[go: up one dir, main page]

WO2023235633A2 - Improved salmonella vectored therapies for treatment of cancer - Google Patents

Improved salmonella vectored therapies for treatment of cancer Download PDF

Info

Publication number
WO2023235633A2
WO2023235633A2 PCT/US2023/024484 US2023024484W WO2023235633A2 WO 2023235633 A2 WO2023235633 A2 WO 2023235633A2 US 2023024484 W US2023024484 W US 2023024484W WO 2023235633 A2 WO2023235633 A2 WO 2023235633A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
cancer
plasmid
seq
gmsc
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.)
Ceased
Application number
PCT/US2023/024484
Other languages
French (fr)
Other versions
WO2023235633A3 (en
Inventor
Roy Curtiss
Shifeng Wang
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 Florida
University of Florida Research Foundation Inc
Original Assignee
University of Florida
University of Florida Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Florida, University of Florida Research Foundation Inc filed Critical University of Florida
Priority to EP23816835.5A priority Critical patent/EP4531880A2/en
Publication of WO2023235633A2 publication Critical patent/WO2023235633A2/en
Publication of WO2023235633A3 publication Critical patent/WO2023235633A3/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Cancer represents a diversity of disease states characterized by unregulated proliferation of cells that are either freely multiplying in blood and/or lymph or organized into tumor masses. After cardiovascular disease, cancer ranks as the second most common cause of death in the US (1 ).
  • Salmonella Typhimurium While obligate anaerobes, such as Bifidobacterium and Clostridium, are highly effective at accumulating and replicating in necrotic tumors, they do not grow in viable tumor tissues, which limits their efficacy as anti-cancer agents. S. Typhimurium is a facultative anaerobe, which can survive and grow in anoxic regions as well as in viable oxygenic regions of tumors. Salmonella also has ability to identify and penetrate tumors by detecting small molecules such as serine and aspartate in tumors, and accumulates in tumors that contain free amino acids, purines and pyrimidines that facilitate Salmonella growth.
  • S. Typhimurium is widely investigated as an anti-cancer agent (see (5)).
  • S. Typhimurium VNP20009 or its derivatives are widely investigated as the anti-cancer agent or as a vector to investigate efficacy of anti-cancer activities (see (5, 9)).
  • VNP20009 carrying a purine auxotrophic mutation (pur/) and lipid A mutation (msbB) and its derivatives demonstrated good anti-tumor efficacy in mice
  • anti-cancer efficacy in human trials was not achieved in phase I clinical trials in patients with metastatic melanoma and renal carcinoma (10, 11).
  • VNP20009-immunized dogs with a variety of malignant tumors bacterial colonization of tumors was observed but only 4 of 35 dogs tested were completely cured (12). Even intratumoral injection in humans with cancer only led to colonization in 2 out of 3 patients (13).
  • the reasons for failure in human clinical trials may be that the parent of VNP20009 is not highly virulent and its genetic construction is not precise.
  • VNP20009 is derived from ATCC 14028, which does not show high virulence and invasiveness compared to other S. Typhimurium strains (14) and we demonstrated that an attenuated aroA mutant of 14028 was not as immunogenic and did not induce as high protective immune levels as did an isogenic derivative of the S. Typhimurium UK-1 strain (15) and furthermore was not as effective as a UK-1-derived strain in ablating colorectal tumors in mice (16).
  • construction of VPN20009 is based on UV- and Tn10 transposon-induced mutations, which may result in other mutations and over-attenuation (17). It has been shown that the design method causes strain VPN20009 lost chemotactic ability (18).
  • the msbB mutation in VNP20009 is a bad choice because it leads to production of penta- acylated lipid A, which is a good pro-inflammatory stimulator in mice, but is an antagonist to inhibit stimulating human innate immunity (19-22).
  • the second S. Typhimurium strain widely used for cancer therapy is A1-R, which is also derived from ATCC 14028 and is a leu-arg auxotroph (23, 24). Notable, the parent of A1-R (25), A1 is screened through nitrosoguanidine mutagenesis (24). A1-R exhibited good tumor- seeking features and has antitumor efficacy against major types of cancer in mice (24- 26), but no clinical trials in humans or dogs have been performed.
  • the 3 rd strain is VXM01, which is based on the S. Typhi strain Ty21a vaccine carrying an eukaryotic expression plasmid for VEGFR2, could induce anti-angiogenic activity when delivered by the oral route in pancreatic cancer. But only 1 of 13 patients showed an improved clinical outcome (27).
  • the 4 th strain tested was ⁇ 4550 delivering IL-2 to induce responses in dogs and humans, respectively (28-30). All the these strains lack specific tumor targeting ability although VPN20009 and A1-R preferentially colonize tumors. Nevertheless, the results showed their targeting ability is not enough for high efficacy.
  • Salmonella has ability to regress tumors because of its natural toxicity and can also be used as vectors to deliver other anti-cancer molecules including cytotoxic agents such as Cytolysin A (ClyA), FAS ligand (FasL) and TNF-related apoptosis- inducing ligand (TRAIL), cytokines such as IL-2, and tumor antigens such as 3urviving and other factors such as tyrosinase which enhance its anti-cancer effectiveness (see (5, 9). FasL and TRAIL are belonging to the TNF ⁇ family.
  • ClyA Cytolysin A
  • FasL FAS ligand
  • TRAIL TNF-related apoptosis- inducing ligand
  • IL-2 tumor antigens
  • tumor antigens such as 3urviving and other factors such as tyrosinase which enhance its anti-cancer effectiveness
  • FasL specifically induces apoptosis in cells that possess the FAS receptor and TRAIL is cytotoxic to many cancer cells via death receptor pathways, which activate caspases 8 and 3 (31, 32).
  • ClyA is a bacterial toxin inducing apoptosis and when delivered by S. Typhimurium reduced tumor growth in mice (33, 34).
  • Cytokine IL-2 is widely investigated for its anti-cancer ability because IL-2 can activate the cytolytic function of NK cells and promotes lymphocyte proliferation (35-37). Cytotoxic agents and cytokines can induce apoptosis or stimulate immune cells to directly kill cancer cells, while tumor antigens such as 3urviving function to sensitize the immune system to fight against cancer cells.
  • Survivin is a member of the inhibitor-of-apoptosis protein family involved in regulation of apoptosis and T-cell responses in anti-tumor immunity. It is over-expressed in many tumor cells. Blocking 3urviving function is thus a promising anti-tumor therapeutic method via induction of immune responses against (38-41).
  • a patent application was filed on use of attenuated derivatives of pathogenic bacteria to deliver recombinant protective antigens from heterologous pathogens to induce protective immunity to the pathogens whose antigens were delivered by the vaccine construct.
  • Salmonella was the chosen pathogen and it has been continuously improved and perfected as a means for using Salmonella as an antigen and DNA vaccine delivery vector (42, 43).
  • the protein is encoded by codon-optimized sequences to enhance mRNA stability and efficiency of transcription and translation in Salmonella (58-60). Since immune responses against recombinant proteins are improved by secretion of antigens rather than their retention in the RASV cytosol (61, 62), we perfected use of type 3 and type 2 secretion systems (T3SS & T2SS) (52, 55, 63) to export proteins out of the RASV or into the periplasmic space to enhance production of outer membrane vesicles that are highly immunogenic (49, 64).
  • T3SS & T2SS type 3 and type 2 secretion systems
  • higher levels of induced protective immunity can be induced by vaccine strains displaying the regulated delayed lysis phenotype than by strains not undergoing lysis (45, 52, 65-68).
  • Figure 3. Plasmid map of pG8R314 encoding OmpA with PLZ4 insert (ompA ⁇ plz4).
  • Figure 9. Plasmid maps of pG8R341 derived from pG8R314 by insertion of sequence encoding GFP3 as an operon fusion and pG8R342 derived from pG8R320 by insertion of sequence encoding EGFP.
  • Plasmid maps of pG8R347 , pG8R348, pG8R349 and pG8R350 are derived from pG8R320 by insertion of sequences for 5’ HLA, HLA leading and Tail peptides and 3’ HLA.
  • pG8R348, pG8R349 and pG8R350 are derived from pG8R347 by insertion of sequence encoding EGFP, neo-antigen BBM963 and MB49, respectively.
  • Figure 13 Plasmid pG8R361 derived from pG8R320 by insertion of sequence from PEF1 ⁇ promoter.
  • Plasmid maps of pG8R362, pG8R363, pG8R364 and pG8R365 Plasmid maps of pG8R362, pG8R363, pG8R364 and pG8R365.
  • pG8R362 and pG8R363 are derived from pG8R320 by insertion of sequence encoding HAC-PD1 fused with human CXCL11 and HAC-PD1 fused with mouse CXCL11, respectively.
  • pG8R364 and pG8R365 are derived from pG8R361 by insertion of sequence encoding HAC-PD1 fused with human CXCL11, HAC-PD1 fused with mouse CXCL11, respectively.
  • Figure 15 Figure 15
  • Figure 16 Plasmid maps of pG8R372, pG8R373, pG8R374 and pG8R375 derived from pG8R320 by insertion of sequence encoding IL2 SS fused with HAC-PD1.
  • Figure 17. Plasmid maps of pG8R380 derived from pG8R320 by insertion of sequence encoding operon fusion of OmpA with LHRH insertion and GFP and pG8R381 derived from pG8R380 by insertion of OmpA with LHRH insertion, respectively.
  • Figure 19 Plasmid maps of pG8R385 derived from pG8R320 by insertion of sequence encoding operon fusion of OmpA with Her2 scFv insertion and GFP and pG8R386 derived from pG8R385 by insertion of OmpA with Her2 scFv insertion, respectively.
  • Figure 20 Plasmid maps of pG8R382, pG8R383 and pG8R384 derived from pG8R320 by insertion of sequence encoding haPD1-IgG, haPD1-IgG and
  • Figure 21 Plasmid maps of pG8R390, pG8R391, and pG8R418.
  • pG8R390 is derived from pG8R381 by insertion of sequence encoding KillerRed fused with neuromodulin N terminal sequence
  • pG8R391 is derived from pG8R386 by insertion of sequence encoding KillerRed fused with neuromodulin N terminal sequence.
  • pG8R418 is derived from pG8R385 by insertion of sequence encoding KillerRed fused with neuromodulin N terminal sequence.
  • Figure 22 The attachment and invasion of bladder cancer cells with strains derived from ⁇ 12614 with O-antigen mutations.
  • A Genotypic characterization of strains using primers specific for waaL, waaG, waaC, ompA and ompA ⁇ plz4. Lane 1, ⁇ 3761; lane 2, ⁇ 12614; lane 3, ⁇ 12812; lane 4, ⁇ 12813; lane 5, ⁇ 12814.
  • B LPS gel profile of strains. All strains are grown in LB media.
  • KillerRed kills HEK293T cell. Time zero is set immediately after irradiation with green light. Time 10 is set immediately after excitation for 10 min (EVOS FL, RFP channel) Figure 28. KillerRed kills HEK293T cell excitation for 10 and 20 minutes. Time zero is set immediately after irradiation with green light.
  • Agents described herein may be administered by oral, intradermal, intravenous, intramuscular, intraocular, intranasal, intrapulmonary, epidermal, subcutaneous, mucosal, or transcutaneous administration.
  • animal host or “subject” as used interchangeably hereinto refer to a human or nonhuman mammal or a vertebrate animal into which a genetically modified Salmonella cell has been administered. In a specific embodiment, the subject is a human.
  • the terms “attenuated” or “attenuation” as used herein refer to the process of rendering certain pathogen virulence attributes needed to cause diseases less able to cause such disease symptoms. In one example, attenuation involves imparting an attenuation mutation in the pathogen.
  • attenuating mutation refers to a mutation imparted into a pathogen that reduces infectivity, virulence, toxicity, induction of disease symptoms, and/or impairment of a subject upon administration of the pathogen (e.g. PIESV strain).
  • pathogen e.g. PIESV strain
  • attenuating mutations include those mutations that facilitate lysis in vivo (e.g. impairing synthesis of essential constituents of peptidoglycan layer), reduce or impair synthesis of LPS or other cell-surface components, and one or more mutations that provide auxotrophy (e.g. dependence on an amino acid, purine, pyrimidine, or vitamin for growth).
  • balanced-lethal vector-host refers to a host Salmonella cell into which a plasmid vector has been introduced such that survival of the host cell is dependent on the maintenance of the plasmid vector and loss of the plasmid vector results in death of the host Salmonella cell.
  • ASD + Expression-Cloning Vector Stable Maintenance and High Level Expression of Cloned Genes in a Salmonella Vaccine Strain. Nat Biotechnol 6, 693–697 (1988) or Galán JE, Nakayama K, Curtiss R 3 rd .
  • biologically active fragment or “biologically active variant” refers to a fragment or variant of a sequence that maintains its biological activity.
  • a biologically active fragment or biologically active variant is a fragment or variant of an antigen amino acid sequence that elicits an immune response in a host.
  • CCTS strain refers to a strain of Salmonella that has one or more attenuating mutations and expresses a gene product that causes selective localization and/or internalization of cells of the CCTS strain by a cancer cell.
  • “codon” means, interchangeably, (i) a triplet of ribonucleotides in an mRNA which is translated into an amino acid in a polypeptide or a code for initiation or termination of translation, or (ii) a triplet of deoxyribonucleotides in a gene whose complementary triplet is transcribed into a triplet of ribonucleotides in an mRNA which, in turn, is translated into an amino acid in a polypeptide or a code for initiation or termination of translation.
  • 5’-TCC-3’ and 5’-UCC-3’ are both “codons” for serine, as the term “codon” is used herein.
  • codon optimized refers to enhancing the ability of the antigen encoding sequence to be expressed in the Salmonella strain by selecting codons that are used for highly expressed genes in Salmonella. Such codon optimization also includes changing the GC content of the antigen encoding sequence to be similar to that used for Salmonella (i.e., ⁇ 52% GC). In addition, the codon optimization can also be used to enhance the stability of the mRNA encoded by the antigen encoding sequence so as to be less likely to be degraded by Rnases.
  • the term “delayed attenuation” as used herein refers to a means of gene regulation such that the attenuation attribute is not expressed during growth of the vaccine strain or during its administration to an animal host but is not expressed after the CCTS strain enters the animal host and is manifest as a consequence of vaccine cell division in vivo with gradual dilution of the virulence gene product by at least half at each cell division in vivo.
  • the term “gene product” refers to a transcript (RNA) or expressed polypeptide encoded by a heterologous gene or nucleic acid that has been introduced into a genetically modified Salmonella cell. In typical embodiments, the gene product causes selected localization to a target cell.
  • the gene product may also cause cytotoxicity to the target cell upon internalization of the genetically modified Salmonella cell and/or cause a targeted immune response to target cells.
  • a "genetically modified Salmonella cell” or “GMSC” refers to a Salmonella cell that comprises an attenuating mutation and/or into which a heterologous gene or nucleic acid, e.g., an exogenous nucleic acid that is foreign to the Salmonella cell, has been introduced.
  • GMSC genetically modified Salmonella cell
  • the term “operably linked” as used herein means that one nucleic acid sequence is linked to another nucleic acid sequence, and therefore the function or expression thereof is influenced by the linked nucleic acid sequence.
  • percentage of sequence identity may refer to the value determined by comparing two optimally aligned sequences (e.g., nucleic acid sequences or polypeptide sequences) of a molecule over a comparison window, wherein the portion of the sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleotide or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the comparison window, and multiplying the result by 100 to yield the percentage of sequence identity.
  • a sequence that is identical at every position in comparison to a reference sequence is said to be 100% identical to the reference sequence, and vice-versa.
  • the term “about” with respect to a numerical value of a sequence length means the stated value with a +/- variance of up to 1-5 percent. For example, about 30 contiguous nucleotides means a range of 27-33 contiguous nucleotides, or any range in between.
  • sequence identity means the stated percentage value with a +/- variance of up to 1-3 percent rounded to the nearest integer. For example, about 90% sequence identity means a range of 87-93%. However, the percentage of sequence identity cannot exceed 100 percent. Thus, about 98% sequence identity means a range of 95-100%.
  • regulated delayed lysis refers to a construction in which the expression of one or more genes specifying synthesis of peptidoglycan precursors such as but not limited to diaminopimelic acid and muramic acid are regulated by a sugar-dependent process such that the genes are expressed in the presence of a sugar such as but not limited to arabinose or rhamnose supplied during cultivation of the strain and cease to be expressed in vivo since the sugar is absent to result in lysis as a consequence of cell division of the CCTS strain in vivo.
  • the genes conferring the regulated delayed lysis phenotype may be either chromosomal and/or plasmid encoded.
  • regulated delayed lysis plasmid refers to a construction in which the expression of one or more genes specifying synthesis of peptidoglycan precursors such as but not limited to diaminopimelic acid and muramic acid that are regulated by a sugar-dependent process are located on a plasmid vector encoding synthesis of one or more foreign antigens or gene products.
  • the term “Salmonella cell” refers to a cell of a Salmonella species or serotype. Examples of a Salmonella serotype include Salmonella Typhimurium and Salmonella Enteritidis. In a more specific embodiment, the Salmonella serotype is S. Typhimurium UK-1.
  • sequence identity refers to the residues in the sequences of the two molecules that are the same when aligned for maximum correspondence over a specified comparison window.
  • target immune response refers to a response by a subject’s immune system against target cells. Immune responses include both cell- mediated immune responses (responses mediated by antigen-specific T cells and non-specific cells of the immune system) and humoral immune responses (responses mediated by antibodies present in the plasma lymph, and tissue fluids and secreted onto mucosal surfaces).
  • target cell refers to a cell of a subject that is of a type to which a genetically modified Salmonella cell is designed for selective localization and/or internalization.
  • Selective localization refers to increased migration of the genetically modified Salmonella cell to a target cell over other cells in a subject.
  • Selective internalization refers to increased internalization of the genetically modified Salmonella cell in the target cell over other cells in the subject.
  • Increased localization to and/or increased internalization means an increase of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70% or more respective to target cells as opposed to other cells in a subject.
  • a target cell internalizes the genetically modified Salmonella cell by active invasion or endocytosis or phagocytosis.
  • the target cell is a cancer cell and the genetically modified Salmonella cell is of a CCTS strain that is selectively internalized by the cancer cell over other cells in the subject.
  • the genetically modified Salmonella cell is engineered to localize at a tumor microenvironment where cancer cells are present without necessarily being internalized into a cancer cell.
  • variant refers to a nucleic acid sequence or amino acid sequence that possesses at least about 85, 90, 95, 96, 97, 98 or 99 percent sequence identity to another nucleic acid sequence or amino acid sequence, respectively. Other relevant definitions are provided infra.
  • CCTS strain embodiments have (i) ability to directly destroy tumor cells, (ii) deliver cargoes that cause tumor cells to self-destruct, (iii) deliver cargoes that enhance abilities to treat tumor cells, and/or (iv) directly and/or indirectly stimulate host immune responses to repress tumor cell growth, metastases and cell death.
  • a potentially desirable feature involves rapid self-destruction of CCTS cells that enables their use for repeat treatments of subjects.
  • a unique attribute of these newly designed and constructed CCTS strains is their ability to simultaneously synthesize and deliver protein cargoes to cancer cells but to also deliver DNA vaccines encoding other effective proteins to be synthesized by the tumor cells to their detriment.
  • the foregoing attributes are achieved by introducing numerous deletion and deletion-insertion mutations to enable and endow the desired phenotypic properties to the strains constructed. These mutations and their associated phenotypes are listed in Table 1 and the suicide vectors needed for their insertion into plasmids and the S. Typhimurium chromosome are listed in Table 2. The distribution of genetic deletion and deletion-insertion mutations and the redundancy in critical modifications ensure both stability and safety of these CCTS strains.
  • genotypes of CCTS strains are listed in Table 3. Table 1. Mutations and associated phenotypes in S. Typhimurium CCTS strains a It is noted that the genes can be inactivated or deleted in multiple ways to confer the same phenotypic traits. Also, though certain allele numbers are indicated elsewhere herein for certain mutations, reference to a certain allele is not limiting and the mutations can be executed in other alleles.
  • Genotype Phenotype ⁇ aroA encodes the first enzyme in the pathway to synthesize aromatic amino acids and derived vitamins (74) ⁇ asdA deletes gene for aspartate semialdehyde dehydrogenase essential for synthesis of diaminopimelic acid (DAP) necessary for peptidoglycan synthesis (75) ⁇ PasdA::TT araC P araBAD asdA makes synthesis of AsdA dependent on presence of arabinose ⁇ PasdA::TT rhaRS PrhaBAD asdA makes synthesis of AsdA dependent on presence of rhamnose ⁇ asdA::TT araC P araBAD c2 inactivates asdA and makes synthesis of C2 repressor dependent on arabinose (76, 77) ⁇ alr and ⁇ dadB deletes the genes for two alanine racemases essential for synthesis of D-alanine necessary for peptidoglycan synthesis (78) ⁇ P
  • Cancers and tumor types that may be treated or attenuated using the methods described herein include but are not limited to bone cancer, bladder cancer, brain cancer, breast cancer, cancer of the urinary tract, carcinoma, cervical cancer, colon cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, liver cancer, lung cancer, lymphoma and leukemia, melanoma, ovarian cancer, pancreatic cancer, pituitary cancer, prostate cancer, rectal cancer, renal cancer, sarcoma, testicular cancer, thyroid cancer, and uterine cancer.
  • a method for treating cancer may include administering a therapeutically effective amount of genetically modified Salmonella cells described herein or therapeutic compositions comprising the same to a subject who has cancer.
  • Treating” or “treatment” of a condition may refer to preventing the condition, slowing the onset or rate of development of the condition, reducing the risk of developing the condition, preventing or delaying the development of symptoms associated with the condition, reducing or ending symptoms associated with the condition, generating a complete or partial regression of the condition, or some combination thereof.
  • a “therapeutically effective amount,” “effective amount” or “effective dose” is an amount of a composition (e.g., a therapeutic composition or cells) that produces a desired therapeutic effect in a subject, such as preventing or treating a target condition or alleviating symptoms associated with the condition.
  • the precise therapeutically effective amount is an amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given subject.
  • This amount will vary depending upon a variety of factors, including but not limited to the characteristics of the therapeutic compound (including activity, pharmacokinetics, pharmacodynamics, and bioavailability), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), the nature of the pharmaceutically acceptable carrier or carriers in the formulation, and the route of administration.
  • One skilled in the clinical and pharmacological arts will be able to determine a therapeutically effective amount through routine experimentation, namely by monitoring a subject's response to administration of a compound and adjusting the dosage accordingly. For additional guidance, see Remington: The Science and Practice of Pharmacy 21 st Edition, Univ.
  • a “route of administration” may refer to any administration pathway known in the art, including but not limited to aerosol, enteral, nasal, ophthalmic, oral, parenteral, rectal, transdermal (e.g., topical cream or ointment, patch), or vaginal.
  • Parenter refers to a route of administration that is generally associated with injection, including infraorbital, infusion, intraarterial, intracapsular, intracardiac, intradermal, intramuscular, intraperitoneal, intrapulmonary, intraspinal, intrasternal, intrathecal, intrauterine, intravenous, subarachnoid, subcapsular, subcutaneous, transmucosal, or transtracheal.
  • the tumor antigen vaccines described herein are administered orally and the compositions that disrupt tumor-derived immune suppression described herein (e.g., YS1646-shSTAT3 Y51646-shIDO1, YS1646-shArg1 or YS1646-shiNOS) are administered intravenously.
  • the compositions that disrupt tumor-derived immune suppression described herein e.g., YS1646-shSTAT3 Y51646-shIDO1, YS1646-shArg1 or YS1646-shiNOS
  • a Bacterial strains, media and bacterial growth. All CCTS strains are derived from the highly virulent S. Typhimurium UK-1 strain (132) since attenuated S. Typhimurium UK-1 strains will induce protective immunity to challenge with all S.
  • LB broth and agar (135) and Purple broth (PB) are used as complex media for propagation, phenotypic analyses and plating.
  • MacConkey agar with 0.5% lactose (Lac) and 0.1% Ara, 0.1% rhamnose and 0.1% mannose (if needed) are used to enumerate bacteria recovered from mice or other animals.
  • Bacterial growth is monitored spectrophotometrically and by plating for colony counts.
  • b. Molecular and genetic procedures. Methods for DNA isolation, restriction enzyme digestion, DNA cloning and use of PCR for construction and verification of bacterial strains and vectors are standard (136). DNA sequence analyses are performed commercially. All oligonucleotide and/or gene syntheses are done commercially with codon optimization to enhance translational efficiency in humans or Salmonella and stabilize mRNA to “destroy” RNase E cleavage sites (59, 60) to prolong mRNA half-life. Plasmids are evaluated by DNA sequencing and ability to specify synthesis of proteins using gel electrophoresis and western blot analyses.
  • DNA vaccine vectors Expression of sequences encoded in DNA vaccine vectors is monitored after electroporation into Vero cells and using antibodies specific to DNA vaccine encoded proteins. Methods for generating mutant strains are described in previous publications (137-145) and in Examples below using the suicide vector delivery strain ⁇ 7213 (thi-1 thr-1 leuB6 glnV44 fhuA21 lacY1 recA1 RP4-2-Tc::Mu ⁇ pir ⁇ asdA4 ⁇ zhf-2::Tn10). Recombinant plasmid constructs are transformed into E.
  • CCTS strain characterization CCTS constructs are evaluated in comparison with vector-control strains for stability of plasmid maintenance, integrity and protein synthesis ability when CCTSs are grown in the presence of arabinose and DAP and with and without IPTG for 50 generations. The IPTG dependence of protein synthesis to overcome the LacI repression of the Ptrc promoter is also verified. IPTG- induced cultures are incubated with chloramphenicol to arrest protein synthesis to determine whether plasmid-specified proteins are stable during the next 4 h. If not, the nucleotide sequence is altered to eliminate protease cleavage sites (with subsequent comparison of both constructs for induction of immune responses).
  • CCTS constructs are evaluated for bile sensitivity, acid tolerance and ability to survive in sera with and without complement (143-145) and for sensitivity to antibiotics used to treat Salmonella infections.
  • Cell culture methods Some tumors are caused by cancer cells with specific targetable receptors or that possess phenotypic properties that can be used to attract specially designed CCTS strains with specific targeting attributes.
  • bladder tumor cells uniquely display a receptor that can bind to a targeting peptide termed PLZ4 (amino acid sequence: CQDGRMGFC) that is absent on normal uroepithelial cells and other cell types throughout the body.
  • PLZ4 amino acid sequence: CQDGRMGFC
  • Nanoparticles coated with PLZ4 specifically target bladder tumor cells but not to other cancer cell types (147-150) . This targeting is observed for bladder tumor cells from mice, dogs and humans (151).
  • CCTS strains displaying PLZ4 can be evaluated by their differential ability to attach to and invade the bladder tumor cell lines 5637, TCCSUP, and T24 (151) . Methods for evaluating the abilities of Salmonella cells to attach to, invade into and survive in cells in culture are well established (152). These methods can be modified as needed for CCTS strains targeting other tumor cell types.
  • Cell imaging Some plasmids have genes encoding fluorescent proteins enabling synthesis of GFP in Salmonella or EGFP or mCherry in animal cells.
  • the fluorescent protein in bacteria or cells will be visualized using the EVOS Automated Cell Imaging System (ThermoFisher Scientific).
  • the Cell Plasma Membrane Staining Kit - Orange Fluorescence - Cytopainter (ab219941, Abcam) was used to label cell membranes.
