt et al, Ed., ASM Press, Washington D.C. pp 1035-1063 (1996)), galE (Hone et al. J. Infect. Dis., 156:164-167 (1987)) and htrB (Raetz, supra), msbB (Reatz, supra; and US Patent No. 7,514,089); and (ix) introduction of a bacteriophage lysis system, such as lysogens encoded by P22 (Rennell et al. Virol, 143:280-289 (1985)), lamda murein transglycosylase (Bienkowska-Szewczyk et al. Mol. Gen. Genet., 184:111-114 (1981)) or S- gene (Reader et al. Virol, 43:623-628 (1971)). The attenuating mutations can be either constitutively expressed or under the control of inducible promoters, such as the temperature sensitive heat shock family of promoters (Neidhardt et al. supra), or the anaerobically induced nirB promoter (Harbome et al. Mol. Micro., 6:2805-2813 (1992)) or repressible promoters, such as uapA (Gorfinkiel et al. J. Biol. Chem., 268:23376-23381 (1993)) or gcv (Stauffer et al. J. Bact, 176:6159-6164 (1994)). In one embodiment, the bacterial delivery system is safe and based on a non-toxic, attenuated Salmonella strain that has a partial deletion of the msbB gene. This deletion diminishes the TNF immune response to bacterial lipopolysaccharides and prevents septic shock. In another embodiment, it also has a partial deletion of the purI gene. This deletion makes the bacteria dependent on external sources of purines and speeds clearance from non-cancerous tissues (13). In mice, the virulence (LD50) of the therapeutic strain is 10,000-fold less than wild-type Salmonella (72, 73). In pre-clinical trials, attenuated Salmonella has been administered systemically into mice and dogs without toxic side effects (17, 27). Two FDA-approved phase I clinical trials have been performed and showed that this therapeutic strain can be safely administered to patients (20). In one embodiment, the strain of bacteria is VNP20009, a derivative strain of Salmonella typhimurium. Deletion of two of its genes - msbB and purI -resulted in its complete attenuation (by preventing toxic shock in animal hosts) and dependence on external sources of purine for survival. This dependence renders the organism incapable of replicating in normal tissue such as the liver or spleen, but still capable of growing in tumors where purine is available. Further, insertion of a failsafe circuit into the bacterial vector prevents unwanted infection and defines the end of therapy without the need for antibiotics to remove the bacteria (e.g., salmonella). II. Vectors/Plasmids In the present compositions and/or methods, DNA, RNA (e.g., a nucleic acid-based gene interfering agent) or protein may be produced by recombinant methods. The nucleic acid is inserted into a replicable vector for expression. Many such vectors are available. The vector components generally include, but are not limited to, one or more of the following: an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence and coding sequence. In some embodiments, for example in the utilization of bacterial delivery agents such as Salmonella, the gene and/or promoter (a sequence of interest) may be integrated into the host cell chromosome or may be presented on, for example, a plasmid/vector. Expression vectors usually contain a selection gene, also termed a selectable marker. This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will not survive in the culture medium. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media. Expression vectors can contain a promoter that is recognized by the host organism and is operably linked to the nucleic acid sequence, such as a nucleic acid sequence coding for an open reading frame. Promoters are untranslated sequences located upstream (5') to the start codon of a structural gene (generally within about 100 to 1000 bp) that control the transcription of particular nucleic acid sequence to which they are operably linked. In bacterial cells, the region controlling overall regulation can be referred to as the operator. Promoters typically fall into two classes, inducible and constitutive. Inducible promoters are promoters that initiate increased levels of transcription from DNA under their control in response to some change in culture conditions, e.g., the presence or absence of a nutrient or a change in temperature. A large number of promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the β-lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, hybrid promoters such as the tac promoter, and starvation promoters (Matin, A. (1994) Recombinant DNA Technology II, Annals of New York Academy of Sciences, 722:277-291). However, other known bacterial promoters are also suitable. Such nucleotide sequences have been published, thereby enabling a skilled worker to operably ligate them to a DNA coding sequence. Promoters for use in bacterial systems also can contain a Shine-Dalgarno (S.D.) sequence operably linked to the coding sequence. Construction of suitable vectors containing one or more of the above-listed components employs standard ligation techniques. Isolated plasmids or DNA fragments are cleaved, tailored, and re-ligated in the form desired to generate the plasmids required. In some embodiments of the invention, the expression vector is a plasmid or bacteriophage vector suitable for use in Salmonella, and the DNA, RNA and/or protein is provided to a subject through expression by an engineered Salmonella (in one aspect attenuated) administered to the patient. The term "plasmid" as used herein refers to any nucleic acid encoding an expressible gene and includes linear or circular nucleic acids and double or single stranded nucleic acids. The nucleic acid can be DNA or RNA and may comprise modified nucleotides or ribonucleotides and may be chemically modified by such means as methylation or the inclusion of protecting groups or cap- or tail structures. One embodiment provides a Salmonella strain comprising a lysis gene or cassette operably linked to an intracellularly induced Salmonella promoter. One embodiment provides an attenuated Salmonella strain comprising a lysis gene or cassette operably linked to an intracellularly induced Salmonella promoter. In one embodiment, the lysis cassette is Lysin E from phage phiX174, phage iEPS5, or lambda phage. In one embodiment, the promoter is a promoter for one of the genes in Salmonella pathogenicity island 2 type III secretion system (SPI2-T3SS) selected from the group SpiC/SsaB (accession no. CBW17423.1), SseF (accession no. CBW17434.1), SseG (accession no. CBW17435.1), SseI (accession no. CBW17087.1), SseJ (accession no. CBW17656.1 or NC_016856.1), SseK1 (accession no. CBW20184.1), SseK2 (accession no. CBW18209.1), SifA (accession no. CBW17257.1), SifB (accession no. CBW17627.1), PipB (accession no. CBW17123.1), PipB2 (accession no. CBW18862.1), SopD2 (accession no. CBW17005.1), GogB (accession no. CBW18646.2), SseL (accession no. CBW18358.1), SteC (accession no. CBW17723.1), SspH1 (accession no. STM14_1483), SspH2 (accession no. CBW18313.1), or SirP (examples/an embodiment of sequences that can be used in the instant compositions/methods are provided for by accession numbers and sequences provided throughout the specification; other sequences, including those with greater than about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% and 100% identity may also be used in the composition/methods of the invention). SpiC/SsaB (accession no. CBW17423.1): 1 MSEEGFMLAV LKGIPLIQDI RAEGNSRSWI MTIDGHPARG EIFSEAFSIS LFLNDLESLP 61 KPCLAYVTLL LAAHPDVHDY AIQLTADGGW LNGYYTTSSS SELIAIEIEK HLALTCILKN 121 VIRNHHKLYS GGV (SEQ ID NO: 1) SseF (accession no. CBW17434.1): 1 MKIHIPSAAS NIVDGNSPPS DIQAKEVSFP PPEIPAPGTP AAPVLLTPEQ IRQQRDYAIH 61 FMQYTIRALG ATVVFGLSVA AAVISGGAGL PIAILAGAAL VIAIGDACCA YHNYQSICQQ 121 KEPLQTASDS VALVVSALAL KCGASLNCAN TLANCLSLLI RSGIAISMLV LPLQFPLPAA 181 ENIAASLDMG SVITSVSLTA IGAVLDYCLA RPSGDDQENS VDELHADPSV LLAEQMAALC 241 QSATTPALMD SSDHTSRGEP (SEQ ID NO: 2) SseG (accession no. CBW17435.1): 1 MKPVSPNAQV GGQRPVNAPE ESPPCPSLPH PETNMESGRI GPQQGKERVL AGLAKRVIEC 61 FPKEIFSWQT VILGGQILCC SAGIALTVLS GGGAPLVALA GIGLAIAIAD VACLIYHHKH 121 HLPMAHDSIG NAVFYIANCF ANQRKSMAIA KAVSLGGRLA LTATVMTHSY WSGSLGLQPH 181 LLERLNDITY GLMSFTRFGM DGMAMTGMQV SSPLYRLLAQ VTPEQRAPE (SEQ ID NO: 3) SseI (accession no. CBW17087.1): 1 MPFHIGSGCL PAIISNRRIY RIAWSDTPPE MSSWEKMKEF FCSTHQAEAL ECIWTICHPP 61 AGTTREDVVS RFELLRTLAY DGWEENIHSG LHGENYFCIL DEDSQEILSV TLDDVGNYTV 121 NCQGYSETHH LTMATEPGVE RTDITYNLTS DIDAAAYLEE LKQNPIINNK IMNPVGQCES 181 LMTPVSNFMN EKGFDNIRYR GIFIWDKPTE EIPTNHFAVV GNKEGKDYVF DVSAHQFENR 241 GMSNLNGPLI LSADEWVCKY RMATRRKLIY YTDFSNSSIA ANAYDALPRE LESESMAGKV 301 FVTSPRWFNT FKKQKYSLIG KM (SEQ ID NO: 4) SseJ (accession no. CBW17656.1): 1 MPLSVGQGYF TSSISSEKFN AIKESARLPE LSLWEKIKAY FFTTHHAEAL ECIFNLYHHQ 61 ELNLTPVQVR GAYIKLRALA SQGCKEQFII ESQEHADKLI IKDDNGENIL SIEVECHPEA 121 FGLAKEINKS HPKPKNISLG DITRLVFFGD SLSDSLGRMF EKTHHILPSY GQYFGGRFTN 181 GFTWTEFLSS PHFLGKEMLN FAEGGSTSAS YSCFNCIGDF VSNTDRQVAS YTPSHQDLAI 241 FLLGANDYMT LHKDNVIMVV EQQIDDIEKI ISGGVNNVLV MGIPDLSLTP YGKHSDEKRK 301 LKDESIAHNA LLKTNVEELK EKYPQHKICY YETADAFKVI MEAASNIGYD TENPYTHHGY 361 VHVPGAKDPQ LDICPQYVFN DLVHPTQEVH HCFAIMLESF IAHHYSTE (SEQ ID NO: 5) sseJ sequence (DNA) - Accession number-NCBI Reference Sequence: NC_016856.1 ATGCCATTGAGTGTTGGACAGGGTTATTTCACATCATCTATCAGTTCTGAAAAATTTAATGCGATAAAAGAAAGC GCACGCCTTCCGGAATTAAGTTTATGGGAGAAAATCAAAGCATATTTCTTTACCACCCACCATGCAGAGGCGCTC GAATGTATCTTTAATCTTTACCACCATCAGGAACTGAATCTAACACCGGTACAGGTTCGCGGAGCCTACATCAAA CTTCGAGCCTTAGCGTCTCAGGGATGTAAAGAACAGTTTATTATAGAATCACAGGAACACGCCGATAAGTTGATT ATTAAAGATGATAATGGTGAAAATATTTTGTCTATTGAGGTTGAATGTCATCCGGAAGCTTTTGGTCTTGCAAAA GAAATCAATAAATCACATCCCAAGCCCAAAAATATTTCTTTGGGTGATATTACCAGACTGGTATTTTTTGGCGAC AGCTTGTCTGACTCCTTAGGGCGTATGTTTGAAAAAACACATCATATCTTACCCTCCTATGGTCAATACTTTGGC GGAAGGTTTACTAATGGATTTACCTGGACTGAGTTTTTATCATCTCCACACTTCTTAGGTAAAGAGATGCTTAAT TTTGCTGAAGGGGGAAGTACATCGGCAAGCTATTCCTGCTTTAATTGCATCGGTGACTTTGTATCAAATACGGAC AGACAAGTCGCATCTTACACCCCTTCTCACCAGGACCTGGCGATATTTTTATTGGGGGCTAATGACTATATGACA CTACACAAAGATAATGTAATAATGGTCGTTGAGCAACAAATTGATGATATTGAAAAAATAATTTCCGGTGGAGTT AATAATGTTCTGGTCATGGGGATTCCCGATTTGTCTTTAACACCTTATGGCAAACATTCTGATGAAAAAAGAAAG CTTAAGGATGAAAGCATCGCTCACAATGCCCTGTTAAAAACTAATGTTGAAGAATTAAAAGAAAAATACCCCCAG CATAAAATATGCTATTACGAGACTGCCGATGCATTTAAGGTGATAATGGAGGCGGCCAGTAATATTGGTTATGAT ACGGAAAACCCTTATACTCACCACGGCTATGTACATGTTCCCGGGGCTAAAGACCCTCAGCTAGATATATGTCCG CAATACGTCTTCAACGACCTTGTCCATCCAACCCAGGAAGTCCATCATTGTTTTGCCATAATGTTAGAAAGTTTT ATAGCTCATCATTATTCCACTGAATAA (SEQ ID NO: 6) sseJ sequence (protein) MPLSVGQGYFTSSISSEKFNAIKESARLPELSLWEKIKAYFFTTHHAEALECIFNLYHHQELNLTPVQVRGAYIK LRALASQGCKEQFIIESQEHADKLIIKDDNGENILSIEVECHPEAFGLAKEINKSHPKPKNISLGDITRLVFFGD SLSDSLGRMFEKTHHILPSYGQYFGGRFTNGFTWTEFLSSPHFLGKEMLNFAEGGSTSASYSCFNCIGDFVSNTD RQVASYTPSHQDLAIFLLGANDYMTLHKDNVIMVVEQQIDDIEKIISGGVNNVLVMGIPDLSLTPYGKHSDEKRK LKDESIAHNALLKTNVEELKEKYPQHKICYYETADAFKVIMEAASNIGYDTENPYTHHGYVHVPGAKDPQLDICP QYVFNDLVHPTQEVHHCFAIMLESFIAHHYSTE (SEQ ID NO: 7) SseK1 (accession no. CBW20184.1): 1 MIPPLNRYVP ALSKNELVKT VTNRDIQFTS FNGKDYPLCF LDEKTPLLFQ WFERNPARFG 61 KNDIPIINTE KNPYLNNIIK AATIEKERLI GIFVDGDFFP GQKDAFSKLE YDYENIKVIY 121 RNDIDFSMYD KKLSEIYMEN ISKQESMPEE KRDCHLLQLL KKELSDIQEG NDSLIKSYLL 181 DKGHGWFDFY RNMAMLKAGQ LFLEADKVGC YDLSTNSGCI YLDADMIITE KLGGIYIPDG 241 IAVHVERIDG RASMENGIIA VDRNNHPALL AGLEIMHTKF DADPYSDGVC NGIRKHFNYS 301 LNEDYNSFCD FIEFKHDNII MNTSQFTQSS WARHVQ (SEQ ID NO: 8) SseK2 (accession no. CBW18209.1): 1 MARFNAAFTR IKIMFSRIRG LISCQSNTQT IAPTLSPPSS GHVSFAGIDY PLLPLNHQTP 61 LVFQWFERNP DRFGQNEIPI INTQKNPYLN NIINAAIIEK ERIIGIFVDG DFSKGQRKAL 121 GKLEQNYRNI KVIYNSDLNY SMYDKKLTTI YLENITKLEA QSASERDEVL LNGVKKSLED VLKNNPEETL ISSHNKDKGH LWFDFYRNLF LLKGSDAFLE AGKPGCHHLQ PGGGCIYLDA DMLLTDKLGT LYLPDGIAIH VSRKDNHVSL ENGIIAVNRS EHPALIKGLE IMHSKPYGDP YNDWLSKGLR HYFDGSHIQD YDAFCDFIEF KHENIIMNTS SLTASSWR (SEQ ID NO: 9) SifA (accession no. CBW17257.1): MPITIGNGFL KSEILTNSPR NTKEAWWKVL WEKIKDFFFS TGKAKADRCL HEMLFAERAP TRERLTEIFF ELKELACASQ RDRFQVHNPH ENDATIILRI MDQNEENELL RITQNTDTFS CEVMGNLYFL MKDRPDILKS HPQMTAMIKR RYSEIVDYPL PSTLCLNPAG APILSVPLDN IEGYLYTELR KGHLDGWKAQ EKATYLAAKI