  • the acquired image was processed using ImageJ software (153).
  • Example 2 Construction of mutant S. Typhimurium strains with deletions of the ompA gene to enable display of altered OmpA proteins with inserted peptides enabling targeting to specific tumor cells.
  • Pan and associates have defined a nine amino acid peptide CQDGRMGFC termed PLZ4 (US patent 10,335,365) that targets a specific receptor present on bladder tumor cells (151).
  • PLZ4 US patent 10,335,365
  • Typhimurium strains with anti-tumor attributes have been constructed to display PLZ4 to preferentially and specifically target bladder tumor cells.
  • the objective was to insert the sequence for PLZ4 into one of the exposed outer loops of the OmpA protein.
  • the OmpA protein was selected since it is the most abundant OMP in the Salmonella outer membrane (105) and could be specified on a plasmid replicon to increase its relative quantity in relation to other OMPs.
  • To construct a strain to test the validity and feasibility of our approach we generated a derivative of ⁇ 12341 to insert the ⁇ ompA11 deletion mutation using the suicide vector pYA4757 (Table 2) to yield the strain ⁇ 12417 (Table 4).
  • the ⁇ ompA11 mutation deletes the entire ompA open reading frame including the start to stop codon sequence.
  • ⁇ 12341 (73, 154) was selected since its viability and virulence are dependent on the supply of three sugars that can be supplied during culture but that are totally absent in animal tissues and since it cannot synthesize LPS O-antigen in vivo thus exposing the outer membrane proteins to enable better and more efficient interactions with eukaryotic cell surfaces in the in vivo environment.
  • S. Typhimurium strains with a diversity of properties for use with a diversity on new plasmid vectors encoding for synthesis of attributes that contribute to tumor therapy, tumor cell destruction and/or to recruit host immunity to target tumor antigens, etc., in addition to tumor cell adherence. All these S. Typhimurium strains listed in Table 4 were constructed using the suicide vectors listed in Table 2 to introduce the mutations described in Table 1. Many were derived from Protective Immunity Enhanced Salmonella Vaccine (PIESV) strains of Self-Destructing Attenuated Adjuvant Salmonella (SDAAS) strains that have been described (PCT/US21/61814 and WO 2021/222696 A1, respectively), which are incorporated herein in their entirety. Table 4. S.
  • PIESV Protective Immunity Enhanced Salmonella Vaccine
  • SDAAS Self-Destructing Attenuated Adjuvant Salmonella
  • strains constructed using the suicide vector pG8R315 are listed in Table 5.
  • One of the examples is ⁇ 12619.
  • the mutations ⁇ waaL46, ⁇ waaG42 and ⁇ waaC41 were introduced into strain ⁇ 12619 to generate a family of strains differing in the presence of the LPS O-antigen, LPS O-antigen and outer LPS core and O-antigen and outer and inner LPS core, respectively.
  • strain ⁇ 12614 with the ⁇ asdA33 ⁇ ompA11 deletion mutations This strain can be transformed with a plasmid encoding ompA ⁇ plz4 to compare the effects of surface modification in Salmonella that affect the targeting ability of Salmonella.
  • the plasmids could be pG8R341, or any plasmid carrying ompA ⁇ plz4 fusion, or other ompA fused with varied targeting peptide sequences.
  • the mutations ⁇ waaL46, ⁇ waaG42 and ⁇ waaC41 were introduced into ⁇ 12614 to generate a series of strains analogous to those generated in ⁇ 12619 resulting in defects in O-antigen, outer core and inner core, respectively. These strains are also listed in Table 5. Table 5. S.
  • Figures 1 and 2 depict the plasmids used as parents or for component segments of derived and constructed plasmids.
  • the derived and constructed plasmids are listed and described in Table 6 with their use and evaluation described in subsequent Examples.
  • Table 7 lists all the nucleotide primers used to construct the plasmids listed in Table 6.
  • a unique and original feature of many of the plasmids designed and constructed is the ability to encode proteins that are synthesized by the CCTS strain to be displayed during targeting and attaching to, invading into and acting within tumor cells in vivo prior to display of regulated lysis within the tumor cell to release the plasmid now serving as a DNA vaccine with unique features to be directed to the nucleus for transcription of encoded sequences that yield products after mRNA translation that exhibit anti-tumor activities.
  • These products with their features as described in later Examples might kill the tumor cell, cause the tumor cell to kill itself (i.e., commit suicide) and/or attract host immune responses that inhibit tumor cell growth and metastases.
  • the pG8R314 plasmid ( Figure 3) with the ompA ⁇ plz4 fusion was constructed by amplifying a 454 bp fragment of the S. Typhimurium UK-1 ( ⁇ 3761) chromosome using primers OmpA-s and OmpA-PLZ4-a and a 707 bp fragment with primers OmpA-PLZ4-s and OmpA-SacIHindIII-a. These two fragments were cloned into plasmid pYA3342 ( Figure 1A) cut with NcoI/HindIII to generate plasmid pG8R314.
  • the sequence encoding the PLZ4 peptide was introduced by both primers OmpA-PLZ4-a and OmpA- PLZ4-s.
  • This plasmid has a gene encoding the PLZ4 peptide inserted into the third exposed loop of the OmpA protein enabling expression of the ompA ⁇ plz4 insertion mutated gene in Salmonella.
  • the PLZ4 peptide was flanked with 2 cysteines forming a disulfide linkage to facilitate its exposure on the loop 3.
  • the suicide vector pG8R315 Figure 4
  • the plasmid pG8R314 was used as the template to generate a 1.1 kb fragment encoding synthesis of OmpA ⁇ PLZ4.
  • This fragment was amplified with primers OmpA-XbaI-s and OmpA- SacIHindIII-a and cut with XbaI/SacI. The fragment was then inserted into suicide plasmid pRE112 ( Figure 1B) cut with XbaI/SacI to generate plasmid pG8R315. This suicide vector is then used to introduce the ompA ⁇ plz4 mutation into the chromosome of the S. Typhimurium strains listed in Table 5. For the construction of pG8R319 ( Figure 5A), we fused two DNA fragments.
  • plasmid pYA4545 ( Figure 1C) as a template
  • a 1,549 bp fragment containing rrfG TT-PCMV-SV40 polyA Trp TT was amplified with primers rrfGTT-s and trpTT-a.
  • plasmid pG8R314 ( Figure 3) as a template
  • a 4,085 bp fragment that includes the whole pG8R314 plasmid was amplified with primers trpTT-s and rrfGTT-a. The two fragments were assembled to generate plasmid pG8R319.
  • the balanced-lethal plasmid has a pBR ori and could express ompA ⁇ plz4 in Salmonella and has a P cmv promoter to be used for gene expression in eukaryotic cells.
  • the P cmv and Ptrc ompA ⁇ plz4 are separated by the trpA TT.
  • plasmid pYA4545 ( Figure 1C) as a template to generate a 8 kb fragment containing the entire pYA4545 plasmid that was amplified with primers pYA4545-TT-BstBI-s and pYA4545-TT-a1 and then extended by PCR with primers pYA4545-TT-BstBI-s and pYA4545-TT-BcII-a.
  • plasmid pG8R314 ( Figure 3) as a template, a 1,242 bp fragment containing Ptrc- ompA was amplified with primers Ptrc-BclI-s and ompA-BstEI-a. The two fragments were then assembled to generated plasmid pG8R320 ( Figure 5B).
  • This regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the cell employs the P cmv promoter to express an inserted gene sequence in eukaryotic cells.
  • the P cmv and Ptrc ompA ⁇ plz4 are separated in the dual plasmid vector pG8R320 by the regulated delayed lysis cassette araC P araBAD GTG murA GTG asdA.
  • Example 5 Display of OmpA ⁇ PLZ4 on the bacterial cell surface enables S. Typhimurium cells to preferentially attach to bladder tumor cells.
  • the ompA ⁇ plz4 mutation was introduced into strain ⁇ 12518 to generate strain ⁇ 12619 using suicide vector ⁇ 7213(pG8R315). Both strains were transformed with plasmid pYA4090 to enable tagging the bacteria with the GFP protein.
  • CXCR3 is expressed on immune cells, such as activated T cells, NK and NKT cells, DCs, but not on naive T cells (161), and a variety of non-immune cells, such as astrocytes, fibroblasts, endothelial cells, epithelial muscle cells, and cancer cells (162, 163).
  • CXCR7 is expressed on multiple immune cells, such as T cells, monocytes, DC cells, B cell and NK cells (164).
  • CXCL11 has diverse functions including inhibiting angiogenesis, increasing immune cell migration, affecting proliferation of different cell types, stimulation of IFN- ⁇ production by immune cells, suppressing M2 macrophage polarization, playing a role in fibroblast directed carcinoma invasion, increasing adhesion and invasion properties, facilitating the migration of certain immune cells, and serving as an adjuvant to anti-cancer therapies (160, 165).
  • CXCL11 mainly works for immune cell migration, differentiation and activation, it could promote cancer cell proliferation and metastasis.
  • Intratumor delivery of CXCL11 has been shown to enhance the efficacy of T-cell infiltration, adoptive T-cell therapy and vaccine efficacy (166-169).
  • CXCL11 in tumor cells will mediate the recruitment of T cells and NK cells to the tumor site to combat tumor development and growth. This will reduce the global toxicity related to overproduction of CXCL11 in non-tumor sites. For these reasons, we determined that the synthesis of CXCL11 by CCST cells would be optimal if the chemokine was synthesized by tumor cells rather than into the environment if synthesized and delivered by the CCST cells being used for combatting bladder cancer.
  • Figure 7 displays diagrams of pG8R321 (A) and pG8R322 (B) that express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after plasmid release of the plasmid in tumor cells employs the P cmv promoter to express the human and mouse CXCL11 chemokines, respectively.
  • pG8R321 To construct pG8R321, we used the CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template to amplify the gene encoding human CXCL11 with primers Human-CXCL11-KpnI-s and Human- CXCL11-Not-a, which was inserted into plasmid pG8R319 cut with KpnI/NotI.
  • the balanced-lethal vector-host combination specifically targets human bladder cancer cells due to the display of the OmpA ⁇ PLZ4 surface protein fusion to induce synthesis of the human CXC11 after invasion into tumor cells to release pG8R321.
  • pG8R322 was similarly constructed using CXCL11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as the template to amplify the gene encoding mouse CXCL11 with primers Mouse-CXCL11- KpnI-s and Mouse-CXCL11-NotI-a. This sequence was then inserted into plasmid pG8R319 cut with KpnI/NotI to generate plasmid pG8R322.
  • the balanced-lethal vector- host targets mouse bladder cancer cells due to the display of the OmpA ⁇ PLZ4 surface protein fusion to induce synthesis of the murine CXC11 after invasion into tumor cells to release pG8R322.
  • Example 7 Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence encoding for synthesis of KillerRed to potentiate tumor cell killing.
  • Photodynamic therapy is an important therapeutic treatment for cancer and other diseases.
  • KillerRed is the first engineered photosensitizer with light-induced cytotoxicity that could be used for precise light-induced cell killing and target protein inactivation (170-175). Upon light activation, KillerRed will produce toxic reactive oxygen species to use for photodynamic therapy against cancer.
  • Plasmid pG8R323 ( Figure 7C) carries the gene encoding a membrane-targeting KillerRed by fusing with Neuromodulin N-terminal sequence (KillerRed mem thereafter) while plasmid pG8R324 ( Figure 7D) carries the gene encoding a mitochondria targeting KillerRed by fusion with mitochondrial location signals (KillerRed mito thereafter). Both plasmids are balanced-lethal plasmids.
  • vectors specify expression of the ompA ⁇ plz4 gene in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after release from the CCST cell in the tumor cell employs the P cmv promoter to express the KillerRed encoding genes to kill tumor cell when induced with light.
  • plasmid pCS2-NXE+mem-KillerRed (Addgene Cat# 45761) as a template to amplify the gene encoding KillerRed mem using primers KillerRed-Mem-KpnI-s and KillerRed-NotIXhoI-a.
  • pG8R324 ( Figure 7D) specifying KillerRed-dMito
  • pKillerRed-dMito (EVROGEN cat# FP964) as a template by amplifying the gene encoding KillerRed mito with primers KillerRed-Mito-KpnI-s and KillerRed-NotIXhoI-a and inserting into plasmid pG8R319 ( Figure 5A) cut with KpnI/NotI to generate plasmid pG8R324.
  • the balanced-lethal vector-host construct carries a mitochondria-targeted KillerRed.
  • KillerRed localized on cellular membranes can be used for effective light-induced cell killing by light-induced production of reactive oxygen species and can also be used to detect tumor cells infected with the CCST cells.
  • the plasmids pG8R321, pG8R322, pG8R323 and pG8R324 ( Figure 7) all have the moderate copy number pBR ori and specify the balanced-lethal phenotype using plasmid encoded expression of the asdA gene.
  • pG8R325 ( Figure 8A) encoding human CXCL11, we amplified a sequence from CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template with primers Human-CXCL11-KpnI-s and Human-CXCL11-NotI-a and cloned into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R325.
  • the lysis vector carries a human CXC11 gene.
  • pG8R326 ( Figure 8B) encoding mouse CXCL11, we amplified a sequence from CXCL11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template with primers Mouse-CXCL11-KpnI-s and Mouse-CXCL11-NotI-a to insert into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI.
  • the pG8R326 lysis vector carries a mouse CXC11 gene.
  • CXCL11 is chemotactic for interleukin-activated T-cells but not unstimulated T- cells, neutrophils or monocytes. It is the dominant ligand for CXCR3.
  • pCS2-NXE+mem-KillerRed Additional ligand for CXCR3.
  • pCS2-NXE+mem-KillerRed Additional ligand for CXCR3.
  • pCS2-NXE+mem-KillerRed Additional pCS2-NXE+mem-KillerRed (Addgene Cat# 45761) as a template to amplify the gene encoding KillerRed mem with primers KillerRed-Mem-KpnI-s and KillerRed-NotIXhoI-a to insert into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI.
  • the pG8R327 lysis vector carries a membrane- targeted KillerRed.
  • pG8R328 Figure 8D
  • pKillerRed-dMito EVROGEN cat# FP964
  • pG8R328 Figure 8D
  • pKillerRed-dMito EVROGEN cat# FP964
  • the lysis vector pG8R328 specifies synthesis of the mitochondria-targeted KillerRed.
  • KillerRed localized on cellular membranes can be used for effective light-induced cell killing by light-induced production of reactive oxygen species.
  • Example 9 Construction of plasmid vectors encoding synthesis of GFP or EGFP to track Salmonella extracellularly and intracellularly to evaluate the targeting ability of CCTS strains to bladder tumors.
  • Figure 9 displays the diagrams of plasmids pG8R341 ( Figure 9A) and pG8R342 ( Figure 9B) used to tag Salmonella with fluorescent proteins.
  • the balanced-lethal plasmid pG8R341 carries a Ptrc promoter which can express the operon fusion of ompA ⁇ plz4 and gfp that enables GFP production in the Salmonella cytosol and displays the synthesized OmpA ⁇ PLZ4 on the cell surface of Salmonella.
  • Ptrc is a prokaryotic promoter that can express at high level protein synthesis under both anaerobic and aerobic conditions and is repressed by LacI (176). Salmonella strains carrying this plasmid in vivo in the absence of arabinose to preclude synthesis of LacI will produce GFP in Salmonella cells present extracellularly and intracellularly.
  • This regulated delayed lysis plasmid pG8R342 ( Figure 9B) has a pUC ori and can express the ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the cell within host cells employs the P cmv promoter to express an inserted egfp in eukaryotic cells.
  • the EGFP is only produced when Salmonella is inside the mammalian cells.
  • plasmid pYA4090 as a template to amplify the gene encoding GFP3 using primers SD-GFP-SacI-gs and GFP-HindIII-ga.
  • the sequence was inserted into plasmid pG8R314 ( Figure 3) cut with SacI/HindIII to generate plasmid pG8R341.
  • the balanced-lethal plasmid uses GFP to track Salmonella with OmpA ⁇ PLZ4.
  • plasmid pYA4685 as a template to amplify the gene encoding EGFP using primers EGFP-KpnI-gs and EGFP-XhoI-ga.
  • the sequence was inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/XhoI to generate plasmid pG8R342.
  • the lysis plasmid uses EGFP to track Salmonella within mammalian cells.
  • Example 10 Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express gene sequences encoding KillerRed to potentiate tumor cell killing and CXCL11 to attract immune cells. A construction that can kill cancer cells and recruit immune cells to tumor cells will have synergic effect to benefit bladder cancer therapy.
  • Figure 10 displays diagrams of such constructions, balanced-lethal plasmids with regulated delayed lysis attributes pG8R343 ( Figure 9A) and pG8R344 ( Figure 9B) with pUC ori that express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the bacterial cell employs the P cmv promoter to express genes encoding KillerRed mem that kill tumor cells and human or mouse CXCL11 that recruit immune cells to combat bladder tumors.
  • a P2A peptide (177-179) was introduced between the genes encoding KillerRed mem and human or mouse CXCL11 to enable ribosome skip to enable synthesis of a peptide bond at the C-terminus of a 2A element leading to cleavage between the end of the 2A sequence and the CXCL11 peptide downstream (177-179).
  • pG8R343 Figure 10A
  • plasmid pCS2-NXE+mem- KillerRed (Addgene plasmid # 45761) as a template to amplify the gene encoding KillerRed mem using primers KillerRed-Mem-KpnI-s and KillerRed-C-P2A-a1.
  • the fragment was used as a template and amplified with primers KillerRed-Mem-KpnI-s and KillerRed-C-P2A-a2 to include the sequence encoding P2A.
  • CXCL11 Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template to amplify the gene encoding human CXCL11 using primers P2A-Human CXCL11-s and Human-CXCL11-NotI-a.
  • the KillerRed Mem-P2A and human CXCL11 fusion sequence was inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R343.
  • the lysis plasmid carries genes encoding KillerRed mem and human CXCL11.
  • the two genes were separated by a P2A peptide (177-179) which can induce cleavage at the C- terminal of the P2A peptide to enable production of both KillerRed mem and human CXCL11 after Salmonella invasion into tumor cells and lysis to release pG8R343 and enable synthesis of KillerRed mem and human CXCL11.
  • pG8R344 Figure 10B
  • the KillerRed mem P2A was generated as above for plasmid pG8R343.
  • CXCL11 Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template to amplify the gene encoding mouse CXCL11 using primers P2A-Mouse CXCL11-s and Mouse- CXCL11-NotI-a.
  • the KillerRed Mem-P2A and mouse CXCL11 fusion was inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R344.
  • the lysis plasmid carries genes encoding KillerRed mem and mouse CXCL11.
  • the two genes were separated by a P2A peptide (177-179) which can induce cleavage at the C- terminal of the P2A peptide to enable production of both KillerRed mem and mouse CXCL11 after Salmonella invasion into tumor cells and lysis to release pG8R344 and enable synthesis of KillerRed mem and mouse CXCL11.
  • Example 11 Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then after invasion express a gene sequence fused with an HLA peptide encoding sequence to potentiate immune responses to tumor cells. Many factors can affect vaccine-induced immune responses.
  • Antigens can be linked to lysosomal or endosomal targeting signals to route the antigen into a MHC class II processing compartment to improve CD4 + T cell responses.
  • a chimeric protein fused with the N-terminal leader peptide with an MHC class I trafficking signal (tail peptide) attached to the C-terminal end of an antigen can strongly improve the presentation of MHC class I and class II epitopes in human and murine dendritic cells, leading to efficient expansion of antigen specific CD4 + and CD8 + T cells and their effector functions (180).
  • tail peptide MHC class I trafficking signal
  • HLAB HLA-B
  • HLAB-Leading- MCS-ga primers for amplify the gene encoding the HLA leading peptide using primers HLAB-Leading-gs and HLAB-Leading- MCS-ga and HLA tail peptide using primers HLAB-tail-MCS-gs and HLAB-tail-XhoI-ga.
  • the sequence generated encoding the above two fragments were inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/XhoI to generate plasmid pG8R345.
  • This plasmid has sequences for HLAB leading and tail peptides which could be used to coupling a selected antigen to MHC Class I Trafficking Signals to increase antigen presentation efficiency (180).
  • This regulated delayed lysis plasmid pG8R345 has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after tumor cell invasion and lysis of the CCST cell employs the P cmv promoter to express the HLA-antigen encoding gene in eukaryotic tumor cells.
  • the egfp is under the control of PCMV which can be expressed in eukaryotic cells.
  • plasmid pYA4685 Figure 2D
  • the sequence was inserted into plasmid pG8R345 ( Figure 11A) cut with KpnI/AvrII to generate plasmid pG8R346.
  • the lysis plasmid carries a gene encoding EGFP and could be used to track the CCTS strain in mammalian cells.
  • pG8R347 Figure 12A
  • HLAB HLA-B
  • NM_005514 Human Untagged Clone
  • Plasmid pG8R3408 To construct plasmid pG8R348, we used plasmid pYA4685 as a template to amplify the gene encoding EGFP using primers encoding EGFP was amplified with primers EGFP(HLAB)-KpnI-gs and EGFP(HLAB)-AvrII-ga and cloned into plasmid pG8R347 ( Figure 12A) cut with KpnI/AvrII to generate plasmid pG8R348.
  • the lysis plasmid carries a gene encoding EGFP and could be used to track the Salmonella in mammalian cells, although this will require replacing the ompA ⁇ plz4 construction that enables targeting to bladder tumor cells with sequences specifying a protein to target other cell types.
  • Tumor neoantigens can be presented by major histocompatibility complex proteins and recognized by T cells to induce anti-tumor immune responses. This approach has been used as therapeutic vaccines in preclinical models to promote tumor specific T-cell responses (185-188). Tumor neoantigens are derived from mutated proteins that lead to the generation of novel immune epitopes that are foreign to the body (189, 190).
  • Vaccines targeting tumor neoantigens are a promising strategy for personalized cancer immunotherapy (188, 190-197). Due to the complex immune tolerance mechanisms in tumors, neoantigen based tumor vaccines are normally combined with immune checkpoint inhibitors. Clinical trials with this combination therapy demonstrated that the induction of neoantigen-specific CD4 + and CD8 + T cell responses and cytotoxic vaccine-induced T cells, had some efficacy in treating bladder cancer (188, 191, 198). Mouse derived BBN963 (199) and MB49 (200, 201) cell lines are commonly used as an in vitro and in vivo model of bladder cancer. Neoantigens have been identified in these two cell lines (202).
  • neoantigens were cloned into vector pG8R347 to generate plasmid pG8R349 ( Figure 12C) and pG8R350 ( Figure 12D) to be used in vaccine trials.
  • the gene encoding neo-antigen BBN963 (202) was cut from with plasmid pUC57-BBN963 with KpnI/AvrII and cloned into plasmid pG8R347 ( Figure 12A) cut with same enzymes to generate plasmid pG8R349.
  • the lysis plasmid carries a gene encoding neo-antigen BBN963.
  • pG8R350 To construct pG8R350, the gene encoding neo-antigen MB49 (202) was cut from with plasmid pJET1.2-MB49 with KpnI/AvrII and cloned into plasmid pG8R347 ( Figure 12A) cut with same enzymes to generate plasmid pG8R350.
  • the lysis plasmid carries a gene encoding neo-antigen MB49.
  • Example 12 Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence under the control of PEF1 ⁇ promoter.
  • the viral derived PCMV promoter is a strong promoter that has been widely used to express genes in eukaryotic cells as in DNA or viral vectors, such as adenovirus. However, it could be silenced in certain cell types due to methylation, leading to considerable variability gene expression in different cell types (203, 204).
  • Human elongation factor-1 ⁇ is a constitutive human promoter that can drive ectopic gene expression homogeneously and persistently in vivo and in vitro (205-207). It can replace PCMV when PCMV has diminished activity due to being silenced.
  • plasmid pLVX EF1 ⁇ IRES Puro N (BEI NR52973) as a template
  • a fragment containing the PEF1 ⁇ promoter was amplified with primers PEF1a-s and PEF1a-KpnI-a.
  • the two fragments were then assembled into plasmid pG8R361.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface to target bladder cancer cells and after lysis of the CCTS cell employs the PEF1 ⁇ promoter to express an inserted gene sequence in eukaryotic cells (208-212), in this case into bladder tumor cells.
  • the PEF1 ⁇ and Ptrc ompA ⁇ plz4 in the dual plasmid vector pG8R361 are separated by the regulated delayed lysis cassette araC P araBAD GTG murA GTG asdA.
  • Example 13 Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence encoding HAC-PD1 to block PD1L1 and activate T cells and CXCL11 to recruit immune cells.
  • the interaction between the Programmed cell death protein-1 (PD-1) and programmed cell death ligand-1 (PD-L1) functions as a T cell checkpoint to regulate T cell responses. Cancer cells upregulate the levels of PD-L1 to evade immune detection and elimination (213).
  • Monoclonal antibodies blocking PD1 and PDL1 have been approved as effective immunotherapies against different tumors (214-219).
  • use of antibodies have inherent limitations that include poor and slow distribution within hypoxic regions of large tumors (220, 221) and immune-related adverse events, such as Fc-mediated cytotoxic immune responses (222) and severe cytokine associated inflammatory and immunological process (223, 224).
  • Fc-mediated cytotoxic immune responses 222
  • severe cytokine associated inflammatory and immunological process 223, 224
  • monoclonal antibodies against PD1/PLD1 they can also reduce circulation of T cell numbers in patients (225-227).
  • HAC high- affinity consensus
  • the HAC- PD1 (228, 229) has an over 40,000-fold higher affinity for PD-L1 than native PD1 (229) and 32- and 12-times higher affinity than the FDA-approved anti-PD-L1 antibodies atezolizumab and durvalumab, respectively (230). At the same dose and schedule through intratumoral injection, it is also more effective than an anti-PD1 antibody in inducing anti-cancer immunity (229).
  • Multiple vectors carrying the gene encoding HAC- PD1 are depicted in Figures 14, 15 and 16.
  • Figure 14 depicts plasmids that contain sequences encoding HAC-PD1 combined with CXCL11 for combinational immunotherapy.
  • Figure 15 depicts plasmids that contain sequences encoding IL2 SS- HAC-PD1 and CXCL11 for combinational immunotherapy.
  • Figure 16 depicts plasmids that carry sequences encoding HAC-PD1, HAC-PD1 and EGFP, CXCL11 and EGFP. All these plasmids display the synthesized OmpA ⁇ PLZ4 on the cell surface to target bladder cancer cells and after lysis of the CCTS cell employ the PCMV or PEF1 ⁇ promoter to express the inserted eukaryotic genes in bladder tumor cells.
  • Plasmid pG8R362 ( Figure 14A), we used plasmid pMal-HAC-PD1 (228, 229) as a template to amplify the gene encoding HAC-PD1 using primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a1 and then extended by PCR with primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a2.
  • CXCL11 Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template
  • the gene encoding human CXCL11 was amplified with primers Linker- hCXCL11-s and Human-CXCL11-NotI-a.
  • the two fragments were inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R362.
  • the lysis plasmid carries the gene encoding HAC-PD1 fused with human CXCL11 with a 36 amino acid linker under the control of a PCMV promoter.
  • the 36 aa linker GGS(GGGSE)5(GGGS)2 was inserted to enable fusion of HAC-PD1 and human CXCL11 (231).
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the fused gene sequence.
  • Plasmid pG8R363 ( Figure 14B), we used plasmid pMal-HAC-PD1 (228, 229) as a template to amplify the gene encoding HAC-PD1 using primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a1 and then extended by PCR with primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a2’).