QSGIEKTTRI LHHANISEST QQNAFLETMA MCGLKQLEIP PPHTHIPIEK MVKEVLLADK TFQAFLVTDP STSQSMLAEI VEAISDQVFH AIFRIDPQAI QKMAEEQLTT LHVRSEQQSG CLCCFL (SEQ ID NO: 10) SifB (accession no. CBW17627.1): MPITIGRGFL KSEMFSQSAI SQRSFFTLLW EKIKDFFCDT QRSTADQYIK ELCDVASPPD AQRLFDLFCK LYELSSPSCR GNFHFQHYKD AECQYTNLCI KDGEDIPLCI MIRQDHYYYE IMNRTVLCVD TQSAHLKRYS DINIKASTYV CEPLCCLFPE RLQLSLSGGI TFSVDLKNIE ETLIAMAEKG NLCDWKEQER KAAISSRINL GIAQAGVTAI DDAIKNKIAA KVIENTNLKN AAFEPNYAQS SVTQIVYSCL FKNEILMNML EESSSHGLLC LNELTEYVTL QVHNSLFSED LSSLVETTKN EAHHQS (SEQ ID NO: 11) PipB (accession no. CBW17123.1): mpitnaspen ilrylhaagt gtkeamksat sprgilewfv nfftcggvrr snerwfrevi gklttsllyv nknaffdgnk ifledvngct iclscgaase ntdpmviiev nkngktvtdk vdserfwnvc rmlklmskhn iqqpdslite dgflnlrgvn lahkdfqged lskidasnad frettlsnvn lvganlccan lhavnlmgsn mtkanlthad ltcanmsgvn ltaailfgsd ltdtklngak ldkialtlak altgadltgs qhtptplpdy ndrtlfphpi f (SEQ ID NO: 12) PipB2 (accession no. CBW18862.1): MERSLDSLAG MAKSAFGAGT SAAMRQATSP KTILEYIINF FTCGGIRRRN ETQYQELIET MAETLKSTMP DRGAPLPENI ILDDMDGCRV EFNLPGENNE AGQVIVRVSK GDHSETREIP LASFEKICRA LLFRCEFSLP QDSVILTAQG GMNLKGAVLT GANLTSENLC DADLSGANLE GAVLFMADCE GANFKGANLS GTSLGDSNFK NACLEDSIMC GATLDHANLT GANLQHASLL GCSMIECNCS GANMDHTNLS GATLIRADMS GATLQGATIM AAIMEGAVLT RANLRKASFI STNLDGADLA EANLNNTCFK DCTLTDLRTE DATMSTSTQT LFNEFYSENI (SEQ ID NO: 13) SopD2 (accession no. CBW17005.1): MPVTLSFGNR HNYEINHSRL ARLMSPDKEE ALYMGVWDRF KDCFRTHKKQ EVLEVLYTLI HGCERENQAE LNVDITGMEK IHAFTQLKEY ANPSQQDRFV MRFDMNQTQV LFEIDGKVID KCNLHRLLNV SENCIFKVME EDEEELFLKI CIKYGEKISR YPELLEGFAN KLKDAVNEDD DVKDEVYKLM RSGEDRKMEC VEWNGTLTEE EKNKLRCLQM GSFNITTQFF KIGYWELEGE 241 VLFDMVHPTL SYLLQAYKPS LSSDLIETNT MLFSDVLNKD YDDYQNNKRE IDAILRRIYR 301 SHNNTLFISE KSSCRNMLI (SEQ ID NO: 14) GogB (accession no. CBW18646.2): 1 MQYAYTSNEA TSNLELLNKW RIESPDIEKE ERNSIYDKII EANHTGSLSI TAHHVTSIPV 61 FPDNLSELNL SSCYTLESIP NLPDGLKSLT ISGNQTIKIS YFPDSLESLS IDMQAYEENY 121 TFPALPYGLK SFTACYGKFL PPLPPHLSSL SLQNFSEILC AELPYKLDKL DLQNCPFLPL 181 MKMLPEELKE LSIELIRTVP GTVIDDILPD KLKKLSINFC DNIKLPVKLP VNLKSINLSS 241 RTPIAWEIPT CNLPAHIDIS TDGYVKLNPE FLTRSDITFS NKPAGDVLSF QPGDVVYGLC 301 KARDRVNTLV NSLYYFSKKD IIIQNTLTDA VWDRKNRAVF NKDEKIAERL NDVQRGIFFR 361 EFLSQHKKYN ITEDKYSDLS NEECWIKTSK AGLEFQTRLR ERSVIFVIDN LVDAISDIAN 421 KTGKHGNSIT AHELRWVYRN RHDDLVKQNV KFFLNGEAIS HEDVFSLVGW DKYKPKNRNR (SEQ ID NO: 15) SseL (accession no. CBW18358.1): 1 MSDEALTLLF SAVENGDQNC IDLLCNLALR NDDLGHRVEK FLFDLFSGKR TGSSDIDKKI 61 NQACLVLHQI ANNDITKDNT EWKKLHAPSR LLYMAGSATT DLSKKIGIAH KIMGDQFAQT 121 DQEQVGVENL WCGARMLSSD ELAAATQGLV QESPLLSVNY PIGLIHPTTK ENILSTQLLE 181 KIAQSGLSHN EVFLVNTGDH WLLCLFYKLA EKIKCLIFNT YYDLNENTKQ EIIEAAKIAG 241 ISESDEVNFI EMNLQNNVPN GCGLFCYHTI QLLSNAGQND PATTLREFAE NFLTLSVEEQ 301 ALFNTQTRRQ IYEYSLQ (SEQ ID NO: 16) SteC (accession no. CBW17723.1): 1 MPFTFQIGNH SCQISERYLR DIIDNKREHV FSTCEKFIDF FRNIFTRRSL ISDYREIYNL 61 LCQKKEHPDI KGPFSPGPFS KRDEDCTRWR PLLGYIKLID ASRPETIDKY TVEVLAHQEN 121 MLLLQMFYDG VLVTETECSE RCVDFLKETM FNYNNGEITL AALGNDNLPP SEAGSNGIYE 181 AFEQRLIDFL TTPATASGYE SGAIDQTDAS QPAAIEAFIN SPEFQKNIRM RDIEKNKIGS 241 GSYGTVYRLH DDFVVKIPVN ERGIKVDVNS PEHRNCHPDR VSKYLNMAND DKNFSRSAIM 301 NINGKDVTVL VSKYIQGQEF DVEDEDNYRM AEALLKSRGV YMHDINILGN ILVKEGVLFF 361 VDGDQIVLSQ ESRQQRSVSL ATRQLEEQIK AHHMIKLKRA ETEGNTEDVE YYKSLITDLD 421 ALIGEEEQTP APGRRFKLAA PEEGTLVAKV LKDELKK (SEQ ID NO: 17) SspH1 (accession no. STM14_1483): 1 MFNIRNTQPS VSMQAIAGAA APEASPEEIV WEKIQVFFPQ ENYEEAQQCL AELCHPARGM 61 LPDHISSQFA RLKALTFPAW EENIQCNRDG INQFCILDAG SKEILSITLD DAGNYTVNCQ 121 GYSEAHDFIM DTEPGEECTE FAEGASGTSL RPATTVSQKA AEYDAVWSKW ERDAPAGESP 181 GRAAVVQEMR DCLNNGNPVL NVGASGLTTL PDRLPPHITT LVIPDNNLTS LPELPEGLRE 241 LEVSGNLQLT SLPSLPQGLQ KLWAYNNWLA SLPTLPPGLG DLAVSNNQLT SLPEMPPALR 301 ELRVSGNNLT SLPALPSGLQ KLWAYNNRLT SLPEMSPGLQ ELDVSHNQLT RLPQSLTGLS 361 SAARVYLDGN PLSVRTLQAL RDIIGHSGIR IHFDMAGPSV PREARALHLA VADWLTSARE 421 GEAAQADRWQ AFGLEDNAAA FSLVLDRLRE TENFKKDAGF KAQISSWLTQ LAEDAALRAK 481 TFAMATEATS TCEDRVTHAL HQMNNVQLVH NAEKGEYDNN LQGLVSTGRE MFRLATLEQI 541 AREKAGTLAL VDDVEVYLAF QNKLKESLEL TSVTSEMRFF DVSGVTVSDL QAAELQVKTA 601 ENSGFSKWIL QWGPLHSVLE RKVPERFNAL REKQISDYED TYRKLYDEVL KSSGLVDDTD 661 AERTIGVSAM DSAKKEFLDG LRALVDEVLG SYLTARWRLN (SEQ ID NO: 18) SspH2 (accession no. CBW18313.1): 1 MPFHIGSGCL PATISNRRIY RIAWSDTPPE MSSWEKMKEF FCSTHQTEAL ECIWTICHPP 61 AGTTREDVIN RFELLRTLAY AGWEESIHSG QHGENYFCIL DEDSQEILSV TLDDAGNYTV 121 NCQGYSETHR LTLDTAQGEE GTGHAEGASG TFRTSFLPAT TAPQTPAEYD AVWSAWRRAA 181 PAEESRGRAA VVQKMRACLN NGNAVLNVGE SGLTTLPDCL PAHITTLVIP DNNLTSLPAL 241 PPELRTLEVS GNQLTSLPVL PPGLLELSIF SNPLTHLPAL PSGLCKLWIF GNQLTSLPVL 301 PPGLQELSVS DNQLASLPAL PSELCKLWAY NNQLTSLPML PSGLQELSVS DNQLASLPTL 361 PSELYKLWAY NNRLTSLPAL PSGLKELIVS GNRLTSLPVL PSELKELMVS GNRLTSLPML 421 PSGLLSLSVY RNQLTRLPES LIHLSSETTV NLEGNPLSER TLQALREITS APGYSGPIIR 481 FDMAGASAPR ETRALHLAAA DWLVPAREGE PAPADRWHMF GQEDNADAFS LFLDRLSETE 541 NFIKDAGFKA QISSWLAQLA EDEALRANTF AMATEATSSC EDRVTFFLHQ MKNVQLVHNA 601 EKGQYDNDLA ALVATGREMF RLGKLEQIAR EKVRTLALVD EIEVWLAYQN KLKKSLGLTS 661 VTSEMRFFDV SGVTVTDLQD AELQVKAAEK SEFREWILQW GPLHRVLERK APERVNALRE 721 KQISDYEETY RMLSDTELRP SGLVGNTDAE RTIGARAMES AKKTFLDGLR PLVEEMLGSY 781 LNVQWRRN (SEQ ID NO: 19) III. Oncolytic Virus Oncolytic viruses (OVs) are an emerging class of cancer therapeutics that offer the benefits of selective replication in tumor cells, delivery of multiple eukaryotic transgene payloads, induction of immunogenic cell death and promotion of antitumor immunity, and a tolerable safety profile that largely does not overlap with that of other cancer therapeutics, such as immune checkpoint inhibitors (ICIs) or chimeric antigen receptors (CARs). The first oncolytic virus immunotherapy to be approved by the FDA was T-VEC, which is used to treat metastatic melanoma. T-VEC is a herpes virus that has been engineered to be less likely to infect healthy cells. OVs directly kill cancer cells. OVs replicate inside cancer cells, causing them to burst and release materials that the immune system can recognize. OVs can be modified to carry genes that boost the treatment's effectiveness. For example, OVs can be engineered to express immune regulators that enhance antitumor immunity. Oncolytic viruses (tumor-targeting viruses to treat cancer) include, but are not limited to, DNA (ssDNA) viruses, double-stranded DNA (dsDNA) viruses, single-stranded RNA (ssRNA) viruses, double-stranded RNA (dsRNA) viruses or a combination thereof. The viruses for use in the invention can include, but are not limited to, adenoviruses, coxsackievirus, herpes simplex virus (HSV), maraba viruses, measles, Newcastle disease virus, picornavirus, reovirus, respiratory syncytial virus, vesicular stomatitis virus, parvoviruses, poxviruses, such as vaccinia virus (VACV) and myxoma virus (MYXV), minute virus of mice (MVM), talimogene laherparepvec (T-VEC, or Imlygic®), and/or poliovirus (such as PVS- RIPO). Any of these viruses can be used in the methods herein. In some embodiments, they are attenuated (similar to that found in some vaccines). See, for example, Gujar et al. Nature Protocols volume 19, pages 2540–2570 (2024), for design, production and testing of oncolytic viruses for cancer immunotherapy. IV. Cancer Treatment Bacteria such as Salmonella, Clostridium and Bifidobacterium have a natural tropism for cancers, such as solid tumors. Types of cancer that can be treated using the methods of the invention include, but are not limited to, solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, osteosarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, pancreatic ductal adenocarcinoma, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodenroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma). In some aspects, the subject is treated with radiation, immunotherapy and/or chemotherapy before, after or during administration of the bacterial cells described herein. V. Administration The invention includes administration of the attenuated bacteria, such as Salmonella, strains described herein and methods for preparing pharmaceutical compositions and administering such as well. Such methods comprise formulating a pharmaceutically acceptable carrier with one or more of the attenuated Salmonella strains described herein. A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF; Parsippany, N.J.) or phosphate buffered saline (PBS). It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of other (undesired) microorganisms. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin. Injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients discussed above. Generally, dispersions are prepared by incorporating the active compound into a vehicle which contains a basic dispersion medium and various other ingredients discussed above. In the case of powders for the preparation of injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously. Oral compositions generally include an inert diluent or an edible carrier. For example, they can be enclosed in gelatin capsules. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. For administration by inhalation, the bacteria are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the bacteria are formulated into ointments, salves, gels, or creams as generally known in the art. Intratumoral injection can be used as well. It is especially advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. When administered to a patient the attenuated Salmonella can be used alone or may be combined with any physiological carrier. In general, the dosage ranges from about 1.0 c.f.u./kg to about 1x1012 c.f.u./kg; optionally from about 1.0 c.f.u./kg to about 1x1010 c.f.u./kg; optionally from about 1.0 c.f.u./kg to about 1x108 c.f.u./kg; optionally from about 1x102 c.f.u./kg to about 1x108 c.f.u./kg; optionally from about 1x104 c.f.u./kg to about 1x108 c.f.u./kg; optionally from about 1x105 c.f.u./kg to about 1x1012 c.f.u./kg; optionally from about 1x105 c.f.u./kg to about 1x1010 c.f.u./kg; optionally from about 1x105 c.f.u./kg to about 1x108 c.f.u./kg. EXAMPLE The following examples are provided in order to demonstrate and further illustrate certain embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof. Example I Introduction Many of the current problems associated with cancer treatment, e.g., metastatic disease and refractory tumors, could be overcome with microbial therapies. Oncolytic viruses (OVs) have the potential to treat many tumors by directly lysing cancer cells and stimulating immune responses that eliminate cancer cells regardless of their location in the body (1-6). Despite this potential, oncolytic viruses have not been effective at treating internal solid tumors (5, 7-10). When injected systemically, OVs are cleared from the blood and do not effectively reach tumors (1, 5, 7-20). This problem could be overcome with a carrier with tumor specific tropism. It has been shown that bacterial therapies predominantly accumulate in tumors after intravenous injections (21-30). Developing a bacterial system to deliver viruses into cancer cells would couple the benefits of these two microbial therapies and focus treatment specifically to cancers. Reported herein, for the first time, is the development of intracellular delivering (ID) Salmonella to carry oncolytic viruses into cancer cells. ID Salmonella is a bacterial system recently developed to deliver macromolecules into cancer cells in tumors (31). ID Salmonella contain a genetic circuit (PsseJ-lysE) that controls the release of plasmids and macromolecules (31). In tumors, Salmonella naturally invades cancer cells (31). After cell invasion, the PsseJ promoter activates lysE, which lyses the bacteria and releases the molecular cargo into the cytoplasm of infected cells (31). Compared to other reported cellular carriers (e.g., exosomes, T cells, and mesenchymal stem cells), Salmonella have a higher therapeutic index. For these other carriers, tumor accumulation is rate limiting and most have accumulation rates below 10% (15, 16). Minute virus of mice (MVM) is a small single-stranded DNA virus of the