  • mice CXCL11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template
  • the gene encoding mouse CXCL11 was amplified with primers Linker- mCXCL11-s and Mouse-CXCL11-NotI-a.
  • the two fragments were inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R363.
  • the lysis plasmid carries a gene encoding HAC-PD1 and fused with mouse CXCL11 with a 36 amino acid linker under the control of a PCMV promoter.
  • the 36 aa linker GGS(GGGSE)5(GGGS)2 was inserted to enable fusion of HAC-PD1 and mouse CXCL11 (231).
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the fused gene sequence.
  • plasmid pG8R364 To generate plasmid pG8R364 ( Figure 14C), a fragment encoding HAC-PD1- hCXCL11 was cut from plasmid pG8R362 ( Figure 14A) with KpnI/NotI and cloned into plasmid pG8R361 ( Figure 13) cut with the same enzymes to generate plasmid pG8R364.
  • the lysis plasmid carries a gene encoding HAC-PD1 that was fused with human CXCL11 using a 36 amino acid linker under the control of the PEF1 ⁇ promoter.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the PEF1 ⁇ promoter to express the fused gene sequence.
  • plasmid pG8R365 Figure 14D
  • a fragment encoding HAC-PD1- mCXCL11 was cut from plasmid pG8R363 ( Figure 14B) with KpnI/NotI and cloned into plasmid pG8R361 ( Figure 13) cut with the same enzymes to generate plasmid pG8R365.
  • the lysis plasmid carries a gene encoding HAC-PD1 that was fused with mouse CXCL11 using a 36 amino acid linker under the control of the PEF1 ⁇ promoter.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the PEF1 ⁇ promoter to express the fused gene sequence.
  • Secretion of proteins increases the levels of therapeutic molecules that can significantly enhance the efficacy of therapy at the site of the disease.
  • the IL2 signal peptide is one of the most commonly used secretion facilitating sequences used for protein production in gene therapy research (232-234).
  • plasmid pG8R366 ( Figure 15A) that carries the sequence encoding the IL2 secretion signal.
  • IL2-s2 primers IL2-s2 and IL2-a2.
  • the sequence was inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI to generate plasmid pG8R366.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express any gene sequence fused to the IL2 secretion signal sequence.
  • HAC-PD1 with human and mouse CXCL11 into pG8R366 to generate plasmids pG8R367 ( Figure 15B) and pG8R368 ( Figure 15C), respectively.
  • the fragment encoding HAC-PD1-hCXCL11 was cut from plasmid pG8R362 ( Figure 14A) with KpnI/NotI and inserted into plasmid pG8R366 ( Figure 15A) cut with the same enzymes to generate plasmid pG8R367.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the gene sequence encoding HAC-PD1-human CXCL11 fused to the IL2 secretion signal.
  • the fragment encoding HAC-PD1-mCXCL11 was cut from plasmid pG8R363 ( Figure 14B) with KpnI/NotI and inserted into plasmid pG8R366 ( Figure 15A) cut with the same enzymes to generate plasmid pG8R368.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the gene sequence encoding HAC-PD1-mouse CXCL11 fused to the IL2 secretion signal.
  • Plasmid pG8R367 ( Figure 15B) as a template to amplify the fragment encoding IL2 SS and HAC-PD1 with primers IL2-s2 and HAC-PD1-a.
  • the fragment was inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R372.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the gene sequence encoding HAC-PD1- fused to the IL2 secretion signal.
  • a pUC ori can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the gene sequence encoding HAC-PD1- fused to the IL2 secretion signal.
  • P cmv promoter to express the gene sequence encoding HAC-PD1- fused to the IL2 secretion signal.
  • Plasmid pG8R373 For the construction of plasmid pG8R373, we used plasmid pG8R367 ( Figure 15B) as a template to amplify the fragment encoding IL2 SS-HAC-PD1 fusion using primers IL2-s2 and HAC-PD1-linker- EGFP-a. And then with plasmid pYA4685 ( Figure 2D) as a template, the gene encoding EGFP was amplified with primers HAC-PD1-linker-EGFP-s and C terminal EGFP- XhoINotI-a.
  • the two fragments were inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R373.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the gene encoding fusion of IL2 secretion signal-HAC-PD1- fused and EGFP.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express the gene encoding the fusion of human CXCL11 and EGFP.
  • the 2 fragments were inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R375.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell in situ employs the P cmv promoter to express the gene encoding fusion of mouse CXCL11 and EGFP.
  • Example 14 Insertion of the nucleotide sequence encoding the ten amino acids of the luteinizing hormone-releasing hormone (LHRH) peptide binding to the LHRH receptor into the third exposed loop of the S.
  • LHRH luteinizing hormone-releasing hormone
  • LHRH Luteinizing hormone-releasing hormone
  • Plasmid pG8R380 ( Figure 17A)
  • plasmid pG8R320 Figure 5B
  • plasmid pYA4090 Figure 2C
  • the gene encoding GFP was amplified with primer GFP-s and GFP-BstBI-a.
  • the above 3 fragments were then inserted into plasmid pG8R320 ( Figure 5B) cut with BclI/BstBI to generate plasmid pG8R380.
  • the regulated delayed lysis plasmid has a pUC ori and can express the ompA ⁇ lhrh and gfp gene sequences in Salmonella to display the synthesized OmpA ⁇ LHRH on the cell surface and GFP in the cytosol and after lysis of the CCTS cell employs the P cmv promoter to express a selected gene to specify synthesis of a desired gene product.
  • plasmid pG8R381 ( Figure 17B), the plasmid pG8R380 was cut with BstBI to remove the gene encoding GFP. The 8.1 kb fragment was self-ligated to generate plasmid pG8R381.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ lhrh and gfp in Salmonella to display the synthesized OmpA ⁇ LHRH on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express a selected gene to specify synthesis of a desired gene product.
  • Example 15 Example 15
  • HAC-PD1 (228, 229) has a 40,000-fold higher affinity for inactivating PD-L1 than native PD1 (229) and a 32- and 12-times higher inactivating ability than the FDA- approved anti-PD-L1 antibodies atezolizumab and durvalumab, respectively (230).
  • HAC-PD1 is also more potent than an anti-PD1 antibody in inducing anti-cancer immunity using the same dose and schedule for intratumor injection (229).
  • HAC-PD1 due to its small size, can leak from cells and thus elicit undesired immune responses against normal tissues. Furthermore, HAC-PD1 has a relatively short half-life and thus requires daily intratumoral injections (229). Thus, a HAC-PD1-IgG chimeric protein (haPD1-IgG, thereafter) can retain HAC-PD1 activity in tumors with a prolonged half-life and enhanced efficacy. Since IgG has a long half-life, use of the chimeric fusion protein can reduce the need for frequent administration.
  • Plasmids pG8R382 ( Figure 18A), pG8R383 ( Figure 18B) and pG8R384 ( Figure 18C) carrying a sequence encoding haPD1-IgG.
  • plasmid pG8R382 ( Figure 18A)
  • plasmid pCMV3-haPD1- IgG (Sinobiological, Project number BWH2-P) as a template to amplify the gene encoding haPD1-IgG using primers haPD1IgG-KpnI-s and haPD1IgG-NotI-a.
  • the gene was inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R382.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the CCTS cell surface to target bladder cancer and after lysis of the CCTS cell in tumor cells employs the P cmv promoter to express the haPD1-IgG to prevent inactivation of T cells.
  • Plasmid pG8R383 ( Figure 18B), we used plasmid pCMV3-haPD1- IgG (Sinobiological, Project number BWH2-P) as a template to amplify the gene encoding haPD1-IgG using primers and haPD1IgG-C-P2A-a.
  • plasmid pCS2- NXE+mem-KillerRed (Addgene plasmid # 45761) as a template
  • the gene encoding KillerRed mem was amplified using primers P2A-KillerRed-Mem-s and KillerRed- NotIXhoI-a and then extended by PCR using primers P2A-gs and KillerRed-NotIXhoI-a.
  • the two fragments were then inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R383.
  • the lysis plasmid carries genes encoding haPD1-IgG and KillerRed mem separated by the P2A peptide under the control of the P cmv promoter to enable synthesis of haPD1-IgG and KillerRed mem in tumor cells invaded by the CCTS strain.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express synthesis of haPD1-IgG to prevent inactivation of T cells and KillerRed mem to kill the tumor cells.
  • Plasmid pG8R384 ( Figure 18C)
  • plasmid pG8R343 Figure 10A
  • plasmid pCMV3-haPD1- IgG (Sinobiological, Project number BWH2-P)
  • the gene encoding haPD1- IgG was amplified with primers P2A-haPD1IgG-s and haPD1IgG-NotI-a.
  • the two fragments were then inserted into plasmid pG8R320 ( Figure 5B) cut with KpnI/NotI to generate plasmid pG8R384.
  • the lysis plasmid carries genes encoding KillerRed mem and haPD1-IgG separated by P2A peptide under the control of a P cmv promoter to enable synthesis of KillerRed mem and haPD1-IgG.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ plz4 in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express KillerRed mem to kill the tumor cell and haPD1-IgG to prevent the inactivation of T cells.
  • the only difference between pG8R343 and pG8R344 is the order of the sequences encoding the synthesis of haPD1-IgG and KillerRed.
  • Example 16 Insertion of the nucleotide sequence encoding the single-chain fragment variable (scFv) targeting HER2 into the third exposed loop of the S.
  • HER2 Human epidermal growth factor receptor 2
  • HER2 Human epidermal growth factor receptor 2
  • scFv Single-chain fragment variables
  • the scFv targets Her2 to bind to ErB2+ cells to potentiate delivery of exogenous DNA and siRNA into ErB2+ cells (249-251).
  • Plasmid #10794 plasmid pG8R385
  • plasmid pYA4090 ( Figure 2C) as a template, the gene encoding GFP was amplified with primer GFP-s and GFP-BstBI-a. The 4 fragments were inserted into plasmid pG8R320 cut with BclI/BstBI to generate plasmid pG8R385.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ Her2 scFv and gfp in Salmonella to display the synthesized OmpA ⁇ Her2 scFv on the CCTS cell surface and GFP in the cytosol and after lysis of the CCTS cell in a cancer cell employs the P cmv promoter to express an inserted gene sequence of importance for cancer cell/tumor therapy.
  • plasmid pG8R386 Figure 19B
  • the plasmid pG8R385 was cut with BstBI to remove the gene encoding GFP.
  • the 8.9 kb fragment was self-ligated to generate plasmid pG8R386.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ Her2 scFv in Salmonella to display the synthesized OmpA ⁇ Her2 scFv on the cell surface and after lysis of the CCTS cell employs the P cmv promoter to express an inserted gene sequence of importance for cancer cell/tumor therapy.
  • Example 17 Construction of universal vaccine vectors to enable expression of both bacterial and eukaryotic genes by insertion of selected nucleotide sequences after the Ptrc or Ptrc bla SSopt promoter and the PCMV promoter, respectively.
  • the regulated delayed lysis plasmid pG8R320 ( Figure 5B) has a pUC ori and can express the bacterial gene ompA ⁇ plz4 under the control of the Ptrc promoter in Salmonella to display the synthesized OmpA ⁇ PLZ4 on the cell surface and after lysis of the cell employs the P cmv promoter to express an inserted gene in eukaryotic cells. It is mainly used with Salmonella strains possessing the ⁇ ompA mutation and mainly targeting bladder cancer cells with PLZ4 peptide. This mutation may not be required in all situations. The targeting to bladder cancer cells limits its usage for other cancer cells.
  • a universal vector without the OmpA ⁇ PLZ4 can be used for other purposes, such as targeting other cancer types or as a dual antigen delivery system.
  • plasmids pG8R388 ( Figure 19A) and pG8R389 ( Figure 19B) as two universal vectors to enable their use to express and deliver proteins of both bacterial and eukaryotic origins.
  • plasmid pYA3342 Figure 1A
  • plasmid pYA3342 Figure 1A
  • the fragment was inserted into plasmid pG8R320 ( Figure 5B) cut with BclI/BstBI to generate plasmid pG8R388.
  • the regulated delayed lysis plasmid has a pUC ori and can express bacterial genes in the Salmonella cytosol and after invasion into a eukaryotic cell and lysis can employ the P cmv promoter to express the eukaryotic gene in animal cells.
  • plasmid pG8R114 ( Figure 2B) as a template to amplify a 237 bp fragment containing the Ptrc promoter with an optimized bla secretion signal (SS) (68) using primers Ptrc-BclI-s and 4545PtrcBlaAAA-a .
  • the fragment was inserted into plasmid pG8R320 ( Figure 5B) cut with BclI/BstBI to generate plasmid pG8R389.
  • the regulated delayed lysis plasmid has a pUC ori and can express bacterial gene fused with bla SSopt in Salmonella and secrete the synthesized protein into the periplasm and invasion into a eukaryotic cell and lysis can employ the P cmv promoter to express the eukaryotic gene in animal cells.
  • Example 18 Construction of dual plasmids to cause CCTS strains to target bladder tumor cells with LHRH peptide or HER2 scFv and then express a gene sequence encoding KillerRed to potentiate tumor cell killing.
  • plasmids pG8R390 Figure 21A
  • pG8R391 Figure 21B
  • pG8R418 Figure 21C
  • KillerRed mem to potentiate tumor cell killing.
  • Figure 20A the gene encoding KillerRed mem was cut from pG8R327 ( Figure 8C) with KpnI/Not and cloned into plasmid pG8R381 ( Figure 17B) cut with KpnI/NotI to generate plasmid pG8R390.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ lhrh in Salmonella to display the synthesized OmpA ⁇ LHRH on the cell surface and after lysis of the CCTS cell in situ employs the P cmv promoter to express the gene encoding KillerRed mem to potentiate tumor cell killing.
  • pG8R391 Figure 20B
  • the gene encoding KillerRed mem was cut from pG8R327 ( Figure 8C) with KpnI/Not and cloned into plasmid pG8R386 ( Figure 19B) cut with KpnI/NotI to generate plasmid pG8R391.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ her2 scFv in Salmonella to display the synthesized OmpA ⁇ Her2 scFv on the CCTS cell surface and after lysis of the CCTS cell after invading into a cancer cell employs the P cmv promoter to express the gene encoding KillerRed mem to potentiate tumor cell killing.
  • the regulated delayed lysis plasmid has a pUC ori and can express ompA ⁇ her2 scFv in Salmonella to display the synthesized OmpA ⁇ Her2 scFv on the CCTS cell surface and after lysis of the CCTS cell after invading into a cancer cell employs the P cmv promoter to express the gene encoding KillerRed mem to potentiate tumor cell killing.
  • Example 19 Construction of recombinant plasmid CCTS strains with potential to attach to and invade into bladder tumor cells to deliver desired cargo to directly and/or indirectly reduce tumor survival.
  • All of the candidate CCTS strains listed in Table 4 possess the ⁇ ompA11 mutation which can be substituted by a chromosomal ompA ⁇ plz4 fusion allele and deletion of the asdA gene to enable establishment of a balanced-lethal vector-host strain after introduction of any of the recombinant plasmid vectors displayed in Figures 3 to 5 and 7 to 21, all of which encode synthesis of a receptor ligand facilitating specific targeting to bladder tumor cells.
  • strains have the ⁇ P murA25 ::TT araC P araBAD murA mutation with a ⁇ asdA mutation to enable the display of the regulated delayed lysis in vivo phenotype that is desirable for delivery of cargoes synthesized by the CCTS strain and also for the delivery of plasmids serving as DNA vaccines to enable synthesis of a desired gene product by tumor cells.
  • the inclusion of the relA mutation facilitates the completeness of lysis by uncoupling the dependance of growth on continued protein synthesis.
  • Most CCTS strains in Table 4 display a means to cause a regulated delayed attenuation in vivo by cessation in the synthesis of the LPS outer core or the LPS O- antigen.
  • This phenotype also enhances the efficiency of CCTS strains to attach to and invade eukaryotic cells and in this case better display the modified outer membrane protein OmpA ⁇ PLZ4 that is the means for targeting specific attachment to bladder tumor cells.
  • These CCTS strains also have a means to display a regulated delayed synthesis of Ptrc regulated gene insertions by the araC P araBAD regulated expression of the lacI gene, an attribute that enhances the efficiency and frequency of in vivo colonization of the CCTS strains in target tissues.
  • ⁇ (wza-wcaM)-8 mutation facilitates complete lysis of strains with the regulated delayed lysis phenotype, enhances levels of plasmid encoded protein synthesis and precludes synthesis of exopolysaccharides that contribute to biofilm formation.
  • Most strains have a ⁇ recF mutation to reduce inter- and intra-plasmid recombination to enhance construct stability and a ⁇ endA mutation to eliminate the endonuclease that could degrade the plasmid vector upon lysis of the CCTS cell.
  • a ⁇ sifA mutation that enables the CCTS strain after invasion into a cell to escape the Salmonella containing vesicle (SCV) or endosome.
  • the DNA vaccine must be free to be directed to the tumor cell nucleus to enable transcription of the inserted gene sequence under the control of the plasmid encoded P CMV or P EF1 ⁇ promoter.
  • the DNA vaccine components of all the plasmids containing sequence from pYA4545 ( Figure 1C) contain multiple sequences that direct the plasmid DNA to the cell nucleus (45).
  • CCTS cells delivering DNA vaccines possess mutations such as the ⁇ sseL and ⁇ tlpA mutations to delay onset of pyroptosis and thus enhance expression of DNA vaccine encoded genes.
  • mutations such as the ⁇ sseL and ⁇ tlpA mutations to delay onset of pyroptosis and thus enhance expression of DNA vaccine encoded genes.
  • many of the strains listed in Table 4 have such mutations and these mutations can be added to other strains using the suicide vectors listed in Table 2.
  • Some of the strains listed in Table 4 have deletion mutations in the pagL, pagP, lpxR, eptA and arnT genes that alter the structure and activities of the LPS lipid A.
  • mutations may or may not contribute to the efficacy of CCTS constructs by altering the degree of inflammation in interacting with TLR4. Further modification of these activities can be accomplished by inclusion of the ⁇ pagP81::P lpp lpxE and/or the ⁇ lpxR93::P lpp lpxF deletion-insertion mutations that cause expression of codon- optimized Francisella tularensis genes to delete the 1’ of 4’ phosphates from lipid A to render it non-toxic but retain ability to bind to and activate TLR4.
  • CCTS gene activities are regulated by the sugars arabinose and rhamnose that must be supplied during in vitro cultivation but are absent in animal tissues
  • the timing of shut off of sugar-regulated gene expression after CCTS cell entry into an animal (human) host can be modulated by whether the sugars supplied during in vitro growth and retained in the cells are or are not quickly metabolized.
  • the mutations ⁇ araBAD65::TT and ⁇ rhaBADSR515 are sometimes added to CCTS strains to delay shut off of the sugar regulated genes for several cell divisions.
  • strains can be achieved by inclusion of mutations such as ⁇ Pt olR67 ::::TT araC P araBAD tolR that acts to increase production and release of outer membrane vesicles and other mutations that alter display or non-display of flagellar and fimbrial appendages or component parts to alter recruitment of innate immunity. These activities are well described in WO 2020/096994 A1 and WO 2021/222696 A1.
  • Example 20 Evaluation of CCTS constructs for ability to attach to and invade bladder tumor cells.
  • CCTS strains with pG8R341 specifying synthesis of GFP by the CCTS strain and pG8R342 in which the EGFP activity must be synthesized by the bladder tumor cell after invasion by the CCTS strain.
  • constructs in ⁇ 12417 ⁇ P murA25 ::TT araC PBAD murA ⁇ waaL46 ⁇ pmi-2426 ⁇ asdA27::TT araC P BAD c2 ⁇ pagL64::TT rhaRS P rhaBAD waaL ⁇ (wza-wcaM)-8 ⁇ relA197::araC PBAD lacI TT ⁇ recF126 ⁇ sifA26 ⁇ ompA11) that was used for our initial work and the much improved strains ⁇ 12735 ( ⁇ P murA25 ::TT araC P araBAD murA ⁇ asdA33 ⁇ waaL46 ⁇ pagL38:::
  • Strains will be grown in LB broth with 0.1% arabinose and with and without 0.1% rhamnose and evaluated for ability to attach to and invade bladder tumor cells as described in Example 1 and Figure 6. It is expected based on the information provided in forgoing Examples that strains grown without rhamnose to prevent synthesis of the LPS O-antigen will be most proficient in attaching to bladder tumor cells. It is also expected that CCTS strains ⁇ 12735 and ⁇ 12736 will be most proficient in inducing synthesis of EGFP.
  • Example 21 Exemplary sequences Sequences and SEQ ID NOs related to embodiments described herein are provided infra before the references.
  • Example 22 Evaluation of the effect of O-antigen on the CCTS constructs for ability to attach to and invade bladder tumor cells.
  • LPS Lipopolysaccharide
  • the LPS mutations, ⁇ waaL46, ⁇ waaG42, and ⁇ waaC41, which enable Salmonella to display defects in the synthesis of O-antigen, outer core and inner core, respectively, were introduced into Salmonella strains. It should be noted that waaC mutant defective in synthesis of the inner core are unable to synthesize and assemble the outer core and O-antigen whereas mutants unable to synthesize the outer LPS core also unable to display the O-antigen. These mutations were introduced into ⁇ 12614 to yield strain ⁇ 12812, ⁇ 12813, and ⁇ 12814, respectively (Table 5). A plasmid pG8R341 carrying multiple copies of ompA ⁇ plz4 was introduced into these strains.
  • PCR with correspondent primers proved that strains derived from ⁇ 12614 have the correct expected genotype and the LPS gel proved that each strain has the right LPS phenotype human bladder cancer cell 5637 and mouse bladder cancer cells MB49 and BBN967 were performed in 24-well culture plates as described previously (152).
  • the ⁇ 12614 lineage strains were grown in LB media until OD600 reached 0.85 ⁇ 0.9.
  • the bacteria were collected and resuspended in DMEM media with 10% fetal bovine serum.
  • a MOI 10:1 was used to infect cells for 1 hour. After infection, half of the monolayers were washed with PBS and lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of attached bacteria.
  • strain ⁇ 12812 with ⁇ waaL46 mutation displays the highest abilities in attachment and invasion compared to the other strains abilities to attach to and invade both human and mouse bladder cells.
  • the LPS mutations, ⁇ waaL46, ⁇ waaG42, and ⁇ waaC41 were also introduced into strain ⁇ 12619 to generate ⁇ 12808, ⁇ 12809, and ⁇ 12810, respectively.
  • the ompA ⁇ plz4 mutation (Table 2) was also introduced into strain ⁇ 12542 (Table 5) to yield strain ⁇ 12811 which has the identical genotypes as strain ⁇ 12810. These strains only have one copy of ompA ⁇ plz4 in the chromosome. PCR with correspondent primers proved that strains derived from ⁇ 12619 have the correct expected genotype and the LPS gel proved that each strain had the right LPS phenotype ( Figure 23 A and B) . All the strains grew on LB with arabinose plates, but not not on LB, LB with DAP and LB with alanine plates ( Figure 23C).
  • strain ⁇ 12417 was transformed with plasmids pG8R385, pG8R418 and pG8R391 that carry the ompA ⁇ her2 ScFV gene.
  • the strains were grown in LB with 0.1% arabinose.1 mM IPTG was added to induce the production of OmpA ⁇ Her2 ScFV for 4 hours.
  • the Salmonella outer member proteins (SOMPs) were prepared as described previously (62).
  • the OmpA ⁇ Her2 in strain ⁇ 12417 carrying any of the above plasmids can be detected in SOMPs portion of the SDS PAGE gel by Coomassie blue staining and western blot using anti-His6 antibody as an expected band around 65.1 kDa, but not the SOMPs from strain ⁇ 12417 (with no plasmid) (Figure 24).
  • Different cell lines have different HER2 expression status (253).
  • Her2 overexpression (SKBR-3, ATCC®HTB30) and low expression cell lines (MDA-MB- 231(ATCC® CRM-HTB-26) and MDA-MB-468 (ATCC® HTB-132)) were used to detect the attachment and invasion of the ⁇ 12417(pG8R385) and ⁇ 12417(pG8R391) strains ( Figure 25).
  • Overnight cultures of ⁇ 12417(pG8R385) and ⁇ 12417(pG8R391) were diluted into LB broth with 0.1% arabinose and grown until OD600 reached 0.9.
  • the bacteria were washed once with PBS and then used to infect SKBR-3, MDA-MB-431 and MDA-MB-468 breast cancer cells at an MOI 10:1 for 1 hour. After infection, half of the monolayers were washed with PBS and lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of attached bacteria. The other half of the monolayers were treated for 1 h with DMEM media containing 100 ug/ml gentamicin to eliminate extracellular bacteria. Monolayers were then lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of internalized bacteria.
  • Example 25 KillerRed kills HEK293T cells.
  • HEK293T cells were transfected with plasmid pG8R327 ( Figure 8C). The fluorescence was observed using EVOS Fl with RFP channel (Thermofisher). The RFP channel in EVOS Fl has 531/40 nm excitation and 593/40 nm whatsoever.
  • KillerRed can absorb 540 ⁇ 580 nm wavelength light and emit an longer 610 nm red light with maximum fluorescence excitation/emission at 585/610 nm.
  • the RFP channel is not the optimal wavelength for KillerRed, strong red fluorescence signals were observed in HEK293T cells transfected with plasmid pG8R327 ( Figure 26). Before RFP channel excitation, the cells showed spindle shapes with smooth surfaces. After excitation for 10 min, membrane blebs appear on the surface of the cells ( Figure 27). The phenomena was also observed in other samples. Further excitation for an additional 10 min led to the cell morphology changing to round instead of spindle shaped ( Figure 28).
  • Example 26 Evaluation of the CCTS strains with targeting peptide in vivo.
  • the safety of CCTS strains uses oral and intravenous routes. Groups of 5 mice are inoculated with CCTS strains at varying doses ranging from 10 4 to 10 9 CFU. The mice are closely monitored for one month to see if any disease symptoms are observed. Studies are conducted using multiple subcutaneous syngeneic tumor models to evaluate the distribution of CCTS strains based on the specific targeting peptide utilized. To establish the subcutaneous syngeneic tumor, approximately 1-5x10 6 tumor cells are injected into the right flank of 6-8 week old mice.
  • mice When tumor sizes reach approximately 100 mm 3 , the mice are administrated with the CCTS strains intravenously and orally using the highest safe dose determined from the previous experiment. At 24, 48, 72 and 96 hours, the spleen, liver, heart, lung, kidney, and tumors is harvested, weighted, homogenized and plated on LB agar with supplements. Fluorescence detection is performed on the tumors and organs to identify the presence of CCTS strains carry plasmids with the KillerRed or EGFP genes. To test the target ability of the CCTS strain displayed PLZ4 peptide, BBN963 or MB49 murine bladder cancer cells are injected into the right flank of C57BL/6 mice.
  • mice are treated with CCTS strains with plasmids carrying the ompA3 ⁇ plz4 gene and monitored as above.
  • high LHRH receptor expression cell lines such as A2780 human ovarian cancer cells (1-5x10 6 ) (235, 241, 243) or human breast cancer cells MCF-7 (ATCC® HTB-22) (254) , MDA-MB-231 (ATCC® HTB-26), HCC1806 (ATCC® CRL-2335) are used to generate xenographs in athymic nu/nu mice.
  • Mice are treated with a CCTS strain with a plasmid specifying the LHRH peptide and monitored as above.
  • Her2 overexpression SKBR-3, ATCC®HTB30
  • low expression cell lines MDA-MB- 231(ATCC® CRM-HTB-26) and MDA-MB-468 (ATCC® HTB-132)
  • mice are treated with a CCTS strain with a plasmid specifying Her2 SCFV and monitored as above.
  • Over 60% of bladder tumors have little immune cell infiltration inside tumors (255).
  • CXCL11 is a cytokine that can attract CD8 cytotoxic T cells (256, 257).
  • mice When tumors in C57BL/6 mice reach around 100 mm 3 , mice are fed with PBS, CCTS(pG8R319), CCTS(pG8R320), CCTS(pG8R322), or CCTS(pG8R326). Tumors will be harvested 24, 48, 72 or 96 hours later. Flow cytometry is used to compare the amount of CD8+ cells in different groups at these time points. To further evaluate the abilities of CCTS strains to destroy tumors, C57BL6 mice carrying subcutaneous bladder tumors are used.
  • mice When tumors reach the size of around 100 mm 3 , groups of 5 tumor-bearing mice are treated with a safe dose of CCTS strains with difference cargos, including CXCL11, haPD1, haPD1-IgG. The treatment could be once or multiple times. Mice are monitored for weight, tumor growth and survival. If tumors regress, mice are maintained and monitored to see if regrowth of tumors does or does not occur. Mice are euthanized once tumor sizes reach 1,500 mm 3 or at the humane endpoint. Tumors are excised at the endpoint and either frozen directly in liquid nitrogen for storage or fixed in 10% formalin for histology or immunohistochemistry.
  • tumors are collected before humane endpoint and split into 3 portions: formalin-fixation for immunohistochemical staining, fresh frozen for RNA and DNA extraction and deep sequencing; and single cell suspension for single cell sequencing.
  • formalin-fixation for immunohistochemical staining
  • fresh frozen for RNA and DNA extraction and deep sequencing
  • single cell suspension for single cell sequencing.
  • tumor-bearing mice are treated with a safe dose of CCTS strains specifying synthesis of KillerRed. Fluorescence imaging of tumors are acquired daily in vivo using an IVIS-Spectrum (PerkinElmer, USA) with excitation wavelength of 570 nm and emission wavelength of 620 nm. A suitable wavelength is used for PDT.
  • tumor-bearing mice are treated with a safe dose of CCTS strains delivering KillerRed. Tumors are either treated with a continuous wave or pulsed laser without causing excessive temperature effect on the skin surface.
  • a validation parameter is described by Shirmanova et .al (259).
  • the PDT is carried at 593 nm, 150 mW/cm 2 , 270 J/cm 2 for the continuous laser wave daily for 7 days, or at 584 nm, 225 mW/cm 2 , 337 J/cm 2 for the pulsed laser on the days 6, 7, and 8 of tumor growth.
  • Skin surface temperature is monitored using an Infrared thermograph.
  • randomly selected tumors in treated and untreated groups is collected and split into 3 portions: formalin-fixation for immunohistochemical staining, fresh frozen for RNA and DNA extraction and deep sequencing; and single cell suspension for single cell sequencing. During these studies, tumors are monitored with the size measured with a caliper twice a week until the mice reach humane endpoint.
  • Example 28 Example 28.
  • Universal vaccine vectors to enable expression of both bacterial and eukaryotic genes by insertion of selected nucleotide sequences after the Ptrc or Ptrc bla SSopt promoter and the PCMV promoter, respectively.
  • a universal vaccine vector is a single vector that enables expression of genes both in prokaryotic and eukaryotic cells, even though it only specifies expression of gene only either in prokaryotic or eukaryotic cells.
  • the regulated delayed lysis plasmids pG8R388 ( Figure 19A) and pG8R389 ( Figure 19B) are examples of two universal vectors to enable expression and delivery of proteins of both bacterial and eukaryotic origins.
  • Plasmid pG8R388 has a pUC ori and can express bacterial genes in the cytosol or on the surface of Salmonella and after invasion into a eukaryotic cell and lysis can employ the P cmv promoter to express the eukaryotic gene(s) in animal cells.
  • Plasmid pG8R389 has a pUC ori and can express a bacterial gene fused with bla SSopt in Salmonella and secrete the synthesized protein into the periplasm and upon invasion into a eukaryotic cell and lysis can employ the P cmv promoter to express the eukaryotic gene in animal cells.
  • the prokaryotic promoter is not limited to Ptrc and could be any promoter that functions in prokaryotic cells.
  • prokaryotic promoters can be used. Multiple prokaryotic promoters can be used to drive the expression of multiple genes. Other secretion signals can be used to replace the bla SSopt.
  • the eukaryotic promoter is not limited to P cmv or PEF1 ⁇ and could be any promoter that functions in eukaryotic cells. Other eukaryotic promoters can be used. Multiple eukaryotic promoters can also be used to drive the expression of multiple genes. Under the control of a prokaryotic promoter, a single gene can be expressed and multiple genes can also be expressed as an operon or protein fusion with or without suitable linkers. These linkers could be flexible, rigid, cleavable or dipeptide linkers.
  • FIG. 17A illustrates plasmid pG8R380, in which an operon fusion links ompA ⁇ lhrh and gfp.
  • Figure 19A and Figure 21C illustrates plasmids pG8R385 and pG8R418, in which an operon fusion links ompA ⁇ her2 SCFV and gfp.
  • a single gene Under the control of a eukaryotic promoter, a single gene can be expressed and multiple genes can also be expressed separated by one or multiple 2A cleavage peptides.
  • the 2A peptide could be P2A, E2A, T2A, F2A or other 2A-like sequences and thus form bi-, tri-, and quad-, penta- or multiple cistronic vectors (263-265).
  • Plasmid pG8R343 (Fig.10), pG8R344 (Fig.10), pG8R383 (Fig.18), pG8R384 (Fig.18) are examples of bicistrion vectors with the P2A peptide.
  • IRES sequences could be used to generate multicistronic constructions for simultaneously expression of multiple genes (177, 179, 266-269). Multiple gene can be linked by a linker described above.
  • Figure 14 and Figure 15B and 15C illustrate examples in which HAC PD1 is linked to CXCL11 through a linker.
  • Figure 16B-D illustrate examples in which HAC PD1 is linked to EGFP through a 5A linker PPVAT.
  • Neuromodulin N-terminal sequence (mem)(SEQ ID NO: SEQ ID NO: 26 (DNA) and SEQ ID NO: 27 (amino acid))
  • GSG P2A sequence SEQ ID NO: 30 (DNA) and SEQ ID NO: 31 (amino acid)
  • Neuromodulin N-terminal sequence (mem)(SEQ ID NO: 34 (DNA) and SEQ ID NO: 35(amino acid))
  • GSG P2A sequence (SEQ ID NO: 42 (DNA) and (SEQ iD NO: 43 (amine acid))
  • Her2 ScFV sequence (SEQ ID NO: 137)
  • HAC-PD1 sequence (SEQ ID NO : 48(DNA) and SEQ ID NO: 49(amino acid) haPD1-lgG sequence (SEQ ID NO: 50 (DNA) and SEQ ID NO: 51(arnino acid))
  • GSG T2A sequence (derived from thoseaasigna virus 2A ) (SEQ. ID NO: 129
  • GSG E2A sequence (derived from equine rhinitis A virus )(SEQ ID NO: 130
  • GSG F2A sequence (derived from foot-and-mouth disease virus) (SEQ ID NO: 131. (amino acid))
  • Attenuated Salmonella typhimurium containing interleukin-2 decreases MC-38 hepatic metastases: a novel anti-tumor agent. Cancer Biother Radiopharm 11:145-153.
  • Ameiss K, Ashraf S, Kong W, Pekosz A, Wu WH, Milich D, Billaud JN, Curtiss R, III.2010 Delivery of woodchuck hepatitis virus-like particle presented influenza M2e by recombinant attenuated Salmonella displaying a delayed lysis phenotype.
  • Plasmidic recombination in Escherichia coli K-12 the role of recF gene function. Mol Gen Genet 189:471-474. 124. Kolodner R, Fishel RA, Howard M.1985. Genetic recombination of bacterial plasmid DNA: effect of RecF pathway mutations on plasmid recombination in Escherichia coli. J Bacteriol 163:1060-1066. 125. Zhang X, Wanda SY, Brenneman K, Kong W, Roland K, Curtiss R, III.2011. Improving Salmonella vector with rec mutation to stabilize the DNA cargoes. BMC Microbiol 11:31. 126.
  • Salmonella maintains the integrity of its intracellular vacuole through the action of SifA. EMBO J 19:3235-3249. 127. Ohlson MB, Huang Z, Alto NM, Blanc MP, Dixon JE, Chai J, Miller SI.2008. Structure and function of Salmonella SifA indicate that its interactions with SKIP, SseJ, and RhoA family GTPases induce endosomal tubulation. Cell Host Microbe 4:434-446. 128.
  • Saltzman DA Katsanis E, Heise CP, Hasz DE, Kelly SM, Curtiss R, III, Leonard AS, Anderson PM.1997. Patterns of hepatic and splenic colonization by an attenuated strain of Salmonella typhimurium containing the gene for human interleukin-2: a novel anti-tumor agent. Cancer Biother Radiopharm 12:37-45. 129. Saltzman DA, Katsanis E, Heise CP, Hasz DE, Vigdorovich V, Kelly SM, Curtiss R, III, Leonard AS, Anderson PM.1997.
  • Antitumor mechanisms of attenuated Salmonella typhimurium containing the gene for human interleukin-2 a novel antitumor agent? J Pediatr Surg 32:301-306. 130. Schödel F, Kelly SM, Peterson DL, Milich DR, Curtiss R, III.1994. Hybrid hepatitis B virus core-pre-S proteins synthesized in avirulent Salmonella typhimurium and Salmonella typhi for oral vaccination. Infect Immun 62:1669- 1676. 131. Augustin LB, Milbauer L, Hastings SE, Leonard AS, Saltzman DA, Schottel JL.2021.
  • CXCR7 functions as a scavenger for CXCL12 and CXCL11.
  • CXCL11 Correlates With Antitumor Immunity and an Improved Prognosis in Colon Cancer. Front Cell Dev Biol 9:646252. 167. Moon EK, Wang LS, Bekdache K, Lynn RC, Lo A, Thorne SH, Albelda SM. 2018. Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines. Oncoimmunology 7:e1395997. 168.
  • mRNA translation influence of the 5' and 3' untranslated regions. Curr Opin Genet Dev 4:310-315. 183. Yamashita A, Takeuchi O.2017. Translational control of mRNAs by 3'- Untranslated region binding proteins. BMB Rep 50:194-200. 184. Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, Tureci O, Sahin U.2006. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood 108:4009-4017. 185.
  • Murine bladder carcinoma cells present antigen to BCG-specific CD4+ T-cells. Cancer Res 52:4286-4290. 202. Saito R, Smith CC, Utsumi T, Bixby LM, Kardos J, Wobker SE, Stewart KG, Chai S, Manocha U, Byrd KM, Damrauer JS, Williams SE, Vincent BG, Kim WY. 2018. Molecular Subtype-Specific Immunocompetent Models of High-Grade Urothelial Carcinoma Reveal Differential Neoantigen Expression and Response to Immunotherapy. Cancer Res 78:3954-3968. 203. Prosch S, Stein J, Staak K, Liebenthal C, Volk HD, Kruger DH.1996.
  • Plasmid vectors harboring cellular promoters can induce prolonged gene expression in hematopoietic and mesenchymal progenitor cells.
  • the EF- 1 ⁇ promoter maintains high-level transgene expression from episomal vectors in transfected CHO-K1 cells. J Cell Mol Med 21:3044-3054.
  • Gill DR Smyth SE, Goddard CA, Pringle IA, Higgins CF, Colledge WH, Hyde SC.2001.
  • nivolumab an anti-PD-1 immune checkpoint inhibitor
  • CheckMate 063 a phase 2, single-arm trial. Lancet Oncol 16:257-265. 219. Tobias J, Steinberger P, Drinic M, Wiedermann U.2021. Emerging targets for anticancer vaccination: PD-1. ESMO Open 6:100278. 220. Lee CM, Tannock IF.2010. The distribution of the therapeutic monoclonal antibodies cetuximab and trastuzumab within solid tumors. BMC Cancer 10:255. 221.
  • Tan S Liu K, Chai Y, Zhang CW, Gao S, Gao GF, Qi J.2018. Distinct PD-L1 binding characteristics of therapeutic monoclonal antibody durvalumab. Protein Cell 9:135-139. 231. Li Y, Cong Y, Jia M, He Q, Zhong H, Zhao Y, Li H, Yan M, You J, Liu J, Chen L, Hang H, Wang S.2021. Targeting IL-21 to tumor-reactive T cells enhances memory T cell responses and anti-PD-1 antibody therapy. Nat Commun 12:951. 232. Sasada R, Onda H, Igarashi K.1987. The establishment of IL-2 producing cells by genetic engineering.

Landscapes

  • Genetics & Genomics (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A genetically modified Salmonella cell (GMSC) engineered to exhibit specific targeting to cells and regulated delayed lysis in vivo, the GMSC comprising a first heterologous nucleic acid that encodes a first gene product that causes the GMSC to be selectively localized to and/or internalized by a target cell in vivo and a second heterologous nucleic acid that encodes a second gene product that facilitates killing of the target cells following internalization.

Description

Improved Salmonella vectored therapies for treatment of cancer
Background
Cancer represents a diversity of disease states characterized by unregulated proliferation of cells that are either freely multiplying in blood and/or lymph or organized into tumor masses. After cardiovascular disease, cancer ranks as the second most common cause of death in the US (1 ).
Bacteria have been used to target cancers since Coley’s observation over 100 years ago that tumors regressed in cancer patients infected with Streptococcus pyogenes (2, 3). Later, he used killed S. pyogenes, known as Coley’s toxin, to treat cancer patients. Unfortunately, the trials of using bacteria as cancer therapy agents stopped for almost 70 years. After Malmgren demonstrated that Clostridium tetani could survive and replicate in necrotic tumors in 1955, studies using bacteria as cancer therapy recommenced and now are widespread in preclinical and clinical studies (4, 5). Many bacteria have been investigated for their anti-cancer ability, including Bifidobacterium infantis (6), Escherichia coli (7), C. tetani, Listeria monocytogenes (8) and Salmonella Typhimurium. While obligate anaerobes, such as Bifidobacterium and Clostridium, are highly effective at accumulating and replicating in necrotic tumors, they do not grow in viable tumor tissues, which limits their efficacy as anti-cancer agents. S. Typhimurium is a facultative anaerobe, which can survive and grow in anoxic regions as well as in viable oxygenic regions of tumors. Salmonella also has ability to identify and penetrate tumors by detecting small molecules such as serine and aspartate in tumors, and accumulates in tumors that contain free amino acids, purines and pyrimidines that facilitate Salmonella growth. As Salmonella are easily genetically manipulated, and attenuated Salmonella still retain their tumor-targeting and natural tumor-regressing capabilities, they became safe enough to evaluate in tumor-bearing mice and humans. Therefore, S. Typhimurium is widely investigated as an anti-cancer agent (see (5)). Currently, many researchers use S. Typhimurium VNP20009 or its derivatives as the anti-cancer agent or as a vector to investigate efficacy of anti-cancer activities (see (5, 9)). While VNP20009 carrying a purine auxotrophic mutation (pur/) and lipid A mutation (msbB) and its derivatives demonstrated good anti-tumor efficacy in mice, anti-cancer efficacy in human trials was not achieved in phase I clinical trials in patients with metastatic melanoma and renal carcinoma (10, 11). In VNP20009-immunized dogs with a variety of malignant tumors, bacterial colonization of tumors was observed but only 4 of 35 dogs tested were completely cured (12). Even intratumoral injection in humans with cancer only led to colonization in 2 out of 3 patients (13). The reasons for failure in human clinical trials may be that the parent of VNP20009 is not highly virulent and its genetic construction is not precise. VNP20009 is derived from ATCC 14028, which does not show high virulence and invasiveness compared to other S. Typhimurium strains (14) and we demonstrated that an attenuated aroA mutant of 14028 was not as immunogenic and did not induce as high protective immune levels as did an isogenic derivative of the S. Typhimurium UK-1 strain (15) and furthermore was not as effective as a UK-1-derived strain in ablating colorectal tumors in mice (16). In addition, construction of VPN20009 is based on UV- and Tn10 transposon-induced mutations, which may result in other mutations and over-attenuation (17). It has been shown that the design method causes strain VPN20009 lost chemotactic ability (18). Also, the msbB mutation in VNP20009 is a bad choice because it leads to production of penta- acylated lipid A, which is a good pro-inflammatory stimulator in mice, but is an antagonist to inhibit stimulating human innate immunity (19-22). The second S. Typhimurium strain widely used for cancer therapy is A1-R, which is also derived from ATCC 14028 and is a leu-arg auxotroph (23, 24). Notable, the parent of A1-R (25), A1 is screened through nitrosoguanidine mutagenesis (24). A1-R exhibited good tumor- seeking features and has antitumor efficacy against major types of cancer in mice (24- 26), but no clinical trials in humans or dogs have been performed. The 3rd strain is VXM01, which is based on the S. Typhi strain Ty21a vaccine carrying an eukaryotic expression plasmid for VEGFR2, could induce anti-angiogenic activity when delivered by the oral route in pancreatic cancer. But only 1 of 13 patients showed an improved clinical outcome (27). The 4th strain tested was χ4550 delivering IL-2 to induce responses in dogs and humans, respectively (28-30). All the these strains lack specific tumor targeting ability although VPN20009 and A1-R preferentially colonize tumors. Nevertheless, the results showed their targeting ability is not enough for high efficacy. Salmonella has ability to regress tumors because of its natural toxicity and can also be used as vectors to deliver other anti-cancer molecules including cytotoxic agents such as Cytolysin A (ClyA), FAS ligand (FasL) and TNF-related apoptosis- inducing ligand (TRAIL), cytokines such as IL-2, and tumor antigens such as 3urviving and other factors such as tyrosinase which enhance its anti-cancer effectiveness (see (5, 9). FasL and TRAIL are belonging to the TNFα family. FasL specifically induces apoptosis in cells that possess the FAS receptor and TRAIL is cytotoxic to many cancer cells via death receptor pathways, which activate caspases 8 and 3 (31, 32). ClyA is a bacterial toxin inducing apoptosis and when delivered by S. Typhimurium reduced tumor growth in mice (33, 34). Cytokine IL-2 is widely investigated for its anti-cancer ability because IL-2 can activate the cytolytic function of NK cells and promotes lymphocyte proliferation (35-37). Cytotoxic agents and cytokines can induce apoptosis or stimulate immune cells to directly kill cancer cells, while tumor antigens such as 3urviving function to sensitize the immune system to fight against cancer cells. Survivin is a member of the inhibitor-of-apoptosis protein family involved in regulation of apoptosis and T-cell responses in anti-tumor immunity. It is over-expressed in many tumor cells. Blocking 3urviving function is thus a promising anti-tumor therapeutic method via induction of immune responses against (38-41). In 1981, a patent application was filed on use of attenuated derivatives of pathogenic bacteria to deliver recombinant protective antigens from heterologous pathogens to induce protective immunity to the pathogens whose antigens were delivered by the vaccine construct. Salmonella was the chosen pathogen and it has been continuously improved and perfected as a means for using Salmonella as an antigen and DNA vaccine delivery vector (42, 43). Traditionally, rendering live vaccines safe to be unable to cause adverse effects or disease symptoms has been accompanied with decreased immunogenicity because of the lessened abilities of the attenuated live vaccine to be invasive to colonize lymphoid tissues and/or with reduced abilities to multiply and/or persist to induce an adequate immune response unless administered in multiple doses (44). We have recently invented multiple means to eliminate all these problems that limit live vaccine efficacy by genetically programing the recombinant attenuated Salmonella vaccines (RASVs) to display the same as or even better infection proficiencies than wild-type Salmonella at the time of RASV administration. We thus invented means to increase invasiveness of RASVs (45) and enhance their ability to better survive against host-defense barriers encountered during mucosal delivery (46- 49). These modifications coupled with engineering strains with regulated delayed attenuation (50, 51) and regulated delayed antigen synthesis (52-55) enable the vaccine constructed strains to colonize internal tissues almost to the same extent as wild-type virulent Salmonella but without causing any disease symptoms (50, 51, 53, 56, 57). The RASVs are also designed to persist in these effector lymphoid tissues to serve as factories for the continuous synthesis and delivery of recombinant protective protein antigens (53). These protein antigens are encoded by pathogen genes to induce protective immunity against the pathogen. Alternatively, the recombinant protein might exert a physiological activity altering a host physiological or immunological activity. In either case, the protein is encoded by codon-optimized sequences to enhance mRNA stability and efficiency of transcription and translation in Salmonella (58-60). Since immune responses against recombinant proteins are improved by secretion of antigens rather than their retention in the RASV cytosol (61, 62), we perfected use of type 3 and type 2 secretion systems (T3SS & T2SS) (52, 55, 63) to export proteins out of the RASV or into the periplasmic space to enhance production of outer membrane vesicles that are highly immunogenic (49, 64). In addition, we developed vaccine constructs with regulated delayed lysis in vivo to release in specified cell compartments a bolus of recombinant proteins (45, 52, 65) or a DNA vaccine designed for maximal import to the nucleus for efficient high-level transcription and translation of encoded sequences (45). We observed in multiple recent studies that higher levels of induced protective immunity can be induced by vaccine strains displaying the regulated delayed lysis phenotype than by strains not undergoing lysis (45, 52, 65-68). We have engineered strains to eliminate or decrease synthesis of serotype-specific LPS O-antigen (69, 70) and other immune- dominant surface antigens to reduce inducing immune responses to Salmonella. Nevertheless, prior immunity including maternal immunity (56, 57) enhances success in immunizing neonates and individuals previously immunized with a different strain. We now term these much-improved vaccine vector strains as Protective Immunity Enhanced Salmonella Vaccine (PIESV) vector strains. Based on accumulated results demonstrating complete biological containment and safety of our self-destructing PIESV vectors encoding for delivery of protective antigens from various bacterial, viral and parasite pathogens in newborn, pregnant, malnourished and immune deficient SCID mice, in multiple studies with mice, chickens, pigs and in a human phase 1 trial with no adverse events, bacteremias or shedding in vaccinated human volunteers of viable recombinant vaccine cells in stools over a 12-day period at oral doses of 1010 CFU (56, 57, 71-73), the NIH Office of Science Policy and Recombinant Advisor Committee granted permission to us to evaluate our genetically modified vaccines at Biosafety level 1 containment and under settings simulating commercial rearing for farm animals and in out-patients for human trials. This reclassification was also approved by the University Florida Institutional Biosafety Committee. We also discovered that these PIESV strains are superior adjuvants in recruiting innate immunity. We subsequently have been designing these adjuvant S. Typhimurium UK-1 derived strains as Self-Destructing Attenuated Adjuvant Salmonella (SDAAS) strains to serve as adjuvants to recruit innate immune responses and enhance induction of immunity induced by subunit, killed, live attenuated and live vectored vaccines. Brief Description of Drawings Figure 1. Plasmid maps of the cloning vector pYA3342, the suicide vector pRE112 and the regulated delayed lysis DNA vaccine vector pYA4545. Figure 2. Plasmid maps of the regulated delayed lysis cloning vectors pG8R110 (with T3SS) and pG8R114 (with T2SS) and the plasmid vectors pYA4090 encoding synthesis of GFP and pYA4685 encoding synthesis of EGFP. Figure 3. Plasmid map of pG8R314 encoding OmpA with PLZ4 insert (ompAΩplz4). Figure 4. Plasmid map of pG8R315 as suicide vector for insertion of sequence encoding ompA with PLZ4 insertion (ompAΩplz4) into the S. Typhimurium chromosome. Figure 5. Plasmid maps of pG8R319 derived from pG8R314 by insertion of eukaryotic expression cassette and pG8R320 derived from pYA4545 by insertions of a prokaryotic expression cassette to express sequence encoding ompA with PLZ4. Figure 6. Salmonella with PLZ4 peptide exposed on surface is attracted to and invades into bladder cancer cells. Figure 7. Plasmid maps of pG8R321, pG8R322, pG8R323 and pG8R324 all derived from pG8R319 by insertion of the nucleotide sequences encoding human CXCL11, Mouse CXCL11, KillerRed fused to the neuromodulin N-terminal sequence and KillerRed fused to mitochondrial targeting signals, respectively. Figure 8. Plasmid maps of pG8R325, pG8R326, pG8R327 and pG8R328 all derived from pG8R320 by insertion of the nucleotide sequences