Protoparvovirus genus that naturally lyses cancer cells (32-37). The prototype strain (MVMp) does not integrate into the genome, is non-infectious, and is non-pathogenic in humans (36, 38, 39). After MVMp infects cells, host polymerase produces NS1 from a double stranded template of the viral DNA (20, 38, 40, 41). NS1 is the initiator protein that creates a site-specific nick in the single-stranded DNA and serves as a helicase to initiate DNA replication (34, 38, 40, 42). This DNA template is used to produce more single-stranded viral genomes, which are assembled in the nucleus into new virus particles (36, 43, 44). Cell lysis kills cells and releases new viral particles that infect neighboring cells and initiates new rounds of infection (5, 10, 20, 45, 46). Because of its small size, the MVMp genome can be included in a standard cloning plasmid (47). When mammalian cells are transfected with an MVMp-containing plasmid, they transcribe NS1 and produce viral particles, similar to natural infection (47, 48). Bacterial delivery of viral DNA is complicated by hairpins in the genomes of many viruses. These hairpins have a role in DNA replication, packaging, and viral infectivity. Palindromic sequences are not well tolerated in Salmonella and are subject to partial or complete deletion (49, 50). MVMp has two imperfect palindromes at the 3’ and 5’ termini that form hairpins in the DNA of the viral genome (32, 34, 41, 43, 47, 51-53). These palindromic regions are primers for replication and are used in virus formation and function (33, 41, 43, 47, 51-53). When transformed into cloning strains of E. coli, homologous recombination forms site-specific deletions in the right-end hairpin of plasmids containing the MVMp genome (51). This deletion is prevented in SURE 2 (‘stop unwanted rearrangement events’ 2) E. Coli (ΔrecB, ΔrecJ, and ΔsbcC), which is restriction minus, endonuclease deficient, and recombination deficient. Salmonella contains two systems for repairing DNA damage and regulating homologous recombination: one for double-stranded breaks (DSBs) and another for single- stranded gap (SSG) repairs (54-56). These systems maintain genetic stability but also modify foreign DNA (54). A key component of the DSB repair system is RecBCD, which is a helicase- nuclease complex that initiates repair by unwinding double-stranded DNA (dsDNA) and creating a single-stranded tail for RecA to bind (54, 57-61). RecA is a central protein in homologous recombination that compares and exchanges complementary DNA sequences (54, 60). Two additional components of the DSB repair system are SbcB and SbcCD (62). SbcCD is a nuclease that cleaves double stranded hairpins (59-61). The primary component of SSG repair system is RecFOR (54, 57). Similar to RecBCD, RecFOR mediates the binding of RecA to single-stranded regions of DNA (54, 59, 61, 63). There is considerable redundancy between these pathways. RecFOR can repair DSBs in the absence of RecBCD, where RecJ functions as an exonuclease to create a single stranded overhang to facilitate the loading of RecA (54, 59, 61). SbcB, in concert with SbcCD, can activate the RecFOR pathway to repair the DSBs in the absence of RecBCD (54, 57-59, 61, 64). Provided herein is the creation of virus-delivering Salmonella (VDS, FIG. 1). To be a suitable carrier for oncolytic viruses, the bacterial vector must (1) preserve the viral genome and (2) deposit it intact into cancer cells. It was hypothesized that 1) DNA delivered by ID Salmonella is expressed by cancer cells, 2) removal of homologous recombination genes from Salmonella prevents hairpin deletion and stabilize the MVMp genome, 3) Salmonella delivery of the MVMp genome produces functional viral particles, and 4) the produced viral particles are infective and kill cancer cells. To test these hypotheses, seven strains of Salmonella were generated with deletions in six genes: recA, recB, recF, sbcB, sbcCD, and recJ. For H1PV and MH1PV sseJ knockouts were created in the strains which could stabilize the hairpins. Virus- delivering Salmonella (VDS) were formed by transforming these homologous-recombination- deficient Salmonella (HRDS) with a plasmid containing the MVMp genome and a second plasmid containing the PsseJ-lysE delivery circuit. Fluorescence microscopy was used to measure the delivery of plasmid DNA to cancer cells with ID Salmonella. Gentamicin invasion assay was used to determine the viability and invasiveness of the HRDS strains. Cancer cells were treated with VDS to measure the production of NS1, the initiation of virus formation, and the cytotoxicity of the delivered virus. Finally, cells were retreated with conditioned media to quantify the infectiveness of released virions. By delivering OVs, VDS can expand the effectiveness of microbial therapy to include more solid tumors and be effective for a broader range of cancer patients. Materials and Methods Bacterial culture All bacterial cultures (SURE 2, Salmonella) were grown in LB (10 g/L sodium chloride, 10 g/L tryptone, and 5 g/L yeast extract). Resistant strains of bacteria were grown in the presence of 100 μg/mL of carbenicillin, 33 μg/mL chloramphenicol, or 50 μg/mL of kanamycin. Bacterial strains and plasmids The MVMp plasmid was provided by Dr. Peter Tattersall (Yale School of Medicine, New Haven, CT). The plasmid is ampicillin resistant and has a ColE1 origin of replication. The ptac-GFP plasmid contains green fluorescent protein (GFP) under control of the constitutive bacterial promoter ptac and a Lysin gene E from ΦX174 bacteriophage under the control of intracellular responsive Salmonella promoter PsseJ (31). This plasmid is chloramphenicol resistant and has a p15A origin of replication. Cell culture Human embryonic kidney cells (HEK-293T, RRID:CVCL_0063), human osteosarcoma cells (U2OS, RRID:CVCL_0042), and murine pancreatic ductal adenocarcinoma cells (KPCY, RRID:CVCL_YM32; ATCC, Manassas, VA) were maintained in low glucose Dulbecco’s modified Eagle medium (DMEM; Sigma Aldrich, St. Louis, MO) with sodium bicarbonate (pH 7.4) and 10% fetal bovine serum (FBS; Cytiva, Marlborough, MA) at 37 °C and 5% CO2. Murine mammary carcinoma cells (4T1, RRID:CVCL 0125) were maintained in Roswell Park Memorial Institute (RPMI 1640; Sigma Aldrich, St. Louis, MO) medium supplemented with 2 g/l of sodium bicarbonate and 10% FBS at 37 °C and 5% CO2. For live-cell microscopy, cells were incubated with DMEM with 20 mM HEPES buffering agent and 10% FBS. GFP expression from a eukaryote promoter after delivery with Salmonella To measure plasmid and gene delivery, two plasmids for delivery and eukaryotic GFP expression were transformed into parental Salmonella (ΔmsbB, ΔpurI, Δxyl) and termed as ID- CMV-Sal. The GFP expression plasmid contained the gene for green fluorescent protein (GFP) under control of the cytomegalovirus (CMV) mammalian expression promoter. The delivery plasmid contained the PsseJ-lysE circuit. Control bacteria were parental Salmonella transformed with CMV-GFP but not the PsseJ-lysE lysis plasmid. Salmonella strains were grown to an OD600 between 0.8-1.0 and added to cultures of 4T1 cells for 2 hours. During this period, the bacteria invaded into the cancer cells. Following the invasion, cells were rinsed with phosphate-buffered saline (PBS) five times and incubated in RPMI with 2 g/L sodium bicarbonate, 10% FBS, and 50 μg/mL gentamicin. The gentamicin in the media eliminates extracellular bacteria. Live cells were imaged 24 hours later to quantify for GFP expression by the cells. The cells were also stained with 10 μg/mL Hoechst 33342 (Invitrogen, Waltham, MA) to identify nuclei. Three regions in each well were randomly selected, and 20 cells per regions were randomly selected and scored based on GFP expression to determine the percentage of the cells expressing GFP. Bacterial gene deletions Seven strains of Salmonella enterica serovar Typhimurium were created to prevent homologous recombination. All genetic deletions (∆recA, ∆recB, ∆sbcCD, ∆sbcB, ∆recF, and ∆recJ) were derived from the parental strain VNP20009 (ΔmsbB, ΔpurI, Δxyl) using a modified lambda red recombination protocol with primers of specific homology regions (68). These genes were deleted to create seven strains (A: ΔrecA; B: ΔrecB; F: ΔrecF; CD: ΔsbcCD; BB’F: ΔrecB, ΔsbcB and ΔrecF; BCDJ: ΔrecB, ΔsbcCD and ΔrecJ; and BB’CDF: ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF, CDS: ΔsbcCD ΔsseJ, BB’CDFS: ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF ΔsseJ). Salmonella was transformed with pkd46 (Yale CGSC E. Coli stock center), grown to OD6000.1, and induced with 10 mM arabinose. Once the OD600 reached 0.6-0.8, the bacteria were centrifuged at 3000 × g for 15 minutes. The pellet was washed twice with ice-cold water. A PCR product for the in-frame deletion of the gene using specific primers (Supplemental Table S1) was amplified from the pkd4 plasmid (accession AY048743.1) containing the FRT- KAN-FRT sequence. The PCR product, which contains 50 bp homology for the gene, was transformed into Salmonella using electroporation. The recovery was plated on kanamycin plates (50 µg/ml) and grown overnight. The colonies were screened for deletion by performing a colony PCR of the junction sites of the inserted PCR amplified products. Colonies with a successful knockout were grown overnight at 43 °C to eliminate pkd46. To create another knockout in the Salmonella strain with a gene deletion, the antibiotic resistance was removed by transforming the pcp20 plasmid into the deleted Salmonella. The colonies were grown overnight at 43 °C to eliminate pcp20. After the elimination of the plasmid, the overnight culture was diluted to approximately 100 CFU/ml and plated on hektoen plates. The colonies which were dark green were confirmed as Salmonella. The colonies were sequentially streaked on hektoen, kanamycin, and chloramphenicol agar to verify the removal of the antibiotics. Hairpin stability and bacterial growth To test hairpin stability, the MVMp plasmid was transformed into each knockout strain by electroporation. Electroporation was performed in 1 mm cuvettes at 1800 V and 25 µF with a time constant of 5 msec. Plasmids were extracted using the ZymoPURE plasmid miniprep kit per the manufacturer’s instructions (Irvine, CA) and sent for sequencing (Massachusetts General Hospital CCIB DNA core facility) To further test hairpin stability, isolated plasmids were digested with SSPI restriction enzyme (New England Biolabs, Ipswich, MA). Strains BB’CDF and CD strains were transformed with a plasmid containing the PsseJ-lysE cassette to deliver the MVMp plasmid and were termed virus-delivering Salmonella (VDS-B and VDS- C, respectively). The growth rate of the knockout strains was determined by inoculating cultures at 5 × 105 CFU/mL, growing at 37 °C, and measuring OD600 every 30 minutes for 16 h on a plate reader. Intracellular invasion To determine the effect of gene deletions on cell invasion, the knockout strains were co-cultured with 4T1 cancer cells. Cells were seeded in a 12-well plate at 30% confluence and incubated for a 24 h in RPMI. Once the cells were 70% confluent, the BB’CDF, CD and BCDJ strains were added to the cells at a multiplicity of infection (MOI) of 20. All Salmonella strains were transformed with a plasmid containing EGFP under a constitutive promoter and grown to an OD600 between 0.8-1.0. Salmonella were added to the cell cultures for 2 hours. Following this invasion period, cultures were rinsed with 1X PBS five times and incubated in RPMI with 2 g/L sodium bicarbonate, 10% FBS, and 50 μg/mL gentamicin. Cells were stained with 10 μg/mL Hoechst 33342 (Invitrogen, Waltham, MA). Multiple regions in a well were randomly selected, and 30 cells/regions were randomly selected and scored based on EGFP expression to quantify the percentage of the cells that were invaded. Virus delivery To measure initiation of virus production, VDS-B and VDS-C were added to the HEK- 293T cells at an MOI of 20. After two hours of invasion, extracellular bacteria were removed with gentamicin. The