encoding human CXCL11, Mouse CXCL11, Killer Red fused to the neuromodulin N-terminal sequence and Killer Red fused to mitochondrial targeting signals, respectively. Figure 9. Plasmid maps of pG8R341 derived from pG8R314 by insertion of sequence encoding GFP3 as an operon fusion and pG8R342 derived from pG8R320 by insertion of sequence encoding EGFP. Figure 10. Plasmid maps of multicistronic pG8R343 and pG8R344 derived from pG8R320 by insertion of sequences encoding KillerRed fused to the neuromodulin N-terminal sequence and human CXCL11 or mouse CXCL11. P2A peptide is used to separate KillerRed and CXCL11 Figure 11. Plasmid maps of pG8R345 derived from pG8R320 by insertion of sequences encoding HLAB leading and tail peptides and pG8R346 derived from pG8R345 by insertion of sequence encoding EGFP. Figure 12. Plasmid maps of pG8R347 , pG8R348, pG8R349 and pG8R350. pG8R347 is derived from pG8R320 by insertion of sequences for 5’ HLA, HLA leading and Tail peptides and 3’ HLA. pG8R348, pG8R349 and pG8R350 are derived from pG8R347 by insertion of sequence encoding EGFP, neo-antigen BBM963 and MB49, respectively. Figure 13. Plasmid pG8R361 derived from pG8R320 by insertion of sequence from PEF1α promoter. Figure 14. Plasmid maps of pG8R362, pG8R363, pG8R364 and pG8R365. pG8R362 and pG8R363 are derived from pG8R320 by insertion of sequence encoding HAC-PD1 fused with human CXCL11 and HAC-PD1 fused with mouse CXCL11, respectively. pG8R364 and pG8R365 are derived from pG8R361 by insertion of sequence encoding HAC-PD1 fused with human CXCL11, HAC-PD1 fused with mouse CXCL11, respectively. Figure 15. Plasmid maps of pG8R366 derived from pG8R320 by insertion of sequence encoding IL2 secretion signal (IL2 SS) and pG8R367 and pG8R368 derived from pG8R366 by insertion of sequence encoding HAC-PD1 and human CXCL11 and HAC- PD1 and mouse CXCL11, respectively. Figure 16. Plasmid maps of pG8R372, pG8R373, pG8R374 and pG8R375 derived from pG8R320 by insertion of sequence encoding IL2 SS fused with HAC-PD1. IL2 SS fused with HAC-PD1 and EGFP, human CXCL11 fused with EGFP, mouse CXCL11 fused with EGFP, respectively. Figure 17. Plasmid maps of pG8R380 derived from pG8R320 by insertion of sequence encoding operon fusion of OmpA with LHRH insertion and GFP and pG8R381 derived from pG8R380 by insertion of OmpA with LHRH insertion, respectively. Figure 18. Plasmid maps of pG8R382, pG8R383 and pG8R384 derived from pG8R320 by insertion of sequence encoding haPD1-IgG, haPD1-IgG and KillerRed fused with neuromodulin N terminal sequence, and KillerRed fused with neuromodulin N terminal sequence and haPD1-IgG, respectively. Figure 19. Plasmid maps of pG8R385 derived from pG8R320 by insertion of sequence encoding operon fusion of OmpA with Her2 scFv insertion and GFP and pG8R386 derived from pG8R385 by insertion of OmpA with Her2 scFv insertion, respectively. Figure 20. Plasmid maps of pG8R388 and pG8R389 derived from pG8R320 by insertion of sequence for Ptrc promoter and Ptrc promoter and optimized Bla secretion signal. Figure 21. Plasmid maps of pG8R390, pG8R391, and pG8R418. pG8R390 is derived from pG8R381 by insertion of sequence encoding KillerRed fused with neuromodulin N terminal sequence and pG8R391 is derived from pG8R386 by insertion of sequence encoding KillerRed fused with neuromodulin N terminal sequence. pG8R418 is derived from pG8R385 by insertion of sequence encoding KillerRed fused with neuromodulin N terminal sequence. Figure 22. The attachment and invasion of bladder cancer cells with strains derived from χ12614 with O-antigen mutations. (A) Genotypic characterization of strains using primers specific for waaL, waaG, waaC, ompA and ompAΩplz4. Lane 1, χ3761; lane 2, χ12614; lane 3, χ12812; lane 4, χ12813; lane 5, χ12814. (B) LPS gel profile of strains. All strains are grown in LB media. (C) The percentage of attachment and invasion of strains in human bladder cancer cell line 5637 and mouse bladder cancer cell line MB49. All strains carry plasmid pG8R314 with multiple copies of ompAΩplz4. ****, P<0.0001, compared with other strains. Figure 23. The attachment and invasion of bladder cancer cells with strains derived from χ12619 with O-antigen mutations. All strains have only one copy of ompAΩplz4 in their chromosome. (A) Genotypic characterization of strains using primers specific for asdA, pagP, pagL, lpxR, waaL, waaG, waaC, ompA and ompAΩplz4. Lane 1, χ3761; lane 2, χ12518; lane 3, χ12619; lane 4, χ12808; lane 5, χ12809; lane 4, χ12810; lane 5, χ12811. (B) The growth of strains on LB, LB+ arabinose (0.1%), LB+DAP (50ug/ml), LB+D-alanine (50 ug/ml) plates. (C) LPS gel profile of the strains. All strains are grown in LB media with 0.1% arabinose. (D) The percentage of attachment and invasion of strains in human bladder cancer cell line 5637 and mouse bladder cancer cell line MB49. *, P<0.05, **, P<0.01 , *** P<0.001, ****, P<0.0001, compared with other LPS mutation strains Figure 24. Display of OmpAΩHer2 ScFV on the bacterial surface. Salmonella outer membrane proteins (OMPs) were isolated and subjected to SDS-PAGE gel and western blot with anti-His antibody. 1, χ12417; 2, χ12417(pG8R385); 3, χ12417(pG8R418); 4, χ12417(pG8R391). Figure 25. Strains carrying ompAΩher2 ScFV display higher attachment and invasion to Her2 over expression cell SKRB-3. ****, P<0.0001. Figure 26. Production of KillerRed in HEK293T cells transfected with plasmid pG8R327. The frames show the KillerRed red fluorescent signals from four different samples (EVOS FL, RFP channel) Figure 27. KillerRed kills HEK293T cell. Time zero is set immediately after irradiation with green light. Time 10 is set immediately after excitation for 10 min (EVOS FL, RFP channel) Figure 28. KillerRed kills HEK293T cell excitation for 10 and 20 minutes. Time zero is set immediately after irradiation with green light. Time 10 and 20 are set immediately after excitation for 10 min or 20 mins (EVOS FL, RFP channel) Definitions As used herein the specification, “a” or “an” may mean one or more, unless clearly indicated otherwise. As used herein in the claims, when used in conjunction with the word “comprising,” the words “a” or “an” may mean one or more than one. The term “administering” or “administration” of an agent as used herein means providing the agent to a subject using any of the various methods or delivery systems for administering agents or pharmaceutical compositions known to those skilled in the art. Agents described herein may be administered by oral, intradermal, intravenous, intramuscular, intraocular, intranasal, intrapulmonary, epidermal, subcutaneous, mucosal, or transcutaneous administration. The terms “animal host” or “subject” as used interchangeably hereinto refer to a human or nonhuman mammal or a vertebrate animal into which a genetically modified Salmonella cell has been administered. In a specific embodiment, the subject is a human. The terms “attenuated” or “attenuation” as used herein refer to the process of rendering certain pathogen virulence attributes needed to cause diseases less able to cause such disease symptoms. In one example, attenuation involves imparting an attenuation mutation in the pathogen. The term “attenuating mutation” refers to a mutation imparted into a pathogen that reduces infectivity, virulence, toxicity, induction of disease symptoms, and/or impairment of a subject upon administration of the pathogen (e.g. PIESV strain). Examples of attenuating mutations include those mutations that facilitate lysis in vivo (e.g. impairing synthesis of essential constituents of peptidoglycan layer), reduce or impair synthesis of LPS or other cell-surface components, and one or more mutations that provide auxotrophy (e.g. dependence on an amino acid, purine, pyrimidine, or vitamin for growth). The term “balanced-lethal vector-host” refers to a host Salmonella cell into which a plasmid vector has been introduced such that survival of the host cell is dependent on the maintenance of the plasmid vector and loss of the plasmid vector results in death of the host Salmonella cell. (See Nakayama, K., Kelly, S. & Curtiss, R. Construction of an ASD+ Expression-Cloning Vector: Stable Maintenance and High Level Expression of Cloned Genes in a Salmonella Vaccine Strain. Nat Biotechnol 6, 693–697 (1988) or Galán JE, Nakayama K, Curtiss R 3rd. Cloning and characterization of the asd gene of Salmonella typhimurium: use in stable maintenance of recombinant plasmids in Salmonella vaccine strains. Gene.1990 Sep 28;94(1):29-35, whose teachings are incorporated by reference). The term “biologically active fragment” or “biologically active variant” refers to a fragment or variant of a sequence that maintains its biological activity. In the context of H. pylori antigen sequences, a biologically active fragment or biologically active variant is a fragment or variant of an antigen amino acid sequence that elicits an immune response in a host. The term “Cancer Cell Targeting Salmonella strain” or “CCTS strain” refers to a strain of Salmonella that has one or more attenuating mutations and expresses a gene product that causes selective localization and/or internalization of cells of the CCTS strain by a cancer cell. As used herein, “codon” means, interchangeably, (i) a triplet of ribonucleotides in an mRNA which is translated into an amino acid in a polypeptide or a code for initiation or termination of translation, or (ii) a triplet of deoxyribonucleotides in a gene whose complementary triplet is transcribed into a triplet of ribonucleotides in an mRNA which, in turn, is translated into an amino acid in a polypeptide or a code for initiation or termination of translation. Thus, for example, 5’-TCC-3’ and 5’-UCC-3’ are both “codons” for serine, as the term “codon” is used herein. The term “codon optimized” or “codon optimization” as used herein refers to enhancing the ability of the antigen encoding sequence to be expressed in the Salmonella strain by selecting codons that are used for highly expressed genes in Salmonella. Such codon optimization also includes changing the GC content of the antigen encoding sequence to be similar to that used for Salmonella (i.e., ~52% GC). In addition, the codon optimization can also be used to enhance the stability of the mRNA encoded by the antigen encoding sequence so as to be less likely to be degraded by Rnases. The term “delayed attenuation” as used herein refers to a means of gene regulation such that the attenuation attribute is not expressed during growth of the vaccine strain or during its administration to an animal host but is not expressed after the CCTS strain enters the animal host and is manifest as a consequence of vaccine cell division in vivo with gradual dilution of the virulence gene product by at least half at each cell division in vivo. The term “gene product” refers to a transcript (RNA) or expressed polypeptide encoded by a heterologous gene or nucleic acid that has been introduced into a genetically modified Salmonella cell. In typical embodiments, the gene product causes selected localization to a target cell. The gene product may also cause cytotoxicity to the target cell upon internalization of the genetically modified Salmonella cell and/or cause a targeted immune response to target cells. A "genetically modified Salmonella cell” or “GMSC” refers to a Salmonella cell that comprises an attenuating mutation and/or into which a heterologous gene or nucleic acid, e.g., an exogenous nucleic acid that is foreign to the Salmonella cell, has been introduced. The term “operably linked” as used herein means that one nucleic acid sequence is linked to another nucleic acid sequence, and therefore the function or expression thereof is influenced by the linked nucleic acid sequence. As used herein, the term “percentage of sequence identity” or “percent sequence identity” may refer to the value determined by comparing two optimally aligned sequences (e.g., nucleic acid sequences or polypeptide sequences) of a molecule over a comparison window, wherein the portion of the sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleotide or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the comparison window, and multiplying the result by 100 to yield the percentage of sequence identity. A sequence that is identical at every position in comparison to a reference sequence is said to be 100% identical to the reference sequence, and vice-versa. The term “about” with respect to a numerical value of a sequence length means the stated value with a +/- variance of up to 1-5 percent. For example, about 30 contiguous nucleotides means a range of 27-33 contiguous nucleotides, or any range in between. The term “about” with respect to a numerical value of percentage of sequence identity means the stated percentage value with a +/- variance of up to 1-3 percent rounded to the nearest integer. For example, about 90% sequence identity means a range of 87-93%. However, the percentage of sequence identity cannot exceed 100 percent. Thus, about 98% sequence identity means a range of 95-100%. The term “regulated delayed lysis” refers to a construction in which the expression of one or more genes specifying synthesis of peptidoglycan precursors such as but not limited to diaminopimelic acid and muramic acid are regulated by a sugar- dependent process such that the genes are expressed in the presence of a sugar such as but not limited to arabinose or rhamnose supplied during cultivation of the strain and cease to be expressed in vivo since the sugar is absent to result in lysis as a consequence of cell division of the CCTS strain in vivo. The genes conferring the regulated delayed lysis phenotype may be either chromosomal and/or plasmid encoded. The term “regulated delayed lysis plasmid” refers to a construction in which the expression of one or more genes specifying synthesis of peptidoglycan precursors such as but not limited to diaminopimelic acid and muramic acid that are regulated by a sugar-dependent process are located on a plasmid vector encoding synthesis of one or more foreign antigens or gene products. The term “Salmonella cell” refers to a cell of a Salmonella species or serotype. Examples of a Salmonella serotype include Salmonella Typhimurium and Salmonella Enteritidis. In a more specific embodiment, the Salmonella serotype is S. Typhimurium UK-1. The term “sequence identity” or “identity,” as used herein in the context of two polynucleotides or polypeptides, refers to the residues in the sequences of the two molecules that are the same when aligned for maximum correspondence over a specified comparison window. As used herein, the term “targeted immune response” refers to a response by a subject’s immune system against target cells. Immune responses include both cell- mediated immune responses (responses mediated by antigen-specific T cells and non- specific cells of the immune system) and humoral immune responses (responses mediated by antibodies present in the plasma lymph, and tissue fluids and secreted onto mucosal surfaces). The term “target cell” refers to a cell of a subject that is of a type to which a genetically modified Salmonella cell is designed for selective localization and/or internalization. Selective localization refers to increased migration of the genetically modified Salmonella cell to a target cell over other cells in a subject. Selective internalization refers to increased internalization of the genetically modified Salmonella cell in the target cell over other cells in the subject. Increased localization to and/or increased internalization means an increase of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70% or more respective to target cells as opposed to other cells in a subject. Typically, a target cell internalizes the genetically modified Salmonella cell by active invasion or endocytosis or phagocytosis. In exemplified embodiments, the target cell is a cancer cell and the genetically modified Salmonella cell is of a CCTS strain that is selectively internalized by the cancer cell over other cells in the subject. In alternative embodiments, the genetically modified Salmonella cell is engineered to localize at a tumor microenvironment where cancer cells are present without necessarily being internalized into a cancer cell. The term “variant” as used herein refers to a nucleic acid sequence or amino acid sequence that possesses at least about 85, 90, 95, 96, 97, 98 or 99 percent sequence identity to another nucleic acid sequence or amino acid sequence, respectively. Other relevant definitions are provided infra. Description Disclosed herein are embodiments directed to designing, constructing and evaluating Cancer Cell Targeting Salmonella (CCTS) strains. CCTS strain embodiments have (i) ability to directly destroy tumor cells, (ii) deliver cargoes that cause tumor cells to self-destruct, (iii) deliver cargoes that enhance abilities to treat tumor cells, and/or (iv) directly and/or indirectly stimulate host immune responses to repress tumor cell growth, metastases and cell death. A potentially desirable feature involves rapid self-destruction of CCTS cells that enables their use for repeat treatments of subjects. A unique attribute of these newly designed and constructed CCTS strains is their ability to simultaneously synthesize and deliver protein cargoes to cancer cells but to also deliver DNA vaccines encoding other effective proteins to be synthesized by the tumor cells to their detriment. The foregoing attributes are achieved by introducing numerous deletion and deletion-insertion mutations to enable and endow the desired phenotypic properties to the strains constructed. These mutations and their associated phenotypes are listed in Table 1 and the suicide vectors needed for their insertion into plasmids and the S. Typhimurium chromosome are listed in Table 2. The distribution of genetic deletion and deletion-insertion mutations and the redundancy in critical modifications ensure both stability and safety of these CCTS strains. Examples of genotypes of CCTS strains are listed in Table 3. Table 1. Mutations and associated phenotypes in S. Typhimurium CCTS strainsa It is noted that the genes can be inactivated or deleted in multiple ways to confer the same phenotypic traits. Also, though certain allele numbers are indicated elsewhere herein for certain mutations, reference to a certain allele is not limiting and the mutations can be executed in other alleles. Genotype Phenotype ∆aroA encodes the first enzyme in the pathway to synthesize aromatic amino acids and derived vitamins (74) ∆asdA deletes gene for aspartate semialdehyde dehydrogenase essential for synthesis of diaminopimelic acid (DAP) necessary for peptidoglycan synthesis (75) ∆PasdA::TT araC ParaBAD asdA makes synthesis of AsdA dependent on presence of arabinose ∆PasdA::TT rhaRS PrhaBAD asdA makes synthesis of AsdA dependent on presence of rhamnose ∆asdA::TT araC ParaBAD c2 inactivates asdA and makes synthesis of C2 repressor dependent on arabinose (76, 77) ∆alr and ∆dadB deletes the genes for two alanine racemases essential for synthesis of D-alanine necessary for peptidoglycan synthesis (78) ∆PdadB::TT araC ParaBAD dadB makes synthesis of DadB dependent on presence of arabinose ∆PdadB::TT rhaRS PrhaBAD dadB makes synthesis of DadB dependent on presence of rhamnose ∆PmurA::TT araC ParaBAD murA makes synthesis of MurA, the first enzyme in the synthesis of muramic acid, dependent on arabinose in growth medium and ceases synthesis in vivo due to absence of arabinose (50, 65) ∆Pfur::TT araC ParaBAD fur makes synthesis of the Fur repressor protein dependent on arabinose in growth medium that ceases in vivo to result in high-level synthesis of all iron regulated proteins to result in attenuation (50, 79) ∆mntR eliminates gene for repressor MntR that regulates MntR- and some Fur-regulated genes for manganese and iron acquisition, respectively ∆PmntR::TT araC ParaBAD mntR makes synthesis of the MntR repressor protein dependent on arabinose in growth medium that ceases in vivo to result in high-level synthesis of all manganese regulated proteins to contribute to attenuation ∆cya encodes enzyme for adenylate cyclase ∆crp encodes adenylate cyclase catabolite represor protein ∆araBAD::TT deletion of genes to eliminate arabinose catabolism with TT inserted to prevent transcription of downstream genes (80-84) ∆araCBAD100::TT Deletion of all genes in the ara operon ∆rhaBADSR deletion of genes to eliminate rhamnose catabolism (85, 86) ∆pagP::Plpp lpxE mutation causes regulated delayed in vivo synthesis of the codon- optimized lpxE gene from Francisella tularensis to cause synthesis of the non-toxic adjuvant form of LPS lipid A lipid A (MPLA) (66) ∆lpxR::Plpp lpxF mutation causes regulated delayed in vivo synthesis of the codon- optimized lpxF gene from Francisella tularensis to cause synthesis of LPS with only the 1’-phosphoryl group (21) ∆pagL and ∆lpxR eliminates two means by which Salmonella alters LPS components in vivo to decrease recruitment of innate immunity by interaction with TLR4 (20) ∆eptA prevents addition of ethanolamine to lipid A (87, 88) ∆arnT prevents addition of 4-amino-4-deoxy-L-arabinose (L-Ara4N) groups to lipid A (89) ∆fliC deletes gene specifying synthesis of the phase I flagellin FliC (79, 90-92) ∆fljB deletes gene specifying synthesis of the phase II flagellin FljB (79, 90-92) ∆fliC180 specifies a truncated FliC protein containing TLR5 recognition domain and CD4 epitope (93) ∆(hin-fljBA) locks in expression of gene for phase I FliC flagellin and precludes synthesis of phase II FljB flagellin (94-100) ∆(agfG-agfC) deletes two operons specifying thin aggregative fimbriae (curli) and an activator for synthesis and export of cellulose and other exopolysaccharides (101) ∆Psaf5::PmurA safA causes constitutive synthesis of Saf fimbriae that facilitate spleen colonization (102) ∆Pstc::PmurA stcA causes constitutive synthesis of Stc fimbriae that facilitate spleen colonization (102) ∆fimH encodes the adhesin tip on Type 1 fimbriae (103, 104) ∆ompA specifies synthesis of a very prevalent outer membrane protein (105) ∆sopB a protein secreted by the Salmonella SPI-I that can cause intestinal inflammation (106-108) ∆pabA & ∆pabB Encode two enzyme subunits of the enzyme synthesizing p- amino benzoic acid (109, 110) ∆pmi eliminates phosphomannose isomerase that precludes synthesis of GDP-mannose that is needed for LPS O-antigen synthesis (70, 79) ΔwaaL & ∆pagL::TT araC ParaBAD waaL (or ∆pagL::TT rhaRS PrhaBAD waaL) make synthesis of the WaaL enzyme that couples O-antigen to the LPS core synthesis (22) dependent on presence of arabinose (or rhamnose) ΔwbaP encodes enzyme that couples LPS core to LPS O-antigen (22, 111-114) ΔwaaC encodes enzyme necessary for assembly of the LPS inner core (111-114) ΔwaaG encodes enzymes essential fir assembly of the outer LPS core (22, 111-114) ∆(wza-wcaM) eliminates 20 genes encoding enzymes needed for synthesis of colanic acid, LPS capsular antigen and other polysaccharides to facilitate lysis, enhance immunogenicity and inhibit biofilm formation (115, 116) ∆relA uncouples growth regulation from a dependence on protein synthesis (117, 118) ∆relA::araC PBAD lacI TT and ∆(traM-traX)::araC ParaBAD lacI makes synthesis of LacI that represses gene expression controlled by Ptrc dependent on presence of arabinose (53, 119) with either inactivation of relA gene (75,76) or deletion of genes encoding conjugational plasmid transfer in Salmonella virulence plasmid (120) ∆spoT eliminates gene for synthesis of ppGpp (117, 121, 122) ∆spvRABCD deletes Salmonella plasmid virulence genes encoding regulatory activator-repressor (R) and four genes conferring invasiveness and virulence; when the spvABCD genes are over expressed increase invasiveness and virulence ∆cysG175::Pspv spvABCD inserts spv operon without the R gene specifying the repressor-activator into a deletion of the cysG gene under control of a promoter not regulated by SpvR ∆PhilA::Ptra∆lacO hilA constitutive Ptrc regulated synthesis of HilA that increases expression of SPI-1 genes for invasion of epithelial cells (45) ∆recF reduces inter- and intra-plasmidic recombination (78, 123-125) ∆endA deletes gene encoding endonuclease I to prevent degradation of released DNA vaccine (82) ∆sifA enables Salmonella to escape from the SCV to enter the cytosol (126, 127) ∆sseL eliminates a gene that enables Salmonella to induce pyroptosis (82) ∆tlpA eliminates a gene that enables Salmonella to induce pyroptosis (82) a ∆ = deletion; TT = transcription terminator; P = promoter Table 2. Suicide vectors for constructing the mutations in Table 1 Genotype Suicide Vector Marker A. Deletion and deletion-insertion mutations to facilitate regulated delayed lysis in vivo ∆PmurA25::TT araC ParaBAD murA pYA4686 Cm ∆asdA33 pYA3736 Cm ∆PasdA55::TT araC ParaBAD asdA pG8R71 Cm ∆PasdA88::TT rhaRS PrhaBAD1 asdA pG8R354 Cm ∆alr-3 pYA3667 Cm ∆dadB4 pYA3668 Cm ∆PdadB66::TT araC ParaBAD dadB pG8R73 Cm ∆PdadB22::TT rhaRS PrhaBAD1 dadB pG8R352 Cm ∆(wza-wcaM)-8 pYA4368 Cm ∆relA1123 pYA3679 Cm B. Mutations enabling regulation of genes that might be present on plasmid vectors in conjunction with strains undergoing regulated delayed lysis in vivo ∆relA197::araC ParaBAD lacI TT pYA4064 Cm ∆asdA27::TT araC ParaBAD c2 pYA4138 Cm ∆(traM-traX)-36::araC ParaBAD lacI TT pG8R329 Cm ∆(traM-traX)-41::araC ParaBAD lacI TT pG8R397 Cm C. Mutations conferring attenuation of virulence ∆aroA21419 pYA3600 Cm ∆cya-27 pMEG080 Tet ∆crp-27 pMEG084 Tet ∆pabA1516 pMEG147 Tet ∆pabB232 pYA3438 Cm D. Mutations conferring regulated delayed attenuation and over production of iron and manganese-regulated proteins to confer cross-protective immunity ∆Pfur33::TT araC ParaBAD fur pYA3722 Cm ∆PmntR44::TT araC ParaBAD mntR pG8R227 Cm E. Mutations altering synthesis of LPS components ∆pmi-2426 pYA3546 Tet ∆pagP8 pYA4288 Cm ∆pagP81::Plpp lpxE pYA4295 Cm ∆pagL7 pYA4284 Cm ∆lpxR9 pYA4287 Cm ∆lpxR93::Plpp lpxF pYA4289 Cm ∆arnT6 pYA4286 Cm ∆eptA4 pYA4283 Cm ∆waaC41 pYA5473 Cm ∆waaG42 pYA4896 Cm ∆waaL46 pYA4900 Cm ∆wbaP45 pYA4899 Cm ∆pagL19::TT araC ParaBAD1 waaL pYA5468 Cm ∆pagL64::TT rhaRS PrhaBAD1 waaL1 pYA5377 Cm ∆pagL38::TT rhaRS PrhaBAD1 waaL2 pG8R296 Cm ∆pagL18::TT araC ParaBAD1 waaC pYA5458 Cm ∆pagL21::TT araC ParaBAD1 waaG pYA5462 Cm F. Mutations blocking catabolism of sugars ∆araBAD65::TT pYA4811 Cm ∆rhaBADSR515 pG8R272 Cm ∆araCBAD100::TT pG8R392 Cm G. Mutations altering synthesis of flagellar components ∆fliC180 pYA3729 Cm ∆fliC2426 pYA3702 Cm ∆fljB217 pYA3548 Tet ∆(hin-fljBA)-209 pG8R306 Cm H. Mutations altering synthesis of fimbrial components ∆(agfG-agfC)-999 pYA4941 Cm ∆Pstc53::PmurA stcA53 pYA5053 Cm ∆stcABCD pYA5007 Tet ∆Psaf55::PmurA safA55 pYA5055 Cm ∆safABCD pYA4586 Tet ∆fimH1019 pYA3545 Tet I. Mutations eliminating or altering outer membrane proteins ∆ompA11 pYA4757 Tet ompA3Ωplz4 pG8R315 Cm J. Mutations decreasing inflammation and enhancing mucosal immunity ∆sopB1925 pYA3733 Cm K. Mutations eliminating or diminishing effective immunogenicity ∆sifA26 pYA3716 Cm L. Mutations decreasing/delaying onset of pyroptosis ∆sseL116 pYA4621 Cm ∆tlpA181 pYA4620 Cm M. Mutations leading to degradation of DNA within Salmonella cells ∆recA62 pYA4680 Cm ∆recF126 pYA3886 Cm ∆endA2311 pYA3652 Cm N. Mutations altering invasion ∆PhilA::Ptra∆lacO hilA pYA4681 Cm a∆ = deletion; TT = transcription terminator; P = promoter Table 3. Genotypes of CCTS strains generated that have been used in past research on anti-cancer therapies. Strain genotype Refs χ4550 ∆cya-1 ∆crp-1 ∆asdA1 (∆zhf-4::Tn10) (from χ4064 STm SR-11) (30, 35, 128- 130) χ8133 ∆cya-27 ∆crp-27 ∆asdA16 (63) χ11091 ∆pabA1516 ∆pabB232 ∆asdA16 ∆msbB48 ∆pagL7 ∆pagP81::Plpp (20) lpxE ∆lpxR93::Plpp lpxF χ12342 ∆waaG42 ∆pagL21::TT araC ParaBAD waaG ∆lpxR9 ∆pagP8 (also (16) with (16) ∆aroA21419) BCT2 ∆pabA1516 ∆pabB232 ∆asdA16 ∆msbB48 ∆pagL7 ∆pagP81::Plpp (131) lpxE ∆lpxR93::Plpp lpxF ∆fimH ∆fliC ∆fljB ∆rfaL (=waaL) ∆pgtEp (from χ11091) Treatment Methods The genetically modified Salmonella cells described herein and therapeutic compositions comprising the same may be used in methods to treat cancer, to attenuate the growth of a tumor or to regress a tumor. The methods described herein may be used to treat or attenuate the growth of any cancer or tumor type. Cancers and tumor types that may be treated or attenuated using the methods described herein include but are not limited to bone cancer, bladder cancer, brain cancer, breast cancer, cancer of the urinary tract, carcinoma, cervical cancer, colon cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, liver cancer, lung cancer, lymphoma and leukemia, melanoma, ovarian cancer, pancreatic cancer, pituitary cancer, prostate cancer, rectal cancer, renal cancer, sarcoma, testicular cancer, thyroid cancer, and uterine cancer. In addition, the methods may be used to treat tumors that are malignant (e.g., primary or metastatic cancers) or benign (e.g., hyperplasia, cyst, pseudocyst, hematoma, and benign neoplasm). In some embodiments, a method for treating cancer may include administering a therapeutically effective amount of genetically modified Salmonella cells described herein or therapeutic compositions comprising the same to a subject who has cancer. “Treating” or “treatment” of a condition may refer to preventing the condition, slowing the onset or rate of development of the condition, reducing the risk of developing the condition, preventing or delaying the development of symptoms associated with the condition, reducing or ending symptoms associated with the condition, generating a complete or partial regression of the condition, or some combination thereof. A “therapeutically effective amount,” “effective amount” or “effective dose” is an amount of a composition (e.g., a therapeutic composition or cells) that produces a desired therapeutic effect in a subject, such as preventing or treating a target condition or alleviating symptoms associated with the condition. The precise therapeutically effective amount is an amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given subject. This amount will vary depending upon a variety of factors, including but not limited to the characteristics of the therapeutic compound (including activity, pharmacokinetics, pharmacodynamics, and bioavailability), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), the nature of the pharmaceutically acceptable carrier or carriers in the formulation, and the route of administration. One skilled in the clinical and pharmacological arts will be able to determine a therapeutically effective amount through routine experimentation, namely by monitoring a subject's response to administration of a compound and adjusting the dosage accordingly. For additional guidance, see Remington: The Science and Practice of Pharmacy 21st Edition, Univ. of Sciences in Philadelphia (USIP), Lippincott Williams & Wilkins, Philadelphia, Pa., 2005. The therapeutic compositions described herein may be administered by any suitable route of administration. A “route of administration” may refer to any administration pathway known in the art, including but not limited to aerosol, enteral, nasal, ophthalmic, oral, parenteral, rectal, transdermal (e.g., topical cream or ointment, patch), or vaginal. “Parenteral” refers to a route of administration that is generally associated with injection, including infraorbital, infusion, intraarterial, intracapsular, intracardiac, intradermal, intramuscular, intraperitoneal, intrapulmonary, intraspinal, intrasternal, intrathecal, intrauterine, intravenous, subarachnoid, subcapsular, subcutaneous, transmucosal, or transtracheal. In one embodiment, the tumor antigen vaccines described herein (e.g., an SVN or CO-SVN Salmonella-based vaccine and associated expression plasmids) are administered orally and the compositions that disrupt tumor-derived immune suppression described herein (e.g., YS1646-shSTAT3 Y51646-shIDO1, YS1646-shArg1 or YS1646-shiNOS) are administered intravenously. Examples Example 1. Materials and Methods. a. Bacterial strains, media and bacterial growth. All CCTS strains are derived from the highly virulent S. Typhimurium UK-1 strain (132) since attenuated S. Typhimurium UK-1 strains will induce protective immunity to challenge with all S. Typhimurium strains whereas other S. Typhimurium strains attenuated with the same mutations often cannot induce protective immunity to some S. Typhimurium strains and definitely not to virulent UK-1 (133, 134). LB broth and agar (135) and Purple broth (PB) (Difco), which is devoid of arabinose (Ara), mannose (Man) and rhamnose (Rha), are used as complex media for propagation, phenotypic analyses and plating. MacConkey agar with 0.5% lactose (Lac) and 0.1% Ara, 0.1% rhamnose and 0.1% mannose (if needed) are used to enumerate bacteria recovered from mice or other animals. Bacterial growth is monitored spectrophotometrically and by plating for colony counts. b. Molecular and genetic procedures. Methods for DNA isolation, restriction enzyme digestion, DNA cloning and use of PCR for construction and verification of bacterial strains and vectors are standard (136). DNA sequence analyses are performed commercially. All oligonucleotide and/or gene syntheses are done commercially with codon optimization to enhance translational efficiency in humans or Salmonella and stabilize mRNA to “destroy” RNase E cleavage sites (59, 60) to prolong mRNA half-life. Plasmids are evaluated by DNA sequencing and ability to specify synthesis of proteins using gel electrophoresis and western blot analyses. Expression of sequences encoded in DNA vaccine vectors is monitored after electroporation into Vero cells and using antibodies specific to DNA vaccine encoded proteins. Methods for generating mutant strains are described in previous publications (137-145) and in Examples below using the suicide vector delivery strain χ7213 (thi-1 thr-1 leuB6 glnV44 fhuA21 lacY1 recA1 RP4-2-Tc::Mu λpir ∆asdA4 ∆zhf-2::Tn10). Recombinant plasmid constructs are transformed into E. coli χ6212 (F λ φ80 Δ(lacZYA-argF) endA1 recA1 hsdR17 deoR thi-1 glnV44 gyrA96 relA1 ΔasdA4) with selection for AsdA+ for initial characterization prior to electroporation into CCTS strains. c. Selection of targeting and effector proteins. Selection of proteins that facilitate targeting to cancer cells or constitute cargo proteins with desired biological effects to be encoded on regulated lysis plasmid vectors for synthesis and delivery by CCTS strains or to be encoded on DNA vaccine vectors for expression in the inoculated animal host are based on prior discoveries and evidence of well-established activities in the published literature. d. CCTS strain characterization. CCTS constructs are evaluated in comparison with vector-control strains for stability of plasmid maintenance, integrity and protein synthesis ability when CCTSs are grown in the presence of arabinose and DAP and with and without IPTG for 50 generations. The IPTG dependence of protein synthesis to overcome the LacI repression of the Ptrc promoter is also verified. IPTG- induced cultures are incubated with chloramphenicol to arrest protein synthesis to determine whether plasmid-specified proteins are stable during the next 4 h. If not, the nucleotide sequence is altered to eliminate protease cleavage sites (with subsequent comparison of both constructs for induction of immune responses). Measurement of LPS core and O-antigen is performed after electrophoresis using silver-stained gels (146). Final CCTS constructs are evaluated for bile sensitivity, acid tolerance and ability to survive in sera with and without complement (143-145) and for sensitivity to antibiotics used to treat Salmonella infections. e. Cell culture methods. Some tumors are caused by cancer cells with specific targetable receptors or that possess phenotypic properties that can be used to attract specially designed CCTS strains with specific targeting attributes. For example, bladder tumor cells uniquely display a receptor that can bind to a targeting peptide termed PLZ4 (amino acid sequence: CQDGRMGFC) that is absent on normal uroepithelial cells and other cell types throughout the body. Nanoparticles coated with PLZ4 specifically target bladder tumor cells but not to other cancer cell types (147-150) . This targeting is observed for bladder tumor cells from mice, dogs and humans (151). CCTS strains displaying PLZ4 can be evaluated by their differential ability to attach to and invade the bladder tumor cell lines 5637, TCCSUP, and T24 (151) . Methods for evaluating the abilities of Salmonella cells to attach to, invade into and survive in cells in culture are well established (152). These methods can be modified as needed for CCTS strains targeting other tumor cell types. f. Cell imaging. Some plasmids have genes encoding fluorescent proteins enabling synthesis of GFP in Salmonella or EGFP or mCherry in animal cells. The fluorescent protein in bacteria or cells will be visualized using the EVOS Automated Cell Imaging System (ThermoFisher Scientific). The Cell Plasma Membrane Staining Kit - Orange Fluorescence - Cytopainter (ab219941, Abcam) was used to label cell membranes. The acquired image was processed using ImageJ software (153). Example 2. Construction of mutant S. Typhimurium strains with deletions of the ompA gene to enable display of altered OmpA proteins with inserted peptides enabling targeting to specific tumor cells. Pan and associates have defined a nine amino acid peptide CQDGRMGFC termed PLZ4 (US patent 10,335,365) that targets a specific receptor present on bladder tumor cells (151). A number of S. Typhimurium strains with anti-tumor attributes have been constructed to display PLZ4 to preferentially and specifically target bladder tumor cells. The objective was to insert the sequence for PLZ4 into one of the exposed outer loops of the OmpA protein. The OmpA protein was selected since it is the most abundant OMP in the Salmonella outer membrane (105) and could be specified on a plasmid replicon to increase its relative quantity in relation to other OMPs. To construct a strain to test the validity and feasibility of our approach, we generated a derivative of χ12341 to insert the ∆ompA11 deletion mutation using the suicide vector pYA4757 (Table 2) to yield the strain χ12417 (Table 4). The ∆ompA11 mutation deletes the entire ompA open reading frame including the start to stop codon sequence. χ12341 (73, 154) was selected since its viability and virulence are dependent on the supply of three sugars that can be supplied during culture but that are totally absent in animal tissues and since it cannot synthesize LPS O-antigen in vivo thus exposing the outer membrane proteins to enable better and more efficient interactions with eukaryotic cell surfaces in the in vivo environment. After demonstrating that χ12417 harboring a multi-copy plasmid encoding the ompAΩplz4 fusion could adhere to bladder tumor cells displaying the receptor for PLZ4 (see below), studies were commenced to evaluate S. Typhimurium strains with a diversity of properties for use with a diversity on new plasmid vectors encoding for synthesis of attributes that contribute to tumor therapy, tumor cell destruction and/or to recruit host immunity to target tumor antigens, etc., in addition to tumor cell adherence. All these S. Typhimurium strains listed in Table 4 were constructed using the suicide vectors listed in Table 2 to introduce the mutations described in Table 1. Many were derived from Protective Immunity Enhanced Salmonella Vaccine (PIESV) strains of Self-Destructing Attenuated Adjuvant Salmonella (SDAAS) strains that have been described (PCT/US21/61814 and WO 2021/222696 A1, respectively), which are incorporated herein in their entirety. Table 4. S. Typhimurium strains constructed and evaluated as CCTS strains. χ12414 ∆waaG42 ∆pagL21::TT araC ParaBAD waaG ∆lpxR9 ∆pagP8 ∆eptA4 ∆arnT6 ∆pmi-2426 ∆relA197::araC ParaBAD lacI TT ∆ompA11 χ12417 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆pmi-2426 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆relA197::araC PBAD lacI TT ∆recF126 ∆sifA26 ∆ompA11 (from χ12341) χ12447 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆pmi-2426 ∆(wza- wcaM)-8 ∆recF126 ∆sifA26 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆endA2113 ∆relA1123 ∆sseL116 ∆tlpA181 ∆ompA11 (from χ12388) χ12452 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆pmi- 2426 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆relA197::araC PBAD lacI TT ∆recF126 ∆sifA26 ∆ompA11 ∆sopB1925 (from χ12417) χ12485 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆pmi-2426 ∆Pfur33::TT araC ParaBAD fur ∆asdA33 ∆relA197::araC PBAD lacI TT ∆(wza-wcaM)-8 ∆PtolR67::::TT araC ParaBAD tolR ∆ompA11 (from χ12473) χ12494 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆pmi-2426 ∆(wza- wcaM)-8 ∆recF126 ∆sifA26 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆endA2113 ∆relA1123 ∆sseL116 ∆tlpA181 ∆ompA11 ∆sopB1925 (from χ12447) χ12508 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆pmi-2426 ∆(wza- wcaM)-8 ∆relA197::araC ParaBAD lacI TT ∆recF126 ∆sifA26 ∆wbaP45 ∆pagL14::TT araC ParaBAD wbaP ∆lpxR9 ∆pagP8 ∆ompA11 (from χ12449) χ12529 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆pmi-2426 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD1 waaL1 ∆(wza-wcaM)-8 ∆relA197::araC ParaBAD lacI TT ∆recF126 ∆sifA26 ∆ompA11 ∆araBAD65::TT ∆rhaBADSR515 (from χ12425) χ12614 ∆asdA33 ∆ompA11 (from χ8958) χ12627 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆pmi-2426 ∆Pfur33::TT araC ParaBAD fur ∆asdA33 ∆relA197::araC PBAD lacI TT ∆(wza-wcaM)-8 ∆PtolR67::::TT araC ParaBAD tolR ∆ompA11 ∆pagP81::Plpp lpxE (from χ12485) χ12628 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆pmi-2426 ∆Pfur33::TT araC ParaBAD fur ∆asdA33 ∆relA197::araC PBAD lacI TT ∆(wza-wcaM)-8 ∆PtolR67::::TT araC ParaBAD tolR ∆ompA11 ∆pagP81::Plpp lpxE ∆lpxR9 (from χ12627) χ12632 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆pmi-2426 ∆Pfur33::TT araC ParaBAD fur ∆asdA33 ∆relA197::araC PBAD lacI TT ∆(wza-wcaM)-8 ∆PtolR67::::TT araC ParaBAD tolR ∆ompA11 ∆pagP81::Plpp lpxE ∆lpxR9 ∆eptA4 ∆arnT6 (from χ12631) χ12654 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆pmi-2426 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆relA197::araC PBAD lacI TT ∆recF126 ∆sifA26 ∆ompA11 ∆endA2311 (from χ12417) χ12655 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL ∆endA2113 ∆relA1123 ∆sseL116 ∆tlpA181 ∆ompA11 (from χ12563) χ12656 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆(wza-wcaM)-8 ∆relA197::araC ParaBAD lacI TT ∆recF126 ∆sifA26 ∆waaL46 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆araBAD65::TT ∆rhaBADSR515 ∆pagP8 ∆lpxR9 ∆ompA11 (from χ12569) χ12657 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆waaL46 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆endA2113 ∆relA1123 ∆sseL116 ∆tlpA181 ∆ompA11 (from χ12601) χ12658 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆waaL46 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆araBAD65::TT ∆rhaBADSR515 ∆pagP8 ∆lpxR9 ∆relA1123 ∆ompA11 (pSTUK201 ∆(traM-traX)- 36::araC ParaBAD lacI TT) (from χ12615) χ12667 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC ParaBAD c2 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆waaL46 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆araBAD65::TT ∆rhaBADSR515 ∆pagP8 ∆lpxR9 ∆relA1123 ∆ompA11(pSTUK206 ∆(traM-traX)- 41::araC ParaBAD lacI TT) (from χ12663) χ12733 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆pmi-2426 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆ompA11 ∆endA2311 ∆relA1123 (from χ12654) χ12734 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆relA197::araC PBAD lacI TT ∆recF126 ∆sifA26 ∆ompA11 ∆endA2311 pmi+ (from χ12654) χ12735 ∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆(wza-wcaM)-8 ∆relA1123 ∆recF126 ∆sifA26 ∆endA2113 ∆sseL116 ∆tlpA181 ∆rhaBADSR515 ∆araBAD65::TT ∆ompA11 (from χ12729) χ12736 ∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD1 waaL1 ∆(wza-wcaM)-8 ∆relA1123 ∆recF126 ∆sifA26 ∆endA2113 ∆sseL116 ∆tlpA181 ∆rhaBADSR515 ∆araBAD65::TT ∆ompA11 (from χ12730) χ12748 ∆PmurA25::TT araC ParaBAD murA ∆asdA27::TT araC PBAD c2 ∆(wza-wcaM)-8 ∆relA197::araC ParaBAD lacI TT ∆recF126 ∆sifA26 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD1 waaL ∆ompA11 ∆sopB1925 (from χ12452) χ12750 ∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆araBAD65::TT ∆rhaBADSR515 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆pagP8 ∆lpxR9 ∆relA1123 (pSTUK206 ∆(traM-traX)-41::araC ParaBAD lacI TT) ∆ompA11 (from χ12688) χ12751 ∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆araBAD65::TT ∆rhaBADSR515 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆pagP8 ∆lpxR9 ∆relA1123 (pSTUK206 ∆(traM-traX)-41::araC ParaBAD lacI TT) ∆ompA11 ∆sopB1925 (from χ12750) χ12753 ∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆araBAD65::TT ∆rhaBADSR515 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆pagP81:: Plpp lpxE ∆lpxR9 ∆relA1123 (pSTUK206 ∆(traM-traX)-41::araC ParaBAD lacI TT) ∆ompA11 (from χ12702) χ12754 ∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆araBAD65::TT ∆rhaBADSR515 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆pagP81:: Plpp lpxE ∆lpxR9 ∆relA1123 (pSTUK206 ∆(traM-traX)-41::araC ParaBAD lacI TT) ∆ompA11 ∆sopB1925 (from χ12753) χ12755 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆ompA11 ∆endA2311 ∆relA1123 (from χ12734) χ12756 ∆waaG42 ∆pagL21::TT araC ParaBAD waaG ∆lpxR9 ∆pagP8 ∆eptA4 ∆arnT6 ∆pmi-2426 ∆relA197::araC ParaBAD lacI TT ∆ompA11 ∆relA1123 (from χ12414) χ12775 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆ompA11 ∆endA2311 ∆relA1123 (pSTUK206 ∆(traM-traX)-41::araC ParaBAD lacI TT) (from χ12755) χ12776 ∆waaG42 ∆pagL21::TT araC ParaBAD waaG ∆lpxR9 ∆pagP8 ∆eptA4 ∆arnT6 ∆pmi-2426 ∆relA197::araC ParaBAD lacI TT ∆ompA11 ∆relA1123 (pSTUK206 ∆(traM- traX)-41::araC ParaBAD lacI TT) (from χ12756) χ12838 ∆PmurA25::TT araC PBAD murA ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD waaL1 ∆(wza-wcaM)-8 ∆recF126 ∆sifA26 ∆ompA11 ∆endA2311 ∆relA1123 ∆asdA33 (from χ12775) χ12846 ∆waaG42 ∆pagL21::TT araC ParaBAD waaG ∆lpxR9 ∆pagP8 ∆eptA4 ∆arnT6 ∆pmi-2426 ∆relA197::araC ParaBAD lacI TT ∆ompA11 ∆relA1123 (pSTUK206 ∆(traM- traX)-41::araC ParaBAD lacI TT) ∆sifA26 (from χ12776) We also constructed strains in which the ompAΩplz4 fusion replaced the wild- type chromosomal ompA gene to use as comparative controls with but one copy of the fusion. These strains constructed using the suicide vector pG8R315 (Table 2) are listed in Table 5. One of the examples is χ12619. The mutations ∆waaL46, ∆waaG42 and ∆waaC41 were introduced into strain χ12619 to generate a family of strains differing in the presence of the LPS O-antigen, LPS O-antigen and outer LPS core and O-antigen and outer and inner LPS core, respectively. We also constructed strain χ
Figure imgf000034_0001
12614 with the ∆asdA33 ∆ompA11 deletion mutations. This strain can be transformed with a plasmid encoding ompAΩplz4 to compare the effects of surface modification in Salmonella that affect the targeting ability of Salmonella. The plasmids could be pG8R341, or any plasmid carrying ompAΩplz4 fusion, or other ompA fused with varied targeting peptide sequences. The mutations ∆waaL46, ∆waaG42 and ∆waaC41 were introduced into χ12614 to generate a series of strains analogous to those generated in χ12619 resulting in defects in O-antigen, outer core and inner core, respectively. These strains are also listed in Table 5. Table 5. S. Typhimurium strains constructed with the ompAΩplz4 fusion χ12518 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 (from χ12516) χ12542 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 ∆waaC41 χ12617 ompAΩplz4 (from χ3761) χ12618 ∆relA4 ∆spoT1 ∆asdA27::TT araC ParaBAD c2 ompAΩplz4 (from χ11001) χ12619 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 ompAΩplz4 (from χ12518) χ12808 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 ompAΩplz4 ∆waaL46 (from χ12619) χ12809 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 ompAΩplz4 ∆waaG42 (from χ12619) χ12810 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 ompAΩplz4 ∆waaC41 (from χ12619) χ12811 ∆alr-3 ∆PdadB66::TT araC ParaBAD dadB ∆PasdA55::TT araC ParaBAD asdA ∆fliC180 ∆pagP81::Plpp lpxE ∆pagL7 ∆lpxR9 ∆waaC41 ompAΩplz4 (from χ12542) χ12614 ∆asdA33 ∆ompA11 (from χ8958) χ12812 ∆asdA33 ∆ompA11 ∆waaL46 (from χ12614) χ12813 ∆asdA33 ∆ompA11 ∆waaG42 (from χ12614) χ12814 ∆asdA33 ∆ompA11 ∆waaC41 (from χ12614) Example 3. Construction of plasmid vectors for use and evaluation in candidate CCTS strains. Figures 1 and 2 depict the plasmids used as parents or for component segments of derived and constructed plasmids. The derived and constructed plasmids are listed and described in Table 6 with their use and evaluation described in subsequent Examples. Table 7 lists all the nucleotide primers used to construct the plasmids listed in Table 6. A unique and original feature of many of the plasmids designed and constructed is the ability to encode proteins that are synthesized by the CCTS strain to be displayed during targeting and attaching to, invading into and acting within tumor cells in vivo prior to display of regulated lysis within the tumor cell to release the plasmid now serving as a DNA vaccine with unique features to be directed to the nucleus for transcription of encoded sequences that yield products after mRNA translation that exhibit anti-tumor activities. These products with their features as described in later Examples might kill the tumor cell, cause the tumor cell to kill itself (i.e., commit suicide) and/or attract host immune responses that inhibit tumor cell growth and metastases. The ability to design and effectively use these newly designed dual function hybrid plasmids is dependent on using CCTS delivery strains that are engineered to express a regulated delayed lysis in vivo phenotype that is a composite function of regulated expression of both chromosomal and plasmid encoded genes. Table 6. Lists of plasmids generated for use in anti-tumor researcha.
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
a
Figure imgf000042_0001
The sizes of all plasmids in number of nucleotide bases is indicated for each plasmid in the accompanying Figures diagramming the plasmids listed in this Table 6. Table 7. List of all the nucleotide primers used to construct the plasmids listed in Table 6.