positive control was the MVMp plasmid transfected into naïve cells using Lipofectamine 3000 per manufacturer’s protocol (Thermo Fisher, Waltham, MA). Thirty-six hours after infection or transfection, cells were scraped from the flasks. Cells were collected and pelleted by centrifugation at 1000g for 5 minutes. Cells were suspended in Laemmli SDS 6x sample buffer (Alfa Aesar, Haverhill, MA). Samples were loaded onto NuPage 4-12% Bis- Tris gels (Invitrogen, Waltham, MA) and separated using electrophoresis. Proteins were transferred to polyvinylidene difluoride membranes (Millipore Sigma, Burlington, MA) and probed using mouse monoclonal anti-NS1 (gift from Dr. Kinjal Majumder) at a 1/500 dilution. Blots were incubated with HRP-conjugated goat anti-mouse IgG (R&D Systems, Catalog # HAF007, RRID:AB_357234; 1/1000 dilution) and visualized using Immobilon Western chemiluminescent HRP substrate (Millipore Sigma, Burlington, MA) on an ImageQuant LAS4000 imaging system (GE Healthcare, Chicago, IL). Delivery was also measured in 4T1, KPCY, and U2OS cells at an MOI of 100. For KPCY and U2OS cells, the anti-NS1 antibody was diluted 1/250 and the anti-mouse IgG was diluted 1/500. Cell viability and death Cell death was quantified after adding VDS to cancer cells. Bacteria (VDS-B or VDS- C) were added to cultures of 4T1 cells at an MOI of 100. Strains without the MVMp plasmid (CD or BB’CDF) were negative controls. The positive control was 4T1 cells transfected with the MVMp plasmid using Lipofectamine 3000. After two hours of invasion, bacteria were cleared with gentamicin. Seventy-two hours after infection/transfection, the cells were fixed with 10% formaldehyde in water for 1 hour at room temperature. Dead cell plaques were stained with crystal violet staining, dead cells were stained with ethidium homodimer and cell viability was determined by MTT assay. For crystal violet staining, the formaldehyde was removed and the cells were stained with 1% crystal violet stain (Acros Organics, Waltham, MA). Cells were incubated in crystal violet for 15 minutes and rinsed with water. Cells were imaged on a Zeiss Axio Observer Z.1 microscope using a 5x objective lens. Images were tiled to image the entire surface. The area of unstained cells was analyzed using MATLAB. To quantify cell death, 1 μg/mL ethidium homodimer (Invitrogen, Waltham, MA) was added to cells 72 hours after infection or transfection. Live cell images were acquired every 10 minutes. The experiment was performed in triplicate, and death was determined in three areas per well. The extent of cell death was determined in ImageJ by measuring the area with red fluorescence compared to the area covered by cells at the time of invasion/transfection. To measure cell viability, MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; Themo Fisher, Waltham, MA) was added to cultures treated with VDS. Cells (4T1, KPCY or U2OS) were seeded on 12-well plates at 30% confluence. Cells were infected with strains VDS-B or BB’CDF (bacterial negative control) at an MOI of 100. The positive control was cells transfected with the MVMp plasmid using lipofectamine. Seventy-two hours after infection/transfection, cells were incubated with 5 mg/ml MTT-solution for 2 hours. After discarding the supernatant, 500 µl isopropanol with 25 N HCl was added to each well. Absorbance was measured at 570 nm (Spectramax ID3). Cell viability was calculated relative to uninfected cells. This assay was performed in triplicate wells for each condition. Re-infection The infectivity of virions formed by VDS was determined by re-infecting cells with conditioned media. Bacteria (VDS-B or BB’CDF) were added to cultures of 4T1 cells for two hours and cleared with gentamicin. Positive control cells were transfected with the MVMp plasmid. Media was collected from infected cells 72 hours following bacterial invasion or transfection. Media was stored at -80 °C until use. Media was thawed at room temperature. The conditioned media was added to fresh media with 50 μg/mL gentamicin, at a 40:60 to HEK- 293T cells. The cells were analyzed 48 hours after re-infection for NS1 protein as described above. The conditioned media, with 50 μg/mL gentamicin, was also added to 4T1 cells plated on 12-wells at 30% confluence. Viability was analyzed 48 hours after re-infection using MTT, as described above. Statistical Analysis A comparison of two populations was performed in Excel (Microsoft Office Professional Plus 2016) using two-tailed, unpaired Student’s t-tests. Comparisons of multiple conditions were performed in GraphPad Prism 9.2.0 using ANOVA with a Bonferroni correction. Values are reported as means ± standard errors (SEMs). Statistical significance was confirmed when P < 0.05. Results ID Salmonella delivers functional plasmid DNA into cancer cells To measure DNA delivery, intracellular delivering (ID) Salmonella enterica serovar Typhimurium (henceforth referred to as Salmonella) was applied to cultures of cancer cells (FIG 2). The delivery of functional DNA is a critical component of a virus carrier system. To visualize DNA delivery and expression, ID Salmonella was transformed with a plasmid containing GFP under control of the cytomegalovirus (CMV) promoter. This promoter only initiates transcription in eukaryotic cells (65). Salmonella with CMV-GFP did not express GFP. In addition to CMV-GFP, ID-CMV-GFP Salmonella contained the PsseJ-lysE circuit, which induces lysis after cell invasion (31). Control Salmonella contained CMV-GFP, but not PsseJ- lysE. When incubated with murine 4T1 mammary cancer cells, ID-CMV-GFP produced GFP (arrows, FIG 2A). Compared to controls, ID-CMV-GFP produced significantly more GFP (P < 0.001, FIG. 2B). This difference indicates that the lysis circuit is needed for delivering plasmids into cancer cells. GFP expression also indicates that after bacterial lysis, released plasmids were transported to the nucleus and delivered genes were translated by recipient cells. Parental Salmonella disables MVMp replication When not modified, parental Salmonella (ΔmsbB, ΔpurI, Δxyl) deletes 97 base pairs from the right-end hairpin of MVMp (FIG. 3). The genome of MVMp contains two palindromes at either end that are primers for replication and are needed for virus formation. The genome for MVMp is on a plasmid that also contains a ColE1 prokaryotic origin of replication, and ampicillin resistance (FIG.3A). Transcription of MVMp is initiated by the P4 promoter within its genome in a mammalian nucleus (44). Replicating this plasmid in SURE 2 recombination-deficient E. coli (ΔrecB, ΔrecJ, and ΔsbcC), maintained both hairpins (FIG. 3B). SSPI digestion of the MVMp plasmid produced three fragments: 5078, 1395 and 732 bp (FIG. 3B, left). The smallest fragment contains the right-end hairpin. When replicated in parental Salmonella, a 97 bp section was deleted from the right hairpin, reducing the third fragment to 635 bp (FIG. 3B, right). The location of this deletion from the center of the palindrome in the right end hairpin was confirmed by sequencing (FIG.3C). Without the right- end hairpin, the MVMp genome is inactive and NS1 is not produced (FIG. 3D). To measure virus initiation, MVMp plasmids were maintained in either SURE 2 or parental Salmonella and transfected into HEK-293T cancer cells. The plasmids with intact hairpins formed the replication initiating NS1 protein (FIG. 3D). Plasmids that were maintained in parental Salmonella did not form NS1 (FIG. 3D). Taken together, these results indicate that parental Salmonella deletes the right-end hairpin, which renders the virus replication incompetent. Deletion of homologous recombination genes stabilizes the right-end hairpin of MVMp To create a bacterial strain to deliver functional virus, homologous recombination genes were deleted from Salmonella (FIG. 4). Seven strains were generated with different combinations of deletions of six genes (recA, recB, recF, sbcB, sbcCD, and recJ) that are components of the homologous-recombination machinery. These were termed homologous- recombination-deficient Salmonella (HRDS) strain A (ΔrecA), strain B (ΔrecB), strain F (ΔrecF), strain CD (ΔsbcCD), strain BB’F (ΔrecB, ΔsbcB and ΔrecF), strain BCDJ (ΔrecB, ΔsbcCD and ΔrecJ), and strain BB’CDF (ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF). The BCDJ strain has the same deletions as SURE 2 E. coli (ΔrecB, ΔsbcCD and ΔrecJ). For H1PV and MH1PV plasmids two more strains with sseJ knockout were created in CD and BB’CDF and were termed as CDS: (ΔsbcCD ΔsseJ), BB’CDFS (ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF ΔsseJ). When the MVMp plasmid was replicated in each of these seven bacterial strains, four of the strains deleted 97 bp from the right-end hairpin (A, B, F and BB’F), as shown by SSPI restriction digest (FIG. 4A) and DNA sequencing (FIG. 4B). Three of the knockout stains maintained the hairpin (CD, BCDJ and BB’CDF; FIGS.4A, B). Two of strains that maintained the right-end hairpin (CD and BB’CDF) also maintained virus function (FIG. 4C). The MVMp plasmid was replicated in CD and BB’CDF, purified, and transfected into HEK-293T cells. After transfection, all of these cells formed NS1 (FIG. 4C), demonstrating the initiation of virus formation. To show the durability of virus replication, the MVMp plasmid was maintained in CD, BB’CDF and SURE 2 for three passages. After each of the generations, both CD and BB’CDF maintained the full-length plasmid and the right- end hairpin (similar to SURE 2, FIG. 4D). Deletion of homologous recombination genes does not impair growth or invasion For strains CD and BB’CDF, which stabilized the right-end hairpin, deletion of homologous recombination genes did not affect bacterial growth or invasiveness into cancer cells (FIG.5). In culture, the growth of strain CD and BB’CDF were comparable with parental Salmonella (FIG. 5A). The growth of strain BCDJ, however, was impaired. The deletions in BCDJ delayed growth by more than six hours (FIG. 5A). At nine hours after inoculation, the density of BCDJ was 4.3 times lower than parental Salmonella (P < 0.0001, FIG. 5B). The densities of CD and BB’CDF were comparable to parental Salmonella at nine hours (FIG.5B). Similar to growth, the CD and BB’CDF strains invaded into cancer cells at comparable rates to parental Salmonella (FIG.5C-F). To measure invasiveness, the Salmonella strains (CD, BB’CDF, BCDJ and parental) were co-cultured with 4T1 murine mammary cancer cells for two hours and extracellular bacteria were cleared with gentamicin. After four hours, bacteria were inside cells (arrows) for the CD, BB’CDF and parental strains (FIG. 5C). Few BCDJ were present inside cells (FIG.5C). There was no difference in the percentage of cells invaded by CD and BB’CDF compared with parental Salmonella (FIG. 5D, E). The percentage of invaded cells was significantly lower for BCDJ compared to parental Salmonella (P < 0.05, FIG. 5F). Because of its poor ability to grow and invade cancer cells, the BCDJ was not used. Engineered Salmonella delivers MVMp to cancer cells and induces lysis To create virus-delivering Salmonella (VDS), HRDS were transformed with two plasmids: one that contains the macromolecular delivery system (PsseJ-lysE) and a second that contains the MVMp genome. Transforming the two strains that maintained the right-end hairpin (BB’CDF and CD) created two strains: VDS-B (BB’CDF) and VDS-C (CD). To test the delivery of MVMp, both strains of VDS were applied to cultures of 4T1 cells (FIG. 6A). After two hours of co-culture, extracellular bacteria were removed with gentamicin (FIG.6A). After a further 36 hours to allow for plasmid release, transport to the nucleus and gene expression, the cells cultured with VDS-B produced the viral NS1 protein (FIG. 6B). NS1 production was comparable to positive control cells that were directly transfected with the MVMp plasmid using lipofectamine (FIG. 6B). As expected, no NS1 was produced by uninfected cells. Cells co-cultured with VDS-C did not produce NS1 (FIG.6B). Bacterial delivery of MVMp with VDS-B caused cell death (FIG. 6C-H). To measure cell death, either VDS-B or control bacteria were applied to 4T1 cells (FIG. 6A). These cells were compared to cells that were directly transfected with the MVMp plasmid. The bacterial control was the BB’CDF strain, which did not contain the MVMp plasmid but was transformed with the delivery (PsseJ-lysE) plasmid, and had the same knockouts (ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF) as VDS-B. Seventy-two hours after bacterial invasion, cells treated with VDS-B formed virus-infected, dead cell plaques (FIG. 6C). The area of the plaques was significantly more than bacterial controls (P < 0.05, FIG. 6D). Over time, VDS-B caused death similarly to transfection controls (arrows in FIGS. 6E-F). In comparison, there was minimal death in bacterial controls (FIGS.6E-F). The area of cell death increased at a similar rate after infection with VDS-B or transfection with MVMp (FIG. 6F). At 72 h, MVMp, delivered by VDS-B, significantly increased the area of cell death compared to control bacteria (P < 0.05, FIG.6G). A secondary method, MTT, also demonstrated that VDS-B caused more cell death than bacterial controls (P < 0.05, FIG. 6H)). Unlike VDS-B, VDS-C did not cause cell death (FIG. 6I). Over time, the area of cell death did not increase for cells treated with VDS-C. At 72 h, there was no difference in cell death between VDS-C and strain HRDS-CD, its bacterial control (FIG.6I). Direct transfection of MVMp caused more cell death than VDS-C (P < 0.001, FIG. 6I). Lack of virus formation and undetectable cell death shows limited utility for VDS-C (compared to VDS-B). VDS creates functional virus particles that infect new cells Delivery of MVMp by VDS forms functional and infective virions (FIG.7). To measure the functionality of produced viruses, VDS was applied to cultures of 4T1 cancer cells for two hours (FIG. 7A). After this time for invasion, the cells were washed and fed with complete media containing gentamicin. After an additional 72 h, the culture medium was removed and added to fresh media with gentamicin, at a 40:60 ratio. The gentamicin in these media prevented bacterial re-infection in the secondary cultures. This media mixture was added to a second culture of HEK-293T cells (FIG.7A). After 48 h in this conditioned media, the HEK- 293T cells that received VDS-B media produced NS1 (FIG. 7B). Conditioned media from primary cultures that had been directly transfected with MVMp also produced NS1. Media from cells treated with VDS-C did not form NS1 (FIG. 7B). These results show that delivery of the MVMp plasmid with VDS-B forms full virions particles that are infective to naïve cells. The viral particles produced by VDS-B are cytotoxic to cancer cells (FIG. 7C). A similar procedure was used to measure induced cell death. There were a couple of key differences. Primary cells were treated with VDS-B or control bacteria (strain BB’CDF) to control for compounds released by bacteria into the conditioned media. Recipient cells in the secondary cultures were 4T1 cancer cells not HEK-293T. The conditioned media from cells treated with VDS-B killed 50% of the recipient cells, which was significantly more than bacterial controls (P < 0.001, FIG. 7C). Cell death was equivalent to cells treated with conditioned media from MVMp transfected cultures (Figure 7C). These results show that the virions produced by treating cells with VDS-B are cytotoxic. VDS-mediated cell death is independent of cancer type When applied to multiple cancer cell types, VDS induced the formation of cytotoxic viral particles that kill cells and infect neighboring cells (FIG.8). To measure virus formation, VDS-B was applied to HEK-293T human embryonic kidney cells, U2OS human osteosarcoma cells, and KPCY murine pancreatic ductal adenocarcinoma cells (FIG. 8A). A similar procedure was used as FIG. 6A. As with 4T1 cells, MVMp is infective in these cells — NS1 was produced by cells that were directly transfected with the MVMp plasmid using lipofectamine (FIG. 8A). When VDS-B was applied for two hours, these cells also produce NS1 (FIG. 8A). VDS-C did not produce NS1 in any of the cell types. The delivery of MVMp with VDS killed osteosarcoma and pancreatic ductal adenocarcinoma cells (FIG. 8B). At 72 h after infection of KPCY cells, VDS-B significantly reduced viability compared to bacterial controls (strain BB’CDF; P < 0.01, FIG.8B). In U2OS cell, MVMp delivered by VDS-B also significantly reduced viability compared to bacteria controls (P < 0.0001, FIG.8B). As with results in FIG 6, VDS-C did not induce death in either of these cell lines. When applied to osteosarcoma and pancreatic ductal adenocarcinoma cells, VDS produced infective viral particles (re-infection, FIG.8C). A similar procedure was used as FIG. 7A. VDS was added to cell cultures for 2 hours, extracellular bacterial were killed with gentamicin, and conditioned media was added to cultures of HEK-293T cells. After 48h in conditioned media, the HEK-293T cells that received media from VDS-B cultures produced NS1 (FIG. 8C). Virus was also present in the culture media from cells transfected with the MVMp plasmid (FIG. 8C). No virus was produced by cultures treated with VDS-C. Viewed together, these data demonstrate that MVMp delivery by VDS resulted in the production of viable virus particles that kill infected cancer cells, independent of cell type. Discussion A bacterial vector was created that delivers oncolytic viruses into cancer cells. The genome for the virus (MVMp) was contained on a plasmid that the bacteria propagated without deleting palindromic sections that are essential for virus infection and replication (FIGS. 3-5). When these virus-delivering Salmonella (VDS) encounter cancer cells, they invade, and deposit the plasmid into the cellular cytoplasm. After the delivered plasmid reaches the nucleus, the cell expresses the viral genes (FIGS. 2, 6). Production of initiating proteins (here NS1) begins the process of forming and assembling viral particles in the cells (FIG.6). Once formed, these virions kill the invaded cancer cells (FIG.6) and spread into the environment. There the viruses invade and kill naïve cells (FIG.7). The release of virions by infected cells perpetuates and amplifies the infection. The results show that deletion of homologous recombination genes enables Salmonella to carry and maintain the MVMp genome (FIGS. 3-5). This parvovirus genome contains a right-end hairpin that is unstable in Salmonella (FIG. 3). VDS was designed to prevent this deletion. Of the seven knockout stains created, one (BB’CDF) performed the best. Previous research has shown that deletion of recA stabilized the herpesvirus genome (66), suggesting that this deletion may stabilize MVMp. However, deletion of the recA gene did not prevent hairpin deletion (FIG.4). It has also been reported that E. coli lacking recB, sbcB, and recF stably propagates the MVMp plasmid (51). In Salmonella, however, neither this combined deletion (strain BB’F) nor the individual ΔrecB (strain B) and ΔrecF (strain F) deletions maintained the right-end hairpin. This difference suggests that there are differences in homologous recombination between Salmonella and E. coli that affect palindromic DNA. All the remaining strains (CD, BCDJ and BB’CDF) did not delete the right-end hairpin. The common knockout in each of these strains (ΔsbcCD) suggests that this nuclease is a primary cause of hairpin deletion. Although these strains maintained the full MVMp genome, two (CD and BCDJ) were not good candidates for viral delivery. Strain BCDJ, which has the same knockouts as SURE 2 E coli, grew poorly and did not efficiently invade into cells (FIG. 5). Strain CD lacked the ability to deliver viral DNA into cancer cells and propagate a virus infection (FIGS. 6-8). The combination of ΔsbcCD with ΔrecB, ΔsbcB, and ΔrecF (BB’CDF) enabled DNA delivery and propagation of parvovirus within several cancer cell lines (FIGS. 6-8). One reason for this difference might be that the deletion of recB makes the BB’CDF strain more attenuated, increases its sensitivity to lysis, and increases plasmid delivery once intracellular. Because of its broad applicability, VDS can be a universal platform for virus delivery to tumors. A strength of oncolytic viruses as a cancer therapy is that the mechanism of cell lysis is independent of the biology of the infected cell and therefore appropriate for treating multiple cancer types. Herein, it was shown that VDS delivers functional viruses to multiple cancer types, including breast carcinoma, pancreatic carcinoma, and osteosarcoma (FIG. 8). In addition, the elimination of homologous recombination genes from VDS, makes it suitable for delivering multiple types of viruses. As demonstrated here, VDS was used to deliver MVMp, which is a single-stranded DNA (ssDNA) virus. VDS, however, could also deliver double- stranded DNA (dsDNA), single-stranded RNA (ssRNA) and double-stranded RNA (dsRNA) viruses. As a cargo vector, VDS enables oncolytic virus treatment of solid tumors. After systemic injection, VDS would preferentially accumulate in tumors over other organs, similar to other Salmonella vectors (31, 67). Delivery of the viral genome would produce infective virus particles that can initiate successive rounds of infection (FIG. 7) and mediate a potent bystander effect. Salmonella delivery would increase the delivery of viruses to the tumor microenvironment. Combining the benefits of Salmonella delivery and oncolytic virus therapy can provide an effective therapy for many cancers. Example II Engineered Salmonella deliver and launch Parvovirus (H1PV) infection in tumors. Salmonella delivers functional H1PV virions that are cytotoxic to liver cancer cells: Two strains of homologous recombination deficient Salmonella, BB’CDF (ΔrecB, ΔsbcB, ΔsbcCD and ΔrecF) and CDS (ΔsbcCD and ΔsseJ) maintained the H1PV right-end hairpin. Two test the stability of the plasmid, SSPI and EcoRI restriction digest was performed and the size of the two bands were same as from the plasmid maintained in the SURE 2 strain (FIG. 14A). To test whether, the plasmid from these strains could generate functional virus, plasmid was transfected into HEK-293T cells. 36 hours post transfection cells made NS1 protein, an early viral protein same as the plasmid maintained in SURE 2 (FIG.14B). To determine whether Salmonella could deliver the viral plasmid into cancer cells, BB’CDF and CDS strains of Salmonella was transformed with the H1PV plasmid and a plasmid containing the intracellular delivery circuit. The strains containing both the plasmid were termed as H1PV VDS-B when transformed in BB’CDF and H1PV VDS-C when transformed into CDS. H1PV VDS-B and VDS-C were co-cultured with HUH7 (human liver cancer cells) at a Multiplicity of infection (MOI) of 100.2 hours post infection, the cells were washed 3X with PBS and added new media with gentamycin.36 hours post cells were scraped and looked for NS1 protein. Both the strains of Salmonella were able to deliver the viral plasmid in human liver cancer cell similar to the transfection of the viral plasmid. (FIG. 14C). The delivery of the H1PV plasmid to HUH7 cells was cytotoxic and resulted in a significant decrease in relative cell viability when compared to the control which was just the respective strain transformed with the plasmid with intracellular delivery circuit (P<0.01) (FIG. 14D). The same trend in the relative cell viability was seen when applied to other human liver cancer cell line HeP3B (P<0.05, P<0.01) (FIG.14E) and SK-Hep-1 (P<0.0001) at an MOI of 50 (FIG. 14F). When applied to the mouse hepatoma cell line (Hepa1-6), H1PV plasmid delivery by VDS-B at a MOI of 100 resulted in the generation of NS1 protein (FIG. 14G). The resulted delivery significantly reduced the cell viability as compared to the control (P<0.05) (FIG.14H). VDS-C strain did not result in as strong delivery of the viral plasmid as seen on the western blot (FIG. 14G). There was also no decrease in the cell viability with VDS-C as compared to the respective control (FIG.14H). Viewed together, these data demonstrate that the homologous recombination strains of Salmonella were able to stably maintain the H1PV plasmid that resulted in a successful viral infection. The same strains when transformed with an intracellular delivery circuit plasmid could deliver the viral plasmid in both human and mouse liver cancer cells. Although, BB’CDF was better at delivering the viral plasmid in the cancer cells than the CDS. Therefore, BB’CDF strain was carried forward for all the in vivo experiments. This delivery resulted in a significant decrease in the cell viability in all the above cell lines. Engineered Salmonella delivers H1PV in tumors that creates functional virions: 1*107 CFU of H1PV VDS-B was injected intravenously (IV) into tumor free mice. The mice injected regained their weights in three days after injection (FIG.15A). After 7 days blood was collected from mice injected with H1PV VDS-B and saline and the serum was analyzed for complete cytokine panel. There was no difference in the cytokine levels between mice injected with H1PV VDS-B and saline (FIG.15B). To confirm the delivery of the viral plasmid inside cells, 4*107 CFU of bacteria was injected in mice intratumorally (IT) in subcutaneous liver hepatoma (Hepa1-6) tumors between 250-450mm3. 