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Example 4. Insertion of the nucleotide sequence encoding the nine amino acids of PLZ4 into the third exposed loop of the S. Typhimurium ompA gene and construction of plasmids to encode synthesis of this fusion or insert it into the S. Typhimurium chromosome. The pG8R314 plasmid (Figure 3) with the ompAΩplz4 fusion was constructed by amplifying a 454 bp fragment of the S. Typhimurium UK-1 (χ3761) chromosome using primers OmpA-s and OmpA-PLZ4-a and a 707 bp fragment with primers OmpA-PLZ4-s and OmpA-SacIHindIII-a. These two fragments were cloned into plasmid pYA3342 (Figure 1A) cut with NcoI/HindIII to generate plasmid pG8R314. Note that the sequence encoding the PLZ4 peptide was introduced by both primers OmpA-PLZ4-a and OmpA- PLZ4-s. This plasmid has a gene encoding the PLZ4 peptide inserted into the third exposed loop of the OmpA protein enabling expression of the ompAΩplz4 insertion mutated gene in Salmonella. The PLZ4 peptide was flanked with 2 cysteines forming a disulfide linkage to facilitate its exposure on the loop 3. To construct the suicide vector pG8R315 (Figure 4), the plasmid pG8R314 was used as the template to generate a 1.1 kb fragment encoding synthesis of OmpAΩPLZ4. This fragment was amplified with primers OmpA-XbaI-s and OmpA- SacIHindIII-a and cut with XbaI/SacI. The fragment was then inserted into suicide plasmid pRE112 (Figure 1B) cut with XbaI/SacI to generate plasmid pG8R315. This suicide vector is then used to introduce the ompAΩplz4 mutation into the chromosome of the S. Typhimurium strains listed in Table 5. For the construction of pG8R319 (Figure 5A), we fused two DNA fragments. With plasmid pYA4545 (Figure 1C) as a template, a 1,549 bp fragment containing rrfG TT-PCMV-SV40 polyA Trp TT was amplified with primers rrfGTT-s and trpTT-a. With plasmid pG8R314 (Figure 3) as a template, a 4,085 bp fragment that includes the whole pG8R314 plasmid was amplified with primers trpTT-s and rrfGTT-a. The two fragments were assembled to generate plasmid pG8R319. The balanced-lethal plasmid has a pBR ori and could express ompAΩplz4 in Salmonella and has a Pcmv promoter to be used for gene expression in eukaryotic cells. The Pcmv and Ptrc ompAΩplz4 are separated by the trpA TT. For the construction of pG8R320 (Figure 5B), we used plasmid pYA4545 (Figure 1C) as a template to generate a 8 kb fragment containing the entire pYA4545 plasmid that was amplified with primers pYA4545-TT-BstBI-s and pYA4545-TT-a1 and then extended by PCR with primers pYA4545-TT-BstBI-s and pYA4545-TT-BcII-a. Then with plasmid pG8R314 (Figure 3) as a template, a 1,242 bp fragment containing Ptrc- ompA was amplified with primers Ptrc-BclI-s and ompA-BstEI-a. The two fragments were then assembled to generated plasmid pG8R320 (Figure 5B). This regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the cell employs the Pcmv promoter to express an inserted gene sequence in eukaryotic cells. The Pcmv and Ptrc ompAΩplz4 are separated in the dual plasmid vector pG8R320 by the regulated delayed lysis cassette araC ParaBAD GTG murA GTG asdA. Example 5. Display of OmpAΩPLZ4 on the bacterial cell surface enables S. Typhimurium cells to preferentially attach to bladder tumor cells. The ompAΩplz4 mutation was introduced into strain χ12518 to generate strain χ12619 using suicide vector χ7213(pG8R315). Both strains were transformed with plasmid pYA4090 to enable tagging the bacteria with the GFP protein. Overnight cultures of χ12518(pYA4090) and χ12619(pYA4090) were diluted into LB broth with 0.1% arabinose and grown until OD600 reached 0.9. The bacteria were washed once with PBS and then used to infect MB49 murine bladder cancer cells and 5637 human bladder cancer cells at MOI 1:100 for 1 hour. The MB49 membrane was stained with Cell Plasma Membrane Staining Kit - Orange Fluorescence - Cytopainter (ab219941, Abcam). As shown in Figure 6, the strain χ12619 with OmpAΩPLZ4 is more attracted to and invades the bladder cancer cells much better than the strain χ12518 without the OmpAΩPLZ4 fusion. Example 6. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express proteins that synthesize the CXCL11 chemokine that attract cells of the immune system. Although the quantity of immune cells in the bladder is not well studied, the bladder has γδ, CD4 and CD8 T cells, macrophages, dendritic cells and NK cells (155). Notably, there is no report of CD8 T cells in the mouse bladder (155). Thus, it is important to recruit T cells to the bladder to potentiate immunotherapy of bladder cancer. CXCL11 functions by binding to the receptors CXCR3 predominantly, as well as CXCR7 (156-160). CXCR3 is expressed on immune cells, such as activated T cells, NK and NKT cells, DCs, but not on naive T cells (161), and a variety of non-immune cells, such as astrocytes, fibroblasts, endothelial cells, epithelial muscle cells, and cancer cells (162, 163). CXCR7 is expressed on multiple immune cells, such as T cells, monocytes, DC cells, B cell and NK cells (164). CXCL11 has diverse functions including inhibiting angiogenesis, increasing immune cell migration, affecting proliferation of different cell types, stimulation of IFN-γ production by immune cells, suppressing M2 macrophage polarization, playing a role in fibroblast directed carcinoma invasion, increasing adhesion and invasion properties, facilitating the migration of certain immune cells, and serving as an adjuvant to anti-cancer therapies (160, 165). Although CXCL11 mainly works for immune cell migration, differentiation and activation, it could promote cancer cell proliferation and metastasis. Intratumor delivery of CXCL11 has been shown to enhance the efficacy of T-cell infiltration, adoptive T-cell therapy and vaccine efficacy (166-169). Locally produced CXCL11 in tumor cells will mediate the recruitment of T cells and NK cells to the tumor site to combat tumor development and growth. This will reduce the global toxicity related to overproduction of CXCL11 in non-tumor sites. For these reasons, we determined that the synthesis of CXCL11 by CCST cells would be optimal if the chemokine was synthesized by tumor cells rather than into the environment if synthesized and delivered by the CCST cells being used for combatting bladder cancer. Figure 7 displays diagrams of pG8R321 (A) and pG8R322 (B) that express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after plasmid release of the plasmid in tumor cells employs the Pcmv promoter to express the human and mouse CXCL11 chemokines, respectively. To construct pG8R321, we used the CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template to amplify the gene encoding human CXCL11 with primers Human-CXCL11-KpnI-s and Human- CXCL11-Not-a, which was inserted into plasmid pG8R319 cut with KpnI/NotI. The balanced-lethal vector-host combination specifically targets human bladder cancer cells due to the display of the OmpAΩPLZ4 surface protein fusion to induce synthesis of the human CXC11 after invasion into tumor cells to release pG8R321. pG8R322 was similarly constructed using CXCL11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as the template to amplify the gene encoding mouse CXCL11 with primers Mouse-CXCL11- KpnI-s and Mouse-CXCL11-NotI-a. This sequence was then inserted into plasmid pG8R319 cut with KpnI/NotI to generate plasmid pG8R322. The balanced-lethal vector- host targets mouse bladder cancer cells due to the display of the OmpAΩPLZ4 surface protein fusion to induce synthesis of the murine CXC11 after invasion into tumor cells to release pG8R322. Example 7. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence encoding for synthesis of KillerRed to potentiate tumor cell killing. Photodynamic therapy is an important therapeutic treatment for cancer and other diseases. KillerRed is the first engineered photosensitizer with light-induced cytotoxicity that could be used for precise light-induced cell killing and target protein inactivation (170-175). Upon light activation, KillerRed will produce toxic reactive oxygen species to use for photodynamic therapy against cancer. Plasmid pG8R323 (Figure 7C) carries the gene encoding a membrane-targeting KillerRed by fusing with Neuromodulin N-terminal sequence (KillerRed mem thereafter) while plasmid pG8R324 (Figure 7D) carries the gene encoding a mitochondria targeting KillerRed by fusion with mitochondrial location signals (KillerRed mito thereafter). Both plasmids are balanced-lethal plasmids. These vectors specify expression of the ompAΩplz4 gene in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after release from the CCST cell in the tumor cell employs the Pcmv promoter to express the KillerRed encoding genes to kill tumor cell when induced with light. To construct pG8R323 (Figure 7C), we used plasmid pCS2-NXE+mem-KillerRed (Addgene Cat# 45761) as a template to amplify the gene encoding KillerRed mem using primers KillerRed-Mem-KpnI-s and KillerRed-NotIXhoI-a. The sequence was then inserted into plasmid pG8R319 (Figure 5A) cut with KpnI/NotI to generate plasmid pG8R323. The balanced-lethal vector-host construct with pG8R323 specifies synthesis of a membrane-targeted KillerRed. To construct pG8R324 (Figure 7D) specifying KillerRed-dMito, we used pKillerRed-dMito (EVROGEN cat# FP964) as a template by amplifying the gene encoding KillerRed mito with primers KillerRed-Mito-KpnI-s and KillerRed-NotIXhoI-a and inserting into plasmid pG8R319 (Figure 5A) cut with KpnI/NotI to generate plasmid pG8R324. The balanced-lethal vector-host construct carries a mitochondria-targeted KillerRed. KillerRed localized on cellular membranes can be used for effective light-induced cell killing by light-induced production of reactive oxygen species and can also be used to detect tumor cells infected with the CCST cells. Example 8. Construction of CCST strains with regulated delayed lysis with regulated delayed lysis high copy number plasmid vectors encoding display of the OmpAΩPLZ4 surface protein fusion and in situ synthesis of CXCL11 and KillerRed. The plasmids pG8R321, pG8R322, pG8R323 and pG8R324 (Figure 7) all have the moderate copy number pBR ori and specify the balanced-lethal phenotype using plasmid encoded expression of the asdA gene. We previously determined during the development of means for DNA vaccine delivery by Salmonella (45) that use of high copy number plasmids with pUC ori acting as DNA vaccines were more effectively delivered by Salmonella cells undergoing lysis after invasion into host cells. To further validate this belief in developing optimal means for using CCST strains, we constructed versions of the pG8R321, pG8R322, pG8R323 and pG8R324 plasmids with the high copy number pUC ori and with regulated delayed lysis attributes to use in Salmonella vector strains also displaying the regulated delayed lysis phenotype (see Table 4). To construct pG8R325 (Figure 8A) encoding human CXCL11, we amplified a sequence from CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template with primers Human-CXCL11-KpnI-s and Human-CXCL11-NotI-a and cloned into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R325. The lysis vector carries a human CXC11 gene. To construct pG8R326 (Figure 8B) encoding mouse CXCL11, we amplified a sequence from CXCL11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template with primers Mouse-CXCL11-KpnI-s and Mouse-CXCL11-NotI-a to insert into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI. The pG8R326 lysis vector carries a mouse CXC11 gene. CXCL11 is chemotactic for interleukin-activated T-cells but not unstimulated T- cells, neutrophils or monocytes. It is the dominant ligand for CXCR3. To construct pG8R327 (Figure 8C), we used pCS2-NXE+mem-KillerRed (Addgene Cat# 45761) as a template to amplify the gene encoding KillerRed mem with primers KillerRed-Mem-KpnI-s and KillerRed-NotIXhoI-a to insert into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI. The pG8R327 lysis vector carries a membrane- targeted KillerRed. To construct pG8R328 (Figure 8D), we used pKillerRed-dMito (EVROGEN cat# FP964) as a template to amplify a sequence encoding KillerRed mito using primers KillerRed-Mito-KpnI-s and KillerRed-NotIXhoI-a to insert into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI. The lysis vector pG8R328 specifies synthesis of the mitochondria-targeted KillerRed. KillerRed localized on cellular membranes can be used for effective light-induced cell killing by light-induced production of reactive oxygen species. Example 9. Construction of plasmid vectors encoding synthesis of GFP or EGFP to track Salmonella extracellularly and intracellularly to evaluate the targeting ability of CCTS strains to bladder tumors. Figure 9 displays the diagrams of plasmids pG8R341 (Figure 9A) and pG8R342 (Figure 9B) used to tag Salmonella with fluorescent proteins. The balanced-lethal plasmid pG8R341 carries a Ptrc promoter which can express the operon fusion of ompAΩplz4 and gfp that enables GFP production in the Salmonella cytosol and displays the synthesized OmpAΩPLZ4 on the cell surface of Salmonella. Ptrc is a prokaryotic promoter that can express at high level protein synthesis under both anaerobic and aerobic conditions and is repressed by LacI (176). Salmonella strains carrying this plasmid in vivo in the absence of arabinose to preclude synthesis of LacI will produce GFP in Salmonella cells present extracellularly and intracellularly. This regulated delayed lysis plasmid pG8R342 (Figure 9B) has a pUC ori and can express the ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the cell within host cells employs the Pcmv promoter to express an inserted egfp in eukaryotic cells. The EGFP is only produced when Salmonella is inside the mammalian cells. To construct pG8R341 (Figure 9A), we used plasmid pYA4090 as a template to amplify the gene encoding GFP3 using primers SD-GFP-SacI-gs and GFP-HindIII-ga. The sequence was inserted into plasmid pG8R314 (Figure 3) cut with SacI/HindIII to generate plasmid pG8R341. The balanced-lethal plasmid uses GFP to track Salmonella with OmpAΩPLZ4. To construct pG8R342 (Figure 9B), we used plasmid pYA4685 as a template to amplify the gene encoding EGFP using primers EGFP-KpnI-gs and EGFP-XhoI-ga. The sequence was inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/XhoI to generate plasmid pG8R342. The lysis plasmid uses EGFP to track Salmonella within mammalian cells. Example 10. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express gene sequences encoding KillerRed to potentiate tumor cell killing and CXCL11 to attract immune cells. A construction that can kill cancer cells and recruit immune cells to tumor cells will have synergic effect to benefit bladder cancer therapy. Figure 10 displays diagrams of such constructions, balanced-lethal plasmids with regulated delayed lysis attributes pG8R343 (Figure 9A) and pG8R344 (Figure 9B) with pUC ori that express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the bacterial cell employs the Pcmv promoter to express genes encoding KillerRed mem that kill tumor cells and human or mouse CXCL11 that recruit immune cells to combat bladder tumors. To enable co-expression of genes encoding KillerRed mem and human or mouse CXCL11 in mammalian cells, a P2A peptide (177-179) was introduced between the genes encoding KillerRed mem and human or mouse CXCL11 to enable ribosome skip to enable synthesis of a peptide bond at the C-terminus of a 2A element leading to cleavage between the end of the 2A sequence and the CXCL11 peptide downstream (177-179). To construct pG8R343 (Figure 10A), we fused two fragments encoding KillerRed mem P2A and human CXCL11. We used plasmid pCS2-NXE+mem- KillerRed (Addgene plasmid # 45761) as a template to amplify the gene encoding KillerRed mem using primers KillerRed-Mem-KpnI-s and KillerRed-C-P2A-a1. The fragment was used as a template and amplified with primers KillerRed-Mem-KpnI-s and KillerRed-C-P2A-a2 to include the sequence encoding P2A. We then used CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template to amplify the gene encoding human CXCL11 using primers P2A-Human CXCL11-s and Human-CXCL11-NotI-a. The KillerRed Mem-P2A and human CXCL11 fusion sequence was inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R343. The lysis plasmid carries genes encoding KillerRed mem and human CXCL11. The two genes were separated by a P2A peptide (177-179) which can induce cleavage at the C- terminal of the P2A peptide to enable production of both KillerRed mem and human CXCL11 after Salmonella invasion into tumor cells and lysis to release pG8R343 and enable synthesis of KillerRed mem and human CXCL11. To construct pG8R344 (Figure 10B), we fused two fragments for KillerRed mem P2A and mouse CXCL11. The KillerRed mem P2A was generated as above for plasmid pG8R343. We used CXCL11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template to amplify the gene encoding mouse CXCL11 using primers P2A-Mouse CXCL11-s and Mouse- CXCL11-NotI-a. The KillerRed Mem-P2A and mouse CXCL11 fusion was inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R344. The lysis plasmid carries genes encoding KillerRed mem and mouse CXCL11. The two genes were separated by a P2A peptide (177-179) which can induce cleavage at the C- terminal of the P2A peptide to enable production of both KillerRed mem and mouse CXCL11 after Salmonella invasion into tumor cells and lysis to release pG8R344 and enable synthesis of KillerRed mem and mouse CXCL11. Example 11. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then after invasion express a gene sequence fused with an HLA peptide encoding sequence to potentiate immune responses to tumor cells. Many factors can affect vaccine-induced immune responses. Antigens can be linked to lysosomal or endosomal targeting signals to route the antigen into a MHC class II processing compartment to improve CD4+ T cell responses. A chimeric protein fused with the N-terminal leader peptide with an MHC class I trafficking signal (tail peptide) attached to the C-terminal end of an antigen can strongly improve the presentation of MHC class I and class II epitopes in human and murine dendritic cells, leading to efficient expansion of antigen specific CD4+ and CD8+ T cells and their effector functions (180). We thus generated plasmid pG8R345 (Figure 11 A) specifying HLAB leading and tail peptides to enable an inserted antigen to be presented to MHC class I and class II pathways. To construct pG8R345 (Figure 11A), we used HLAB (HLA-B) (NM_005514) Human Untagged Clone (ORIGENE Cat# SC124484) as a template to amplify the gene encoding the HLA leading peptide using primers HLAB-Leading-gs and HLAB-Leading- MCS-ga and HLA tail peptide using primers HLAB-tail-MCS-gs and HLAB-tail-XhoI-ga. The sequence generated encoding the above two fragments were inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/XhoI to generate plasmid pG8R345. This plasmid has sequences for HLAB leading and tail peptides which could be used to coupling a selected antigen to MHC Class I Trafficking Signals to increase antigen presentation efficiency (180). This regulated delayed lysis plasmid pG8R345 has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after tumor cell invasion and lysis of the CCST cell employs the Pcmv promoter to express the HLA-antigen encoding gene in eukaryotic tumor cells. We then inserted egfp into plasmid pG8R345 to generate plasmid pG8R346 (Figure 11 B). The egfp is under the control of PCMV which can be expressed in eukaryotic cells. To construct pG8R346, we used plasmid pYA4685 (Figure 2D) as a template to amplify the gene encoding EGFP using primers EGFP(HLAB)-KpnI-gs and EGFP(HLAB)-AvrII-ga. The sequence was inserted into plasmid pG8R345 (Figure 11A) cut with KpnI/AvrII to generate plasmid pG8R346. The lysis plasmid carries a gene encoding EGFP and could be used to track the CCTS strain in mammalian cells. The 5’ and 3’ terminal nucleotide sequences of eukaryotic genes affect the translation of the gene (181-183). Thus, we generated plasmid pG8R347 (Figure 12A) that includes the 5’ and 3’ terminal nucleotide sequence of HLAB gene. To construct pG8R347, we used HLAB (HLA-B) (NM_005514) Human Untagged Clone (ORIGENE Cat# SC124484) as a template to amply the sequence of the 5’ terminal of HLAB using primers HLAB-5' Leading-gs and HLAB-Leading-MCS-ga and the 3’ terminal of HLAB using primers HLAB-tail-MCS-gs and HLAB-3' tail-XhoI-ga. The above two fragments were cloned into pG8R320(Figure 5B) cut with KpnI/XhoI to generate pG8R347. The lysis plasmid has sequences for HLAB 5’ terminus, leading and tail peptides and 3’ terminus which could be used to coupling antigen to MHC Class I Trafficking Signals to increase antigen presentation efficiency (180). The inclusion of 5’ and 3’ termini of HLAB increases the transcript stability and translational efficiency (184). Similar, we inserted the egfp gene into plasmid pG8R347 to generate plasmid pG8R348 (Figure 12B). To construct plasmid pG8R348, we used plasmid pYA4685 as a template to amplify the gene encoding EGFP using primers encoding EGFP was amplified with primers EGFP(HLAB)-KpnI-gs and EGFP(HLAB)-AvrII-ga and cloned into plasmid pG8R347 (Figure 12A) cut with KpnI/AvrII to generate plasmid pG8R348. The lysis plasmid carries a gene encoding EGFP and could be used to track the Salmonella in mammalian cells, although this will require replacing the ompAΩplz4 construction that enables targeting to bladder tumor cells with sequences specifying a protein to target other cell types. Tumor neoantigens can be presented by major histocompatibility complex proteins and recognized by T cells to induce anti-tumor immune responses. This approach has been used as therapeutic vaccines in preclinical models to promote tumor specific T-cell responses (185-188). Tumor neoantigens are derived from mutated proteins that lead to the generation of novel immune epitopes that are foreign to the body (189, 190). Vaccines targeting tumor neoantigens are a promising strategy for personalized cancer immunotherapy (188, 190-197). Due to the complex immune tolerance mechanisms in tumors, neoantigen based tumor vaccines are normally combined with immune checkpoint inhibitors. Clinical trials with this combination therapy demonstrated that the induction of neoantigen-specific CD4+ and CD8+ T cell responses and cytotoxic vaccine-induced T cells, had some efficacy in treating bladder cancer (188, 191, 198). Mouse derived BBN963 (199) and MB49 (200, 201) cell lines are commonly used as an in vitro and in vivo model of bladder cancer. Neoantigens have been identified in these two cell lines (202). These neoantigens were cloned into vector pG8R347 to generate plasmid pG8R349 (Figure 12C) and pG8R350 (Figure 12D) to be used in vaccine trials. To construct pG8R349, the gene encoding neo-antigen BBN963 (202) was cut from with plasmid pUC57-BBN963 with KpnI/AvrII and cloned into plasmid pG8R347 (Figure 12A) cut with same enzymes to generate plasmid pG8R349. The lysis plasmid carries a gene encoding neo-antigen BBN963. To construct pG8R350, the gene encoding neo-antigen MB49 (202) was cut from with plasmid pJET1.2-MB49 with KpnI/AvrII and cloned into plasmid pG8R347 (Figure 12A) cut with same enzymes to generate plasmid pG8R350. The lysis plasmid carries a gene encoding neo-antigen MB49. Example 12. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence under the control of PEF1α promoter. The viral derived PCMV promoter is a strong promoter that has been widely used to express genes in eukaryotic cells as in DNA or viral vectors, such as adenovirus. However, it could be silenced in certain cell types due to methylation, leading to considerable variability gene expression in different cell types (203, 204). Human elongation factor-1α is a constitutive human promoter that can drive ectopic gene expression homogeneously and persistently in vivo and in vitro (205-207). It can replace PCMV when PCMV has diminished activity due to being silenced. We therefore generated the regulated delayed lysis plasmid pG8R361 (Figure 13) that has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the PEF1α promoter to express inserted genes in eukaryotic cells. For the construction of pG8R361 (Figure 13), we fused two fragments. We used plasmid pG8R320 (Figure 5B) as a template to generate a 7.4 kb fragment with primers 4545-(ForPEF1a) KpnIXmaINotIXhoI-s and 4545(ForPEF1a)-a2. Then, with plasmid pLVX EF1α IRES Puro N (BEI NR52973) as a template, a fragment containing the PEF1α promoter was amplified with primers PEF1a-s and PEF1a-KpnI-a. The two fragments were then assembled into plasmid pG8R361. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface to target bladder cancer cells and after lysis of the CCTS cell employs the PEF1α promoter to express an inserted gene sequence in eukaryotic cells (208-212), in this case into bladder tumor cells. The PEF1α and Ptrc ompAΩplz4 in the dual plasmid vector pG8R361 are separated by the regulated delayed lysis cassette araC ParaBAD GTG murA GTG asdA. Example 13. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence encoding HAC-PD1 to block PD1L1 and activate T cells and CXCL11 to recruit immune cells. The interaction between the Programmed cell death protein-1 (PD-1) and programmed cell death ligand-1 (PD-L1) functions as a T cell checkpoint to regulate T cell responses. Cancer cells upregulate the levels of PD-L1 to evade immune detection and elimination (213). Monoclonal antibodies blocking PD1 and PDL1 have been approved as effective immunotherapies against different tumors (214-219). However, use of antibodies have inherent limitations that include poor and slow distribution within hypoxic regions of large tumors (220, 221) and immune-related adverse events, such as Fc-mediated cytotoxic immune responses (222) and severe cytokine associated inflammatory and immunological process (223, 224). For monoclonal antibodies against PD1/PLD1, they can also reduce circulation of T cell numbers in patients (225-227). To overcome these shortcomings, a soluble fragment of the PD1 ectodomain, the high- affinity consensus (HAC)-PD1, was identified as an alternative agent that exhibits improved antitumor responses and avoids antibody limitations (228, 229). The HAC- PD1 (228, 229) has an over 40,000-fold higher affinity for PD-L1 than native PD1 (229) and 32- and 12-times higher affinity than the FDA-approved anti-PD-L1 antibodies atezolizumab and durvalumab, respectively (230). At the same dose and schedule through intratumoral injection, it is also more effective than an anti-PD1 antibody in inducing anti-cancer immunity (229). Multiple vectors carrying the gene encoding HAC- PD1 are depicted in Figures 14, 15 and 16. Figure 14 depicts plasmids that contain sequences encoding HAC-PD1 combined with CXCL11 for combinational immunotherapy. Figure 15 depicts plasmids that contain sequences encoding IL2 SS- HAC-PD1 and CXCL11 for combinational immunotherapy. Figure 16 depicts plasmids that carry sequences encoding HAC-PD1, HAC-PD1 and EGFP, CXCL11 and EGFP. All these plasmids display the synthesized OmpAΩPLZ4 on the cell surface to target bladder cancer cells and after lysis of the CCTS cell employ the PCMV or PEF1α promoter to express the inserted eukaryotic genes in bladder tumor cells. To construct plasmid pG8R362 (Figure 14A), we used plasmid pMal-HAC-PD1 (228, 229) as a template to amplify the gene encoding HAC-PD1 using primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a1 and then extended by PCR with primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a2. Then with CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template, the gene encoding human CXCL11 was amplified with primers Linker- hCXCL11-s and Human-CXCL11-NotI-a. The two fragments were inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R362. The lysis plasmid carries the gene encoding HAC-PD1 fused with human CXCL11 with a 36 amino acid linker under the control of a PCMV promoter. The 36 aa linker GGS(GGGSE)5(GGGS)2 was inserted to enable fusion of HAC-PD1 and human CXCL11 (231). The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the fused gene sequence. To construct plasmid pG8R363 (Figure 14B), we used plasmid pMal-HAC-PD1 (228, 229) as a template to amplify the gene encoding HAC-PD1 using primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a1 and then extended by PCR with primers HAC- PD1-KpnI-s and HAC-PD1-Linker-a2’). Then with Cxcl11 (NM_019494) Mouse Tagged ORF Clone (mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template, the gene encoding mouse CXCL11 was amplified with primers Linker- mCXCL11-s and Mouse-CXCL11-NotI-a. The two fragments were inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R363. The lysis plasmid carries a gene encoding HAC-PD1 and fused with mouse CXCL11 with a 36 amino acid linker under the control of a PCMV promoter. The 36 aa linker GGS(GGGSE)5(GGGS)2 was inserted to enable fusion of HAC-PD1 and mouse CXCL11 (231). The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the fused gene sequence. To generate plasmid pG8R364 (Figure 14C), a fragment encoding HAC-PD1- hCXCL11 was cut from plasmid pG8R362 (Figure 14A) with KpnI/NotI and cloned into plasmid pG8R361 (Figure 13) cut with the same enzymes to generate plasmid pG8R364. The lysis plasmid carries a gene encoding HAC-PD1 that was fused with human CXCL11 using a 36 amino acid linker under the control of the PEF1α promoter. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the PEF1α promoter to express the fused gene sequence. To generate plasmid pG8R365 (Figure 14D), a fragment encoding HAC-PD1- mCXCL11 was cut from plasmid pG8R363 (Figure 14B) with KpnI/NotI and cloned into plasmid pG8R361 (Figure 13) cut with the same enzymes to generate plasmid pG8R365. The lysis plasmid carries a gene encoding HAC-PD1 that was fused with mouse CXCL11 using a 36 amino acid linker under the control of the PEF1α promoter. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the PEF1α promoter to express the fused gene sequence. Secretion of proteins increases the levels of therapeutic molecules that can significantly enhance the efficacy of therapy at the site of the disease. The IL2 signal peptide is one of the most commonly used secretion facilitating sequences used for protein production in gene therapy research (232-234). To increase the secretion of HAC-PD1, we first generated plasmid pG8R366 (Figure 15A) that carries the sequence encoding the IL2 secretion signal. We amplified the IL2 SS using primers IL2-s2 and IL2-a2. The sequence was inserted into plasmid pG8R320 (Figure 5B) cut with KpnI to generate plasmid pG8R366. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express any gene sequence fused to the IL2 secretion signal sequence. We then inserted HAC-PD1 with human and mouse CXCL11 into pG8R366 to generate plasmids pG8R367 (Figure 15B) and pG8R368 (Figure 15C), respectively. To generate plasmid pG8R367, the fragment encoding HAC-PD1-hCXCL11 was cut from plasmid pG8R362 (Figure 14A) with KpnI/NotI and inserted into plasmid pG8R366 (Figure 15A) cut with the same enzymes to generate plasmid pG8R367. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the gene sequence encoding HAC-PD1-human CXCL11 fused to the IL2 secretion signal. To generate plasmid pG8R368, the fragment encoding HAC-PD1-mCXCL11 was cut from plasmid pG8R363 (Figure 14B) with KpnI/NotI and inserted into plasmid pG8R366 (Figure 15A) cut with the same enzymes to generate plasmid pG8R368. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the gene sequence encoding HAC-PD1-mouse CXCL11 fused to the IL2 secretion signal. We also generated plasmid pG8R372 (Figure 16A) which only has HAC-PD1 fused to the IL2 secretion signal. To generated plasmid pG8R372, we used plasmid pG8R367 (Figure 15B) as a template to amplify the fragment encoding IL2 SS and HAC-PD1 with primers IL2-s2 and HAC-PD1-a. The fragment was inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R372. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the gene sequence encoding HAC-PD1- fused to the IL2 secretion signal. To facilitate tracking of HAC-PD1 in mammalian cells, we tagged HAC-PD1 with EGFP to generate plasmid pG8R373 (Figure 16B). For the construction of plasmid pG8R373, we used plasmid pG8R367 (Figure 15B) as a template to amplify the fragment encoding IL2 SS-HAC-PD1 fusion using primers IL2-s2 and HAC-PD1-linker- EGFP-a. And then with plasmid pYA4685 (Figure 2D) as a template, the gene encoding EGFP was amplified with primers HAC-PD1-linker-EGFP-s and C terminal EGFP- XhoINotI-a. The two fragments were inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R373. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the gene encoding fusion of IL2 secretion signal-HAC-PD1- fused and EGFP. Similarly, we generate pG8R374 (Figure 16C) and pG8R375 (Figure 16D) in which CXCL11 fused with EGFP to track the behavior of CXCL11. To generate plasmid pG8R374, we used CXCL11 (NM_005409) Human Tagged ORF Clone (human CXCL11(Myc-DDK-tagged), ORIGENE Cat# RC210320) as a template to amplify the gene encoding human CXCL11 using primers Human-CXCL11-KpnI-s and hCXCL11- EGFP-a. And then with plasmid pYA4685 (Figure 2D) as a template, the gene encoding EGFP was amplified with primers HAC-PD1-linker-EGFP-s and C-terminal EGFP- XhoINotI-a. The two fragments were inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R374. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express the gene encoding the fusion of human CXCL11 and EGFP. To generate plasmid pG8R375 (Figure 16D), we used CXCL11 (NM_019494) Mouse Tagged ORF Clone ( mouse CXCL11(Myc-DDK-tagged), ORIGENE Cat# MR222244) as a template to amplify the gene encoding mouse CXCL11 using primers Mouse-CXCL11-KpnI-s and mCXCL11-EGFP-a. And then with plasmid pYA4685 (Figure 2D) as a template, the gene encoding EGFP was amplified with primers HAC- PD1-linker-EGFP-s and C-terminal EGFP-XhoINotI-a. The 2 fragments were inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R375. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell in situ employs the Pcmv promoter to express the gene encoding fusion of mouse CXCL11 and EGFP. Example 14. Insertion of the nucleotide sequence encoding the ten amino acids of the luteinizing hormone-releasing hormone (LHRH) peptide binding to the LHRH receptor into the third exposed loop of the S. Typhimurium ompA gene to cause CCTS strains to target endometrial, bladder, ovarian, prostate and breast tumors with overexpression of LHRH receptors. Luteinizing hormone-releasing hormone (LHRH) receptors are overexpressed in many cancers, including endometrial, bladder, ovarian, prostate and breast cancers (235-242), while limited in normal healthy tissues. LHRH has been employed to efficiently guide anticancer and imaging agents to cancer cells, thereby increasing the amount of these substances in tumors, but limiting delivery to normal tissues to reduce unnecessary exposure and toxicity (235, 241, 243, 244). We thus generated regulated delayed lysis plasmids pG8R380 (Figure 17A) and pG8R381 (Figure 17B) that have a pUC ori and can express ompAΩlhrh in Salmonella to display the synthesized OmpAΩLHRH on the cell surface to target endometrial, bladder, ovarian, prostate and breast cancers and after lysis of the CCTS cell in the invaded tumor cell employ the Pcmv promoter to express a selected gene of importance to tumor therapy or identification. To construct plasmid pG8R380 (Figure 17A), we used plasmid pG8R320 (Figure 5B) as a template to amplified a 540 bp fragment using primers Ptrc-BclI-s and LHRH-a and a 707 bp fragment using primes LHRH-s and OmpAGFP-BstBI-a. And then with plasmid pYA4090 (Figure 2C) as a template, the gene encoding GFP was amplified with primer GFP-s and GFP-BstBI-a. The above 3 fragments were then inserted into plasmid pG8R320 (Figure 5B) cut with BclI/BstBI to generate plasmid pG8R380. The regulated delayed lysis plasmid has a pUC ori and can express the ompAΩlhrh and gfp gene sequences in Salmonella to display the synthesized OmpAΩLHRH on the cell surface and GFP in the cytosol and after lysis of the CCTS cell employs the Pcmv promoter to express a selected gene to specify synthesis of a desired gene product. To construct plasmid pG8R381 (Figure 17B), the plasmid pG8R380 was cut with BstBI to remove the gene encoding GFP. The 8.1 kb fragment was self-ligated to generate plasmid pG8R381. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩlhrh and gfp in Salmonella to display the synthesized OmpAΩLHRH on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express a selected gene to specify synthesis of a desired gene product. Example 15. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells and then express a gene sequence encoding haPD1-IgG to block PD1L1 inactivation of T cell functions and a gene sequence encoding KillerRed mem to kill tumor cells. The HAC-PD1 (228, 229) has a 40,000-fold higher affinity for inactivating PD-L1 than native PD1 (229) and a 32- and 12-times higher inactivating ability than the FDA- approved anti-PD-L1 antibodies atezolizumab and durvalumab, respectively (230). HAC-PD1 is also more potent than an anti-PD1 antibody in inducing anti-cancer immunity using the same dose and schedule for intratumor injection (229). However, HAC-PD1, due to its small size, can leak from cells and thus elicit undesired immune responses against normal tissues. Furthermore, HAC-PD1 has a relatively short half-life and thus requires daily intratumoral injections (229). Thus, a HAC-PD1-IgG chimeric protein (haPD1-IgG, thereafter) can retain HAC-PD1 activity in tumors with a prolonged half-life and enhanced efficacy. Since IgG has a long half-life, use of the chimeric fusion protein can reduce the need for frequent administration. We therefore generated plasmids pG8R382 (Figure 18A), pG8R383 (Figure 18B) and pG8R384 (Figure 18C) carrying a sequence encoding haPD1-IgG. To construct plasmid pG8R382 (Figure 18A), we used plasmid pCMV3-haPD1- IgG (Sinobiological, Project number BWH2-P) as a template to amplify the gene encoding haPD1-IgG using primers haPD1IgG-KpnI-s and haPD1IgG-NotI-a. The gene was inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R382. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the CCTS cell surface to target bladder cancer and after lysis of the CCTS cell in tumor cells employs the Pcmv promoter to express the haPD1-IgG to prevent inactivation of T cells. To construct plasmid pG8R383 (Figure 18B), we used plasmid pCMV3-haPD1- IgG (Sinobiological, Project number BWH2-P) as a template to amplify the gene encoding haPD1-IgG using primers and haPD1IgG-C-P2A-a. Then with plasmid pCS2- NXE+mem-KillerRed (Addgene plasmid # 45761) as a template, the gene encoding KillerRed mem was amplified using primers P2A-KillerRed-Mem-s and KillerRed- NotIXhoI-a and then extended by PCR using primers P2A-gs and KillerRed-NotIXhoI-a. The two fragments were then inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R383. The lysis plasmid carries genes encoding haPD1-IgG and KillerRed mem separated by the P2A peptide under the control of the Pcmv promoter to enable synthesis of haPD1-IgG and KillerRed mem in tumor cells invaded by the CCTS strain. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express synthesis of haPD1-IgG to prevent inactivation of T cells and KillerRed mem to kill the tumor cells. To construct plasmid pG8R384 (Figure 18C), we used plasmid pG8R343 (Figure 10A) as a template to amplify the gene encoding KillerRed mem using primers KillerRed-Mem-KpnI-s and KillerRed-C-P2A-a2. And then with plasmid pCMV3-haPD1- IgG (Sinobiological, Project number BWH2-P) as a template, the gene encoding haPD1- IgG was amplified with primers P2A-haPD1IgG-s and haPD1IgG-NotI-a. The two fragments were then inserted into plasmid pG8R320 (Figure 5B) cut with KpnI/NotI to generate plasmid pG8R384. The lysis plasmid carries genes encoding KillerRed mem and haPD1-IgG separated by P2A peptide under the control of a Pcmv promoter to enable synthesis of KillerRed mem and haPD1-IgG. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩplz4 in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express KillerRed mem to kill the tumor cell and haPD1-IgG to prevent the inactivation of T cells. Note that the only difference between pG8R343 and pG8R344 is the order of the sequences encoding the synthesis of haPD1-IgG and KillerRed. Example 16. Insertion of the nucleotide sequence encoding the single-chain fragment variable (scFv) targeting HER2 into the third exposed loop of the S. Typhimurium ompA gene to cause CCTS strains to target bladder, prostate and breast tumors. Human epidermal growth factor receptor 2 (HER2) is overexpressed in bladder, gastric, prostate and breast cancers (239-242, 245). Single-chain fragment variables (scFv, ~25kDa) penetrate tumors better than large IgG (246) and have faster clearance rates from the circulation to provide greater efficacy (247, 248). The scFv targets Her2 to bind to ErB2+ cells to potentiate delivery of exogenous DNA and siRNA into ErB2+ cells (249-251). We thus generated regulated delayed lysis plasmids pG8R385 (Figure 19A) and pG8R386 (Figure 19B) that have a pUC ori to synthesize scFv targeting Her2 on the Salmonella cell surface to target attaching to and invading bladder, gastric, prostate and breast cancers and after lysis of the CCTS cell in cancer cells employs the Pcmv promoter to drive expression of desired gene sequences important for tumor therapy. To construct plasmid pG8R385 (Figure 19A), we used plasmid pG8R320 (Figure 5B) as a template to amplify a 536 bp fragment using primers Ptrc-BclI-s and OmpA-Her2-a and a 694 bp fragment using primers OmpA-Her2-s and ompAGFP- BstBI-a. And then with plasmid pACgp67B-Her2 (Addgene Plasmid #10794) as a template (251), the gene encoding Her2 scFv was amplified with primers Her2-s and Her2His-a. With plasmid pYA4090 (Figure 2C) as a template, the gene encoding GFP was amplified with primer GFP-s and GFP-BstBI-a. The 4 fragments were inserted into plasmid pG8R320 cut with BclI/BstBI to generate plasmid pG8R385. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩHer2 scFv and gfp in Salmonella to display the synthesized OmpAΩHer2 scFv on the CCTS cell surface and GFP in the cytosol and after lysis of the CCTS cell in a cancer cell employs the Pcmv promoter to express an inserted gene sequence of importance for cancer cell/tumor therapy. To construct plasmid pG8R386 (Figure 19B), the plasmid pG8R385 was cut with BstBI to remove the gene encoding GFP. The 8.9 kb fragment was self-ligated to generate plasmid pG8R386. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩHer2 scFv in Salmonella to display the synthesized OmpAΩHer2 scFv on the cell surface and after lysis of the CCTS cell employs the Pcmv promoter to express an inserted gene sequence of importance for cancer cell/tumor therapy. Example 17. Construction of universal vaccine vectors to enable expression of both bacterial and eukaryotic genes by insertion of selected nucleotide sequences after the Ptrc or Ptrc bla SSopt promoter and the PCMV promoter, respectively. The regulated delayed lysis plasmid pG8R320 (Figure 5B) has a pUC ori and can express the bacterial gene ompAΩplz4 under the control of the Ptrc promoter in Salmonella to display the synthesized OmpAΩPLZ4 on the cell surface and after lysis of the cell employs the Pcmv promoter to express an inserted gene in eukaryotic cells. It is mainly used with Salmonella strains possessing the ∆ompA mutation and mainly targeting bladder cancer cells with PLZ4 peptide. This mutation may not be required in all situations. The targeting to bladder cancer cells limits its usage for other cancer cells. Thus, a universal vector without the OmpAΩPLZ4 can be used for other purposes, such as targeting other cancer types or as a dual antigen delivery system. We generated plasmids pG8R388 (Figure 19A) and pG8R389 (Figure 19B) as two universal vectors to enable their use to express and deliver proteins of both bacterial and eukaryotic origins. To construct pG8R388 (Figure 20A), we used plasmid pYA3342 (Figure 1A) as a template to amplify a 145 bp fragment containing the Ptrc promoter using primers Ptrc- BclI-s and 4545Ptrc-a. The fragment was inserted into plasmid pG8R320 (Figure 5B) cut with BclI/BstBI to generate plasmid pG8R388. The regulated delayed lysis plasmid has a pUC ori and can express bacterial genes in the Salmonella cytosol and after invasion into a eukaryotic cell and lysis can employ the Pcmv promoter to express the eukaryotic gene in animal cells. To construct pG8R389 (Figure 20B), we used plasmid pG8R114 (Figure 2B) as a template to amplify a 237 bp fragment containing the Ptrc promoter with an optimized bla secretion signal (SS) (68) using primers Ptrc-BclI-s and 4545PtrcBlaAAA-a . The fragment was inserted into plasmid pG8R320 (Figure 5B) cut with BclI/BstBI to generate plasmid pG8R389. The regulated delayed lysis plasmid has a pUC ori and can express bacterial gene fused with bla SSopt in Salmonella and secrete the synthesized protein into the periplasm and invasion into a eukaryotic cell and lysis can employ the Pcmv promoter to express the eukaryotic gene in animal cells. Example 18. Construction of dual plasmids to cause CCTS strains to target bladder tumor cells with LHRH peptide or HER2 scFv and then express a gene sequence encoding KillerRed to potentiate tumor cell killing. To validate the function of the LHRH peptide and HER2 scFv to target bladder tumor cells, we generated plasmids pG8R390 (Figure 21A), pG8R391 (Figure 21B) and pG8R418 (Figure 21C) with KillerRed mem to potentiate tumor cell killing. To construct pG8R390 (Figure 20A), the gene encoding KillerRed mem was cut from pG8R327 (Figure 8C) with KpnI/Not and cloned into plasmid pG8R381 (Figure 17B) cut with KpnI/NotI to generate plasmid pG8R390. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩlhrh in Salmonella to display the synthesized OmpAΩLHRH on the cell surface and after lysis of the CCTS cell in situ employs the Pcmv promoter to express the gene encoding KillerRed mem to potentiate tumor cell killing. To construct pG8R391 (Figure 20B), the gene encoding KillerRed mem was cut from pG8R327 (Figure 8C) with KpnI/Not and cloned into plasmid pG8R386 (Figure 19B) cut with KpnI/NotI to generate plasmid pG8R391. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩher2 scFv in Salmonella to display the synthesized OmpAΩHer2 scFv on the CCTS cell surface and after lysis of the CCTS cell after invading into a cancer cell employs the Pcmv promoter to express the gene encoding KillerRed mem to potentiate tumor cell killing. To construct pG8R391 (Figure 21C), the gene encoding KillerRed mem was cut from pG8R327 (Figure 8C) with KpnI/Not and cloned into plasmid pG8R385 (Figure 19A) cut with KpnI/NotI to generate plasmid pG8R418. The regulated delayed lysis plasmid has a pUC ori and can express ompAΩher2 scFv in Salmonella to display the synthesized OmpAΩHer2 scFv on the CCTS cell surface and after lysis of the CCTS cell after invading into a cancer cell employs the Pcmv promoter to express the gene encoding KillerRed mem to potentiate tumor cell killing. Example 19. Construction of recombinant plasmid CCTS strains with potential to attach to and invade into bladder tumor cells to deliver desired cargo to directly and/or indirectly reduce tumor survival. All of the candidate CCTS strains listed in Table 4 possess the ∆ompA11 mutation which can be substituted by a chromosomal ompAΩplz4 fusion allele and deletion of the asdA gene to enable establishment of a balanced-lethal vector-host strain after introduction of any of the recombinant plasmid vectors displayed in Figures 3 to 5 and 7 to 21, all of which encode synthesis of a receptor ligand facilitating specific targeting to bladder tumor cells. Most strains have the ∆PmurA25::TT araC ParaBAD murA mutation with a ∆asdA mutation to enable the display of the regulated delayed lysis in vivo phenotype that is desirable for delivery of cargoes synthesized by the CCTS strain and also for the delivery of plasmids serving as DNA vaccines to enable synthesis of a desired gene product by tumor cells. The inclusion of the relA mutation facilitates the completeness of lysis by uncoupling the dependance of growth on continued protein synthesis. Most CCTS strains in Table 4 display a means to cause a regulated delayed attenuation in vivo by cessation in the synthesis of the LPS outer core or the LPS O- antigen. This phenotype also enhances the efficiency of CCTS strains to attach to and invade eukaryotic cells and in this case better display the modified outer membrane protein OmpAΩPLZ4 that is the means for targeting specific attachment to bladder tumor cells. These CCTS strains also have a means to display a regulated delayed synthesis of Ptrc regulated gene insertions by the araC ParaBAD regulated expression of the lacI gene, an attribute that enhances the efficiency and frequency of in vivo colonization of the CCTS strains in target tissues. The presence of the ∆(wza-wcaM)-8 mutation facilitates complete lysis of strains with the regulated delayed lysis phenotype, enhances levels of plasmid encoded protein synthesis and precludes synthesis of exopolysaccharides that contribute to biofilm formation. Most strains have a ∆recF mutation to reduce inter- and intra-plasmid recombination to enhance construct stability and a ∆endA mutation to eliminate the endonuclease that could degrade the plasmid vector upon lysis of the CCTS cell. Also present, is a ∆sifA mutation that enables the CCTS strain after invasion into a cell to escape the Salmonella containing vesicle (SCV) or endosome. This is important for release of DNA vaccines by CCTS cells since the DNA vaccine must be free to be directed to the tumor cell nucleus to enable transcription of the inserted gene sequence under the control of the plasmid encoded PCMV or PEF1α promoter. In this regard, the DNA vaccine components of all the plasmids containing sequence from pYA4545 (Figure 1C) contain multiple sequences that direct the plasmid DNA to the cell nucleus (45). Since Salmonella infection into host cells induces pyroptosis that acts to destroy the nuclear organization and function, it is important that CCTS cells delivering DNA vaccines possess mutations such as the ∆sseL and ∆tlpA mutations to delay onset of pyroptosis and thus enhance expression of DNA vaccine encoded genes. Thus, many of the strains listed in Table 4 have such mutations and these mutations can be added to other strains using the suicide vectors listed in Table 2. Some of the strains listed in Table 4 have deletion mutations in the pagL, pagP, lpxR, eptA and arnT genes that alter the structure and activities of the LPS lipid A. These mutations may or may not contribute to the efficacy of CCTS constructs by altering the degree of inflammation in interacting with TLR4. Further modification of these activities can be accomplished by inclusion of the ∆pagP81::Plpp lpxE and/or the ∆lpxR93::Plpp lpxF deletion-insertion mutations that cause expression of codon- optimized Francisella tularensis genes to delete the 1’ of 4’ phosphates from lipid A to render it non-toxic but retain ability to bind to and activate TLR4. Since many CCTS gene activities are regulated by the sugars arabinose and rhamnose that must be supplied during in vitro cultivation but are absent in animal tissues, the timing of shut off of sugar-regulated gene expression after CCTS cell entry into an animal (human) host can be modulated by whether the sugars supplied during in vitro growth and retained in the cells are or are not quickly metabolized. Thus the mutations ∆araBAD65::TT and ∆rhaBADSR515 are sometimes added to CCTS strains to delay shut off of the sugar regulated genes for several cell divisions. Additional refinements of strains can be achieved by inclusion of mutations such as ∆PtolR67::::TT araC ParaBAD tolR that acts to increase production and release of outer membrane vesicles and other mutations that alter display or non-display of flagellar and fimbrial appendages or component parts to alter recruitment of innate immunity. These activities are well described in WO 2020/096994 A1 and WO 2021/222696 A1. Example 20. Evaluation of CCTS constructs for ability to attach to and invade bladder tumor cells. CCTS strains with pG8R341 specifying synthesis of GFP by the CCTS strain and pG8R342 in which the EGFP activity must be synthesized by the bladder tumor cell after invasion by the CCTS strain. Based on the discussion in Example 19, we will be comparing constructs in χ12417 (∆PmurA25::TT araC PBAD murA ∆waaL46 ∆pmi-2426 ∆asdA27::TT araC PBAD c2 ∆pagL64::TT rhaRS PrhaBAD waaL ∆(wza-wcaM)-8 ∆relA197::araC PBAD lacI TT ∆recF126 ∆sifA26 ∆ompA11) that was used for our initial work and the much improved strains χ12735 (∆ PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆pagL38::TT rhaRS PrhaBAD2 waaL2 ∆(wza-wcaM)-8 ∆relA1123 ∆recF126 ∆sifA26 ∆endA2113 ∆sseL116 ∆tlpA181 ∆rhaBADSR515 ∆araBAD65::TT ∆ompA11) and χ12736 (∆PmurA25::TT araC ParaBAD murA ∆asdA33 ∆waaL46 ∆pagL64::TT rhaRS PrhaBAD1 waaL1 ∆(wza-wcaM)-8 ∆relA1123 ∆recF126 ∆sifA26 ∆endA2113 ∆sseL116 ∆tlpA181 ∆rhaBADSR515 ∆araBAD65::TT ∆ompA11) (see Table 4). Strains will be grown in LB broth with 0.1% arabinose and with and without 0.1% rhamnose and evaluated for ability to attach to and invade bladder tumor cells as described in Example 1 and Figure 6. It is expected based on the information provided in forgoing Examples that strains grown without rhamnose to prevent synthesis of the LPS O-antigen will be most proficient in attaching to bladder tumor cells. It is also expected that CCTS strains χ12735 and χ12736 will be most proficient in inducing synthesis of EGFP. Example 21. Exemplary sequences Sequences and SEQ ID NOs related to embodiments described herein are provided infra before the references. Example 22. Evaluation of the effect of O-antigen on the CCTS constructs for ability to attach to and invade bladder tumor cells. Clinical trials showed only 3 out of 25 patients had Salmonella colonization at the tumor sites after intravenous injection (10), indicating that targeting efficiency of Salmonella should be increased. The OmpAΩPLZ4 fusion enables Salmonella displaying a bladder cancer targeting peptide on the surface of Salmonella to target bladder cancer cells. Lipopolysaccharide (LPS) is the structure that covers the Salmonella surface. Modifications of LPS in Salmonella can potentially affect Salmonella tumor targeting. LPS comprises Kdo-lipid A, inner core, outer core and O-antigen side chain (252). Kdo-lipid A is essential to the survival of Salmonella. The LPS mutations, ΔwaaL46, ΔwaaG42, and ΔwaaC41, which enable Salmonella to display defects in the synthesis of O-antigen, outer core and inner core, respectively, were introduced into Salmonella strains. It should be noted that waaC mutant defective in synthesis of the inner core are unable to synthesize and assemble the outer core and O-antigen whereas mutants unable to synthesize the outer LPS core also unable to display the O-antigen. These mutations were introduced into χ12614 to yield strain χ12812, χ12813, and χ12814, respectively (Table 5). A plasmid pG8R341 carrying multiple copies of ompAΩplz4 was introduced into these strains. PCR with correspondent primers proved that strains derived from χ12614 have the correct expected genotype and the LPS gel proved that each strain has the right LPS phenotype human bladder cancer cell 5637 and mouse bladder cancer cells MB49 and BBN967 were performed in 24-well culture plates as described previously (152). The χ12614 lineage strains were grown in LB media until OD600 reached 0.85~0.9. The bacteria were collected and resuspended in DMEM media with 10% fetal bovine serum. A MOI 10:1 was used to infect cells for 1 hour. After infection, half of the monolayers were washed with PBS and lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of attached bacteria. The other half of cells was incubated for 1 h with DMEM media containing 100 ug/ml gentamicin to eliminate extracellular bacteria. Monolayers were then lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of internalized bacteria. As shown in Figure 22C, strain χ12812 with ΔwaaL46 mutation displays the highest abilities in attachment and invasion compared to the other strains abilities to attach to and invade both human and mouse bladder cells. The LPS mutations, ΔwaaL46, ΔwaaG42, and ΔwaaC41 were also introduced into strain χ12619 to generate χ12808, χ12809, and χ12810, respectively. The ompAΩplz4 mutation (Table 2) was also introduced into strain χ12542 (Table 5) to yield strain χ12811 which has the identical genotypes as strain χ12810. These strains only have one copy of ompAΩplz4 in the chromosome. PCR with correspondent primers proved that strains derived from χ12619 have the correct expected genotype and the LPS gel proved that each strain had the right LPS phenotype (Figure 23 A and B) . All the strains grew on LB with arabinose plates, but not not on LB, LB with DAP and LB with alanine plates (Figure 23C). The evaluation of the abilities of Salmonella cells to attach to and invade into human bladder cancer cell 5637 and mouse bladder cancer cells MB49 and BBN967 were performed in 24-well culture plates as described previously (152). The χ12619 lineage strains were grown in LB media with 0.1% arabinose until OD600 reaches 0.85~0.9. The bacteria were collected and resuspended in DMEM media with 10% fetal bovine serum. A MOI of 10:1 was used to infect cells for 1 hour. After infection, half of the monolayers were washed with PBS and lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of attached bacteria. The other half of the monolayers was incubated for 1 h with DMEM media containing 100 ug/ml gentamicin to eliminate extracellular bacteria. Monolayers were then lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of internalized bacteria. As shown in Figure 23C, strain χ12809 with ΔwaaG46 mutation displayed the highest abilities in attachment, however, strain χ12808 with ΔwaaL46 displays highest abilities of invasion to both human and mouse bladder cells. Example 24. Display of OmpAΩHer2 ScFV on the bacterial surface enables S. Typhimurium cells to preferentially attach to tumor cells with higher Her2 production levels. To evaluate the surface display of OmpAΩHer2 ScFV, strain χ12417 was transformed with plasmids pG8R385, pG8R418 and pG8R391 that carry the ompAΩher2 ScFV gene. The strains were grown in LB with 0.1% arabinose.1 mM IPTG was added to induce the production of OmpAΩHer2 ScFV for 4 hours. The Salmonella outer member proteins (SOMPs) were prepared as described previously (62). The OmpAΩHer2 in strain χ12417 carrying any of the above plasmids can be detected in SOMPs portion of the SDS PAGE gel by Coomassie blue staining and western blot using anti-His6 antibody as an expected band around 65.1 kDa, but not the SOMPs from strain χ12417 (with no plasmid) (Figure 24). Different cell lines have different HER2 expression status (253). Her2 overexpression (SKBR-3, ATCC®HTB30) and low expression cell lines (MDA-MB- 231(ATCC® CRM-HTB-26) and MDA-MB-468 (ATCC® HTB-132)) were used to detect the attachment and invasion of the χ12417(pG8R385) and χ12417(pG8R391) strains (Figure 25). Overnight cultures of χ12417(pG8R385) and χ12417(pG8R391) were diluted into LB broth with 0.1% arabinose and grown until OD600 reached 0.9. The bacteria were washed once with PBS and then used to infect SKBR-3, MDA-MB-431 and MDA-MB-468 breast cancer cells at an MOI 10:1 for 1 hour. After infection, half of the monolayers were washed with PBS and lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of attached bacteria. The other half of the monolayers were treated for 1 h with DMEM media containing 100 ug/ml gentamicin to eliminate extracellular bacteria. Monolayers were then lysed with PBS containing 0.1% sodium deoxycholate to assess the total number of internalized bacteria. The strain χ12417(pG8R385) and χ12417(pG8R391) producing OmpAΩPLZ4 attached to and invaded to the highest levels the Her2 overexpression cell line SKBR-3, but not to the Her2 low expression cell lines MDA-MB-431 and MDA-MB-468. Example 25. KillerRed kills HEK293T cells. HEK293T cells were transfected with plasmid pG8R327 (Figure 8C). The fluorescence was observed using EVOS Fl with RFP channel (Thermofisher). The RFP channel in EVOS Fl has 531/40 nm excitation and 593/40 nm émission. KillerRed can absorb 540~580 nm wavelength light and emit an longer 610 nm red light with maximum fluorescence excitation/emission at 585/610 nm. Although the RFP channel is not the optimal wavelength for KillerRed, strong red fluorescence signals were observed in HEK293T cells transfected with plasmid pG8R327 (Figure 26). Before RFP channel excitation, the cells showed spindle shapes with smooth surfaces. After excitation for 10 min, membrane blebs appear on the surface of the cells (Figure 27). The phenomena was also observed in other samples. Further excitation for an additional 10 min led to the cell morphology changing to round instead of spindle shaped (Figure 28). This observation is at lower level but consistent with previous reports (170, 173, 175). Example 26. Evaluation of the CCTS strains with targeting peptide in vivo. The safety of CCTS strains uses oral and intravenous routes. Groups of 5 mice are inoculated with CCTS strains at varying doses ranging from 104 to 109 CFU. The mice are closely monitored for one month to see if any disease symptoms are observed. Studies are conducted using multiple subcutaneous syngeneic tumor models to evaluate the distribution of CCTS strains based on the specific targeting peptide utilized. To establish the subcutaneous syngeneic tumor, approximately 1-5x106 tumor cells are injected into the right flank of 6-8 week old mice. When tumor sizes reach approximately 100 mm3, the mice are administrated with the CCTS strains intravenously and orally using the highest safe dose determined from the previous experiment. At 24, 48, 72 and 96 hours, the spleen, liver, heart, lung, kidney, and tumors is harvested, weighted, homogenized and plated on LB agar with supplements. Fluorescence detection is performed on the tumors and organs to identify the presence of CCTS strains carry plasmids with the KillerRed or EGFP genes. To test the target ability of the CCTS strain displayed PLZ4 peptide, BBN963 or MB49 murine bladder cancer cells are injected into the right flank of C57BL/6 mice. Mice are treated with CCTS strains with plasmids carrying the ompA3Ωplz4 gene and monitored as above. To test the target ability of LHRH peptide, high LHRH receptor expression cell lines, such as A2780 human ovarian cancer cells (1-5x106) (235, 241, 243) or human breast cancer cells MCF-7 (ATCC® HTB-22) (254) , MDA-MB-231 (ATCC® HTB-26), HCC1806 (ATCC® CRL-2335) are used to generate xenographs in athymic nu/nu mice. Mice are treated with a CCTS strain with a plasmid specifying the LHRH peptide and monitored as above. To test the target ability of Her2 ScFV, Her2 overexpression (SKBR-3, ATCC®HTB30) and low expression cell lines (MDA-MB- 231(ATCC® CRM-HTB-26) and MDA-MB-468 (ATCC® HTB-132)), respectively, are used to generate xenographs in athymic nu/nu mice. Mice are treated with a CCTS strain with a plasmid specifying Her2 SCFV and monitored as above. Over 60% of bladder tumors have little immune cell infiltration inside tumors (255). CXCL11 is a cytokine that can attract CD8 cytotoxic T cells (256, 257). The abilities of CCTS strains delivering plasmids pG8R319 (pBR ori, ompAΩplz4), pG8R320 (pUC ori, ompA3Ωplz4), pG8R322 (pBR ori, ompA3Ωplz4, cxcl11) and pG8R326 (pUC ori, ompA3Ωplz4, cxcl11) are compared to convert an immune “cold” to immune “hot” tumor. When tumors in C57BL/6 mice reach around 100 mm3, mice are fed with PBS, CCTS(pG8R319), CCTS(pG8R320), CCTS(pG8R322), or CCTS(pG8R326). Tumors will be harvested 24, 48, 72 or 96 hours later. Flow cytometry is used to compare the amount of CD8+ cells in different groups at these time points. To further evaluate the abilities of CCTS strains to destroy tumors, C57BL6 mice carrying subcutaneous bladder tumors are used. When tumors reach the size of around 100 mm3, groups of 5 tumor-bearing mice are treated with a safe dose of CCTS strains with difference cargos, including CXCL11, haPD1, haPD1-IgG. The treatment could be once or multiple times. Mice are monitored