5 days post injection tumor was harvested, processed, and stained for viral protein, NS1 on a western blot (FIG.15C). NS1 band (83KDa) was visible in the tumor samples of the mice (n=3) injected with H1PV VDS-B, whereas the mice injected with saline (n=3) had no NS1 band (FIG.15D). To measure the delivery of the functional virions and their ability to start de novo infection, mice with tumors injected with 4*107 CFU of H1PV VDS-B were harvested and processed. Functional virions were harvested from the tumors and the supernatant containing the virus was put on HEK-293T cells. 72 hours post infection, the HEK-293T cells were analyzed for the NS1 viral protein (FIG. 15E). The tumors injected with H1PV VDS-B had NS1 band when the supernatant from these tumors were put on HEK-293T cells whereas the supernatant from tumors injected with saline and H1PV VDS-C did not show up a NS1 band on the immunoblot (FIG.15F). These results taken together demonstrate that engineered Salmonella can deliver viral plasmid into the cancer cells in vivo. The plasmid can then generate functional virions that can start de novo infection. H1PV delivery by Salmonella reduces tumor volumes and increases survival: 4*107 CFU of H1PV VDS-B was injected IT in Hepa1-6 tumors between the size of 30-130mm3. BB’CDF was used as a control and the same amount was injected. The tumors were measured every three days until the maximum tumor burden was reached (1000mm3). Injections were given until the last control died, a total of six injections were given every six days to both control and experimental group (FIG.16A). Mice injected with H1PV VDS-B had reduced tumor volumes as compared to the mice injected with mice injected with control (FIG.16B). All control mice (n=5) reached maximum tumor burden and were euthanized, where the last control mice died on day 33 (FIG.16C). For the experimental group none of the mice (n=4) were even close to tumor burden. They remained of the same size as when they were started (FIG.16C). There was an 84% reduction in tumor volume between the control and the experimental group which was highly significant (P<0.01) (FIG.16D). The delivery of virus by Salmonella significantly increased the survival of mice as compared to the control (P<0.01) (FIG. 16E). These results show that the H1PV delivery by engineered Salmonella significantly reduces tumor volume and increases survival. Intravenous (IV) delivery of H1PV VDS-B is as efficacious as intratumoral (IT) delivery: 4*107 CFU of H1PV VDS-B or BB’CDF was injected IV in Hepa1-6 tumors between the size of 50-180mm3.6 injections were given, six days apart and tumors were measured every three days until maximum tumor burden was reached (FIG.17A). All the mice(n=) in control reached maximum tumor burden where the last control died on day 21, while the mice in the experimental group (n=) were almost the same size as when they started (FIG.17B). There was a significant reduction in tumor volumes in the experimental group as compared to the control on day 15 (FIG. 17D). The efficacy was stopped when the first control mice died. The mice in experimental also survived significantly longer than the mice in the control group (P<0.05) (FIG.17E). When IV injected mice were compared with IT injected mice of H1PV VDS-B there was no difference in the tumor volumes (FIG. 17E) while there was a significant reduction in the tumor volumes of the in the IV group as compared to mice injected with IT injection of BB’CDF (P<0.05) (FIG. 4E). There was a 79% reduction in tumor volumes in the IV injected group as compared to the group injected with BB’CDF IT (FIG. 17E). There was also no difference in the survival of the mice in the group where H1PV VDS-B was injected IV and H1PV VDS-B injected IT (FIG.17F). There was a significant difference in the survival of the group injected with H1PV VDS-B IV as compared to the group injected with BB’CDF IT (FIG. 17F). Taken together, these results prove that Salmonella can deliver the virus effectively in the tumors when administered systemically. This solves the problem of the virus getting sequestered into different organs and thereby the viral load decreases in tumor. There was difference in the efficacy and survival of viral delivery IT and IV which circumvents the problem of reduction in viral load in tumors. H1PV delivery by Salmonella causes reduction in tumor weights, cancer cells and increases immunostimulatory innate immune cells: 4*107 CFU of H1PV VDS-B was injected IT in Hepa1-6 tumor model with BB’CDF as a control. 3 injections were given, six days apart and five days after the last injection mice were euthanized and tumor were harvested (FIG.18A). The tumors were measured every three days until the first control mice died. There was a significant reduction in tumor volumes of the mice in experimental group (n=4) as compared to the control (n=3) (P<0.05). There was 78% reduction in tumor volumes in the experimental group as compared to the control (FIG. 18B). This reduction in tumor volumes also manifested in tumor weights. After the tumor was harvested, necrosis was removed from all the mice and the live tumor weight was measured. The average tumor weight of the control group was 1.87gms as compared to 1.22gms of the experimental group (FIG.18C). There was a 35% reduction in the live tumor weights between the control and the experimental group (FIG. 18C). This reduction in tumor weights was significant (P<0.05) (FIG.18C). The tumor was then stained for Epcam positive cells which is pan cancer cell marker. There was over a 2-fold reduction in the Epcam positive cells in mice injected with H1PV VDS-B as compared to mice injected with BB’CDF (P<0.05) (FIG.18D). The tumor was also stained for all the innate cells; Natural killer cells (Npk46 marker), Macrophages (F4/80 marker). There was a significant increase in the NK cells (P<0.05) (FIG. 17E) and Macrophages (P<0.05) (FIG. 18F) in the experimental group as compared to the control group. The H1PV VDS-B tumors also have a significant decrease in the CD45+ cells as compared to the control tumors which is a pan immune cell marker (P<0.05) (FIG.18G). Since all the other cells (NK cells, Macrophages and CD3+ cells) had a significant increase in mice injected with H1PV VDS-B. This reduction could be attributed to the reduction in neutrophils. So, there was a significant reduction in the neutrophil population in the experimental group as compared to the control group (P<0.01) (FIG. 18H). The increase in neutrophils is associated with tissue damage and poor prognosis of the tumor. In the population of macrophages, cells were stained for tumor inhibiting macrophages (M1) (CD80+) and tumor promoting macrophages (M2) (CD206+) (FIG.18I). There was over a 2-fold increase in the ratio of M1 to M2 macrophages (P<0.05), suggesting that tumors injected with H1PV VDS-B were shifting to immunostimulatory microenvironment than immunosuppressive microenvironment (FIG.18J). H1PV delivery be engineered Salmonella activates adaptive immunity and generates tumor memory: 4*107 CFU of H1PV VDS-B was injected IT with Hepa1-6 tumor model with BB’CDF as a control. 3 injections were given, six days apart and five days after the last injection mice were euthanized and tumor were harvested (FIG.19A). The tumor was processed and stained for pan T cells (CD3 marker), cytotoxic T cells (CD8 marker), Helper T cells (CD4 marker), Regulatory T cells (CD25 marker). Mice injected with H1PV VDS-B had over a 4-fold increase in the CD3+ cells as compared to the bacterial control group (P<0.05) (FIG. 19B). The cells were then stained for CD8 T cells and regulatory T cells (FIG. 19C). There was a 12-fold increase in the ratio of CD8+ to CD4+CD25+ cells (P<0.05) (FIG. 19D). There was a significant increase in CD4+CD25- T cells which will probably be Th1cells (P<0.05) (FIG.19E). Cells that were CD8+ were further stained for an activation maker (CD44) (FIG. 19F). There was approximately a six-fold increase in the activated T cells in the experimental group as compared to the control group (P<0.05) (FIG. 19G). Of the cells that were activated, how many of them were still granulated meaning T cells that were not exhausted. The granulatory of the T cells was determined using side scatter. There was an approximately a four-fold increase in the T cells that were granulated in the H1PV VDS-B mice group as compared to the control group (P<0.05) (FIG.19H). The virus delivery can recruit both virus specific T cells and cancer specific T cells. To check whether the T cells recruited in the tumor microenvironment were cancer specific. The mice were injected with H1PV VDS-B three times, six days apart with 4*107 CFU. Five days post the last injection, spleens were then extracted from the mice. The spleen was processed and the splenocytes were then injected intraperitonially (IP) in the naïve mice. Two weeks post the injection of splenocytes naïve mice were rechallenged with 2.5 million Hepa1-6 cells (FIG. 19I). To determine the take rate of the Hepa1-6 cells, a million cells were injected in the naïve mice. The mice not injected with splenocytes derived from H1PV VDS-B mice tumors took in 75% mice of the mice whereas the mice injected with the splenocytes no mice grafted tumors (FIG.19J). The tumors injected were monitored for 66 days. All the tumors in the mice injected with saline grew while the mice injected with the splenocytes had pin pricks but then regressed and the mice remained tumor free for 66 days (FIG.19K). Together these results demonstrate that the viral delivery by engineered Salmonella activates adaptive immune system. This delivery recruit tumor specific T cells and these T cells are activated generating a tumor specific immunity. Engineered Salmonella colonize tumor and is safe 1*107 CFU BB’CDF strain of Salmonella with the intracellular delivery plasmid was injected intravenously in the mice having a Hepa1-6. 72 hours post injection tumor along with liver, spleen, lungs, kidney, heart, and brain were harvested to determine the colonization of bacteria in different organs (FIG.20A). Engineered Salmonella colonized tumors significantly higher than any other healthy organs (FIG. 20B). The colonization in the tumors was over 50 million-fold higher than in liver and spleen (FIG.20B). Engineered strain cleared out of healthy organs faster than the parental VNP20009 strain transformed with the delivery plasmid (FIG. 20C). Colonization of BB’CDF as compared to the parental strain was approximately 0.02 times in the liver, spleen, lungs, and brain; 0.0063 times in the kidneys; 0.03 times in the heart whereas the colonization in the tumor was almost double (FIG. 20C). Weights were measured for the mice injected with 1*107 CFU of BB’CDF, parental strain and saline (FIG.20D). The mice injected with engineered Salmonella gained weights in 3 days and cleared out the infection quickly whereas the mice injected with parental Salmonella took 7 days to reach the same weight gain (FIG. 20E). 7 days post injection, blood from mice was collected using cardiac puncture and the serum was analyzed for cytokines (FIG. 20D). There was no difference in the cytokine levels between BB’CDF and saline (FIG.20F). There was a significant difference in the levels of IL-6 (P<0.05) (FIG.20G) and TNFα (P<0.01) (FIG. 20H) between BB’CDF and parental Salmonella, the two most important cytokines responsible for cytokine storm. To determine the maximum tolerable dose (MTD), BB’CDF and parental Salmonella strain were injected IV in mice. 5*107 CFU of BB’CDF bacteria was very well tolerated in mice with weight loss of around 10% but with the parental strain the weight loss was around 15% (FIG. 20I). After 7 days the mice were rechallenged with another same dose of the bacteria. After the second dose of parental Salmonella, mice died whereas mice injected with BB’CDF regained their weights after 3 days of injection (FIG. 20I). Mice were then injected with 1*108 CFU of BB’CDF strain. After the first injection, mice lost around 12% of their body weights but after second injection they lost about 18% of their body weights (FIG.20I). Taken together, all these results indicate that the engineered Salmonella strain colonizes tumors and clears out of healthy organs better than the parental strain. The strain is also significantly safer than the parental strain. Bibliography 1. Zou H, Mou XZ, & Zhu B (2023) Combining of Oncolytic Virotherapy and Other Immunotherapeutic Approaches in Cancer: A Powerful Functionalization Tactic. Glob Chall 7(1):2200094. 2. Shi T, Song X, Wang Y, Liu F, & Wei J (2020) Combining Oncolytic Viruses With Cancer Immunotherapy: Establishing a New Generation of Cancer Treatment. Front Immunol 11:683. 3. Kaufman HL, Kohlhapp FJ, & Zloza A (2015) Oncolytic viruses: a new class of immunotherapy drugs. Nature Reviews Drug Discovery 14(9):642-662. Lin D, Shen Y, & Liang T (2023) Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 8(1):156. Hu PY, et al. (2020) The limiting factors of oncolytic virus immunotherapy and the approaches to overcome them. Appl Microbiol Biotechnol 104(19):8231-8242. Li K, et al. (2022) Advances in the clinical development of oncolytic viruses. Am J Transl Res 14(6):4192-4206. Mondal M, Guo J, He P, & Zhou D (2020) Recent advances of oncolytic virus in cancer therapy. Hum Vaccin Immunother 16(10):2389-2402. Mahasa KJ, et al. (2020) Mesenchymal stem cells used as carrier cells of oncolytic adenovirus results in enhanced oncolytic virotherapy. Sci Rep 10(1):425. Rahman MM & McFadden G (2021) Oncolytic Viruses: Newest Frontier for Cancer Immunotherapy. Cancers (Basel) 13(21). Kwan A, Winder N, & Muthana M (2021) Oncolytic Virotherapy Treatment of Breast Cancer: Barriers and Recent Advances. Viruses 13(6). Davis JJ & Fang B (2005) Oncolytic virotherapy for cancer treatment: challenges and solutions. J Gene Med 7(11):1380-1389. Reale A, Calistri A, & Altomonte J (2021) Giving Oncolytic Viruses a Free Ride: Carrier Cells for Oncolytic Virotherapy. Pharmaceutics 13(12). Fukuhara H, Ino Y, & Todo T (2016) Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci 107(10):1373-1379. Ferguson MS, Lemoine NR, & Wang Y (2012) Systemic delivery of oncolytic viruses: hopes and hurdles. Adv Virol 2012:805629. Roy DG & Bell JC (2013) Cell carriers for oncolytic viruses: current challenges and future directions. Oncolytic Virother 2:47-56. Willmon C, et al. (2009) Cell carriers for oncolytic viruses: Fed Ex for cancer therapy. Mol Ther 17(10):1667-1676. Power AT, et al. (2007) Carrier cell-based delivery of an oncolytic virus circumvents antiviral immunity. Mol Ther 15(1):123-130. Shalhout SZ, Miller DM, Emerick KS, & Kaufman HL (2023) Therapy with oncolytic viruses: progress and challenges. Nat Rev Clin Oncol 20(3):160-177. Allaume X, et al. (2012) Retargeting of rat parvovirus H-1PV to cancer cells through genetic engineering of the viral capsid. J Virol 86(7):3452-3465. Bretscher C & Marchini A (2019) H-1 Parvovirus as a Cancer-Killing Agent: Past, Present, and Future. Viruses 11(6). Kasinskas RW & Forbes NS (2006) Salmonella typhimurium specifically chemotax and proliferate in heterogeneous tumor tissue in vitro. Biotechnol Bioeng 94(4):710- 721. Forbes NS (2010) Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer 10(11):785-794. Wang WK, Lu MF, Kuan YD, & Lee CH (2015) The treatment of mouse colorectal cancer by oral delivery tumor-targeting Salmonella. Am J Cancer Res 5(7):2222-2228. Raman V, Van Dessel N, O'Connor OM, & Forbes NS (2019) The motility regulator flhDC drives intracellular accumulation and tumor colonization of Salmonella. J Immunother Cancer 7(1):44. Arrach N, et al. (2010) High-throughput screening for salmonella avirulent mutants that retain targeting of solid tumors. Cancer Res 70(6):2165-2170. Kim JE, et al. (2015) Salmonella typhimurium Suppresses Tumor Growth via the Pro- Inflammatory Cytokine Interleukin-1beta. Theranostics 5(12):1328-1342. Phan TX, et al. (2015) Activation of inflammasome by attenuated Salmonella typhimurium in bacteria-mediated cancer therapy. Microbiology and immunology 59(11):664-675. Kawaguchi K, et al. (2018) Tumor targeting Salmonella typhimurium A1-R in combination with gemcitabine (GEM) regresses partially GEM-resistant pancreatic cancer patient-derived orthotopic xenograft (PDOX) nude mouse models. Cell Cycle 17(16):2019-2026. Hoffman RM (2016) Tumor-Targeting Salmonella typhimurium A1-R: An Overview. Methods Mol Biol 1409:1-8. Igarashi K, et al. (2018) Tumor-targeting Salmonella typhimurium A1-R combined with recombinant methioninase and cisplatinum eradicates an osteosarcoma cisplatinum-resistant lung metastasis in a patient-derived orthotopic xenograft (PDOX) mouse model: decoy, trap and kill chemotherapy moves toward the clinic. Cell Cycle 17(6):801-809. Raman V, et al. (2021) Intracellular delivery of protein drugs with an autonomously lysing bacterial system reduces tumor growth and metastases. Nat Commun 12(1):6116. Cotmore SF & Tattersall P (2014) Parvoviruses: Small Does Not Mean Simple. Annu Rev Virol 1(1):517-537. Cotmore SF & Tattersall P (2005) Genome packaging sense is controlled by the efficiency of the nick site in the right-end replication origin of parvoviruses minute virus of mice and LuIII. J Virol 79(4):2287-2300. Cotmore SF & Tattersall P (2013) Parvovirus diversity and DNA damage responses. Cold Spring Harb Perspect Biol 5(2). Blechacz B & Russell SJ (2004) Parvovirus vectors: use and optimisation in cancer gene therapy. Expert Rev Mol Med 6(16):1-24. Ros C, Bayat N, Wolfisberg R, & Almendral JM (2017) Protoparvovirus Cell Entry. Viruses 9(11). Herrero YCM, et al. (2004) Parvovirus H-1 infection of human glioma cells leads to complete viral replication and efficient cell killing. Int J Cancer 109(1):76-84. Cornelis JJ, et al. (2004) Cancer gene therapy through autonomous parvovirus-- mediated gene transfer. Curr Gene Ther 4(3):249-261. Brandenburger A & Russell S (1996) A novel packaging system for the generation of helper-free oncolytic MVM vector stocks. Gene Ther 3(10):927-931. Majumder K, et al. (2020) The NS1 protein of the parvovirus MVM Aids in the localization of the viral genome to cellular sites of DNA damage. PLoS Pathog 16(10):e1009002. Cotmore SF & Tattersall P (2007) Parvoviral host range and cell entry mechanisms. Adv Virus Res 70:183-232. Op De Beeck A & Caillet-Fauquet P (1997) The NS1 protein of the autonomous parvovirus minute virus of mice blocks cellular DNA replication: a consequence of lesions to the chromatin? J Virol 71(7):5323-5329. Marchini A, Bonifati S, Scott EM, Angelova AL, & Rommelaere J (2015) Oncolytic parvoviruses: from basic virology to clinical applications. Virol J 12:6. Mattola S, et al. (2021) Concepts to Reveal Parvovirus-Nucleus Interactions. Viruses- Basel 13(7). Goradel NH, et al. (2021) Oncolytic virotherapy: Challenges and solutions. Curr Probl Cancer 45(1):100639. Hill C & Carlisle R (2019) Achieving systemic delivery of oncolytic viruses. Expert Opin Drug Deliv 16(6):607-620. Merchlinsky MJ, et al. (1983) Construction of an infectious molecular clone of the autonomous parvovirus minute virus of mice. J Virol 47(1):227-232. Lang SI, et al. (2005) The infectivity and lytic activity of minute virus of mice wild- type and derived vector particles are strikingly different. J Virol 79(1):289-298. Hagan CE & Warren GJ (1983) Viability of palindromic DNA is restored by deletions occurring at low but variable frequency in plasmids of Escherichia coli. Gene 24(2- 3):317-326. Leach DR (1994) Long DNA palindromes, cruciform structures, genetic instability and secondary structure repair. Bioessays 16(12):893-900. Boissy R & Astell CR (1985) An Escherichia coli recBCsbcBrecF host permits the deletion-resistant propagation of plasmid clones containing the 5'-terminal palindrome of minute virus of mice. Gene 35(1-2):179-185. Tattersall SFCaP (1996) Parvovirus DNA Replication. in DNA Replication in Eukaryotic Cells (Cold Spring Harbor Laboratory Press), pp 799-813. Li L, Cotmore SF, & Tattersall P (2013) Parvoviral left-end hairpin ears are essential during infection for establishing a functional intranuclear transcription template and for efficient progeny genome encapsidation. J Virol 87(19):10501-10514. Buljubasic M, et al. (2019) RecBCD- RecFOR-independent pathway of homologous recombination in Escherichia coli. DNA Repair (Amst) 83:102670. Cano DA, Pucciarelli MG, Garcia-del Portillo F, & Casadesus J (2002) Role of the RecBCD recombination pathway in Salmonella virulence. J Bacteriol 184(2):592-595. Rocha EP, Cornet E, & Michel B (2005) Comparative and evolutionary analysis of the bacterial homologous recombination systems. PLoS Genet 1(2):e15. Galitski T & Roth JR (1997) Pathways for homologous recombination between chromosomal direct repeats in Salmonella typhimurium. Genetics 146(3):751-767. Smith GR (1989) Homologous recombination in E. coli: multiple pathways for multiple reasons. Cell 58(5):807-809. Spies M & Kowalczykowski SC (2004) Homologous Recombination by the RecBCD and RecF Pathways (2005 ASM Press, Washington, D.C.). Lloyd RG & Low KB (1996) Homologous recombination (ASM Press, Washington, DC) 2 Ed. Kowalczykowski SC, Dixon DA, Eggleston AK, Lauder SD, & Rehrauer WM (1994) Biochemistry of homologous recombination in Escherichia coli. Microbiol Rev 58(3):401-465. Lee J, et al. (2021) Crystal structure of the nuclease and capping domain of SbcD from Staphylococcus aureus. Journal of microbiology (Seoul, Korea) 59(6):584-589. 63. Zhang X, et al. (2011) Improving Salmonella vector with rec mutation to stabilize the DNA cargoes. BMC Microbiol 11:31. 64. Miesel L & Roth JR (1996) Evidence that SbcB and RecF pathway functions contribute to RecBCD-dependent transductional recombination. Journal of Bacteriology 178(11):3146-3155. 65. Zuniga RA, et al. (2019) Development of a new promoter to avoid the silencing of genes in the production of recombinant antibodies in chinese hamster ovary cells. J Biol Eng 13:59. 66. Cicin-Sain L, Brune W, Bubic I, Jonjic S, & Koszinowski UH (2003) Vaccination of mice with bacteria carrying a cloned herpesvirus genome reconstituted in vivo. J Virol 77(15):8249-8255. 67. Forbes NS, Munn LL, Fukumura D, & Jain RK (2003) Sparse initial entrapment of systemically injected Salmonella typhimurium leads to heterogeneous accumulation within tumors. Cancer Res 63(17):5188-5193. 68. Mosberg JA, Lajoie MJ, & Church GM (2010) Lambda red recombineering in Escherichia coli occurs through a fully single-stranded intermediate. Genetics 186(3):791-799. All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In the event that the definition of a term incorporated by reference conflicts with a term defined herein, this specification shall control.

Claims

WHAT IS CLAIMED IS: 1. An engineered bacterial cell comprising: a) a lysis gene or lysis cassette operably linked to an intracellularly induced Salmonella promoter; b) one or more of the bacterial cell genes selected from the group consisting of recA, recB, recF, sbcB, sbcCD, recJ, sseJ or any combination thereof are knocked out; and c) a nucleic acid sequence coding for an oncolytic virus genome.
2. The bacterial cell of claim 1, wherein the knock out renders the bacterial cell homologous-recombination-deficient (HRDS).
3. The bacterial cell of claim 1, wherein gene sbcCD is knocked out.
4. The bacterial cell of claim 1, wherein genes recB, sbcB, sbcCD and recF are knocked out.
5. An engineered bacterial cell comprising: a) a lysis gene or lysis cassette operably linked to an intracellularly induced Salmonella promoter; and b) bacterial cell genes recB, sbcB, sbcCD and recF knocked out.
6. The bacterial cell of claim 1, wherein the bacterial cell is an intratumoral bacteria cell.
7. The bacterial cell of claim 1, wherein the bacterial cell is a Clostridium, Escherichia coli, Bifidus, Listeria, Yersinia, or Salmonella cell.
8. The bacterial cell of claim 1, wherein the bacterial cell is a Salmonella cell.
9. The bacterial cell of claim 1, wherein the bacterial cell is an attenuated cell.
10. The bacterial cell of claim 9, wherein the bacteria cell has at least a partial deletion of msbB gene.
11. The bacterial cell of claim 10, wherein the bacteria cell has at a partial deletion of purI gene.
12. The bacterial cell of claim 1, wherein viral genome and the lysis gene or lysis cassette operably linked to an intracellularly induced Salmonella promoter are coded for on separate expression vectors.
13. The bacterial cell of claim 1, wherein the lysis cassette is Lysin E (lysE).
14. The bacterial cell of claim 13, wherein lysE is from phage phiX174, the lysis cassette of phage iEPS5, or the lysis cassette from lambda phage.
15. The bacterial cell of claim 1, wherein the intracellularly induced Salmonella promoter is a promoter for one of the genes in Salmonella pathogenicity island 2 type III secretion system (SPI2-T3SS).
16. The bacterial cell of claim 1, wherein the promoter is selected from the group SpiC/SsaB, SseF, SseG, SseI, SseJ, SseK1, SseK2, SifA, SifB, PipB, PipB2, SopD2, GogB, SseL, SteC, SspH1, SspH2, or SirP.
17. The bacterial cell of claim 16, wherein the intracellularly induced Salmonella promoter comprises SseJ.
18. The bacterial cell of claim 1, wherein sseJ is deleted and flhDC circuit is induced by salicylic acid.
19. The bacterial cell of claim 1, wherein the virus comprises a DNA (ssDNA) virus, double-stranded DNA (dsDNA), single-stranded RNA (ssRNA), double-stranded RNA (dsRNA) viruses or a combination thereof.
20. The bacterial cell of claim 1, wherein the virus genome is selected from the group consisting of adenoviruses, herpes simplex virus (HSV), parvoviruses or poxviruses, vaccinia virus, measles virus, picornavirus, reovirus, or paramyxovirus.
21. A composition comprising a population of cells of any of claims 1 to 20 and a pharmaceutically acceptable carrier.
22. A method to colonize a tumor and/or tumor associated cells comprising administering a population of the bacterial cells of claim 1 to a subject in need thereof.
23. The method of claim 22, wherein the tumor associated cells are intratumoral immune cells, vascular endothelial cells, or stromal cells within tumors.
24. A method to treat cancer comprising administering to subject in need thereof an effective amount of a population of the bacterial cells of claim 1 to treat said cancer.
25. A method of inhibiting tumor growth/proliferation or reducing the volume/size of a tumor comprising administering to subject in need thereof an effective amount of a population of the bacterial cells of claim 1 to suppress tumor growth or reduce the volume of the tumor.
26. A method to treat, reduce formation/number or inhibit spread of metastases comprising administering to subject in need thereof an effective amount of a population of the bacterial cells of claim 1 to treat, reduce formation/number or inhibit spread of metastases.
27. The method of any of claims 22 to 26, wherein the tumor, tumor associated cells, cancer, or metastases are a lung, liver, kidney, breast, prostate, pancreatic, skin, colon, head and neck, ovarian, immune (such as lymphoma) and/or gastroenterological tumor, tumor associated cells, cancer or metastases.
28. The bacterial cell of claim 1, wherein the virus genome is from H-1PV.
PCT/US2024/059002 2023-12-08 2024-12-06 Delivery of oncolytic viruses with engineered salmonella Pending WO2025122955A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363607690P 2023-12-08 2023-12-08
US63/607,690 2023-12-08

Publications (1)

Publication Number Publication Date
WO2025122955A1 true WO2025122955A1 (en) 2025-06-12

Family

ID=95980603

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/059002 Pending WO2025122955A1 (en) 2023-12-08 2024-12-06 Delivery of oncolytic viruses with engineered salmonella

Country Status (1)

Country Link
WO (1) WO2025122955A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100178684A1 (en) * 2006-12-21 2010-07-15 Woo Savio L C Transgenic oncolytic viruses and uses thereof
WO2022174226A1 (en) * 2021-02-09 2022-08-18 University Of Massachusetts Intracellular delivery of therapeutic proteins designed to invade and autonomously lyse and methods of use thereof
US20230132250A1 (en) * 2020-03-11 2023-04-27 Nature Technology Corporation Bacterial host strains

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100178684A1 (en) * 2006-12-21 2010-07-15 Woo Savio L C Transgenic oncolytic viruses and uses thereof
US20230132250A1 (en) * 2020-03-11 2023-04-27 Nature Technology Corporation Bacterial host strains
WO2022174226A1 (en) * 2021-02-09 2022-08-18 University Of Massachusetts Intracellular delivery of therapeutic proteins designed to invade and autonomously lyse and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FERNA ´NDEZ SILVIA, SOROKIN ALEXEI, ALONSO JUAN C: "Genetic Recombination in Bacillus subtilis 168: Effects of recU and recS Mutations on DNA Repair and Homologous Recombination", JOURNAL OF BACTERIOLOGY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 180, no. 13, 1 July 1998 (1998-07-01), US , pages 3405 - 3409, XP093327835, ISSN: 0021-9193 *
MARCHINI ANTONIO, DAEFFLER LAURENT, POZDEEV VITALY I., ANGELOVA ASSIA, ROMMELAERE JEAN: "Immune Conversion of Tumor Microenvironment by Oncolytic Viruses: The Protoparvovirus H-1PV Case Study", FRONTIERS IN IMMUNOLOGY, vol. 10, 1 August 2019 (2019-08-01), pages 1848, XP055803151, DOI: 10.3389/fimmu.2019.01848 *

Similar Documents

Publication Publication Date Title
Vassaux et al. Bacterial gene therapy strategies
Van Pijkeren et al. A novel Listeria monocytogenes-based DNA delivery system for cancer gene therapy
US20090208534A1 (en) Attenuated salmonella as a delivery system for sirna-based tumor therapy
JP2024026075A (en) Modified vaccinia vector
JP2019530467A (en) Self-limiting Cas9 network (SLiCES) plasmid and its lentiviral system for improved safety
JP6908593B2 (en) Delivery vehicle
CN1902317B (en) Plasmid maintenance
JP5841432B2 (en) Plasmid for transformation
CN106795527A (en) Oncolytic virus
JP7539723B2 (en) Compositions and methods for organ-protecting expression and regulation of encoding ribonucleic acids
Nafissi et al. Construction and characterization of an in-vivo linear covalently closed DNA vector production system
KR20100075884A (en) Methods to increase transgene expression from bacterial based delivery systems by co-expressing suppressors of the eukaryotic type i interferon response
JP7774263B2 (en) Bacterial cells designed to invade and autonomously lyse for intracellular delivery of therapeutic proteins
WO2021074379A1 (en) Bacterial delivery of viruses into eukaryotic cells
Fu et al. Synergistic antitumoral effects of human telomerase reverse transcriptase‐mediated dual‐apoptosis‐related gene vector delivered by orally attenuated Salmonella enterica Serovar Typhimurium in murine tumor models
KR102683896B1 (en) Expression Vector for Secreting GM-CSF and Anticancer Recombinant Microbials Transformed thereby
US20220387575A1 (en) Construction method of recombinant drug-resistant mycobacterium bovis bacillus calmette-guerin (bcg) strain and pharmaceutical composition for treating tuberculosis (tb)
WO2025122955A1 (en) Delivery of oncolytic viruses with engineered salmonella
WO2018012570A1 (en) Method for producing vaccinia virus expressing foreign gene
EA011282B1 (en) Bacterial packaging strains useful for generation and production of recombinant double-stranded rna nucleocapsids and uses thereof
WO2022151078A1 (en) Oncolytic virus and application thereof
WO2004046319A2 (en) Bacteriophage having modified holin and uses thereof
US20250000913A1 (en) Salmonella engineered for nontoxic colonization of tumors
US20250195631A1 (en) Antigen delivering salmonella for use as a tumor homing beacon to refocus preexisting, vaccine generated t cells to combat cancer
WO2021186042A1 (en) Plasmid comprising alopecia antigen

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: 24901666

Country of ref document: EP

Kind code of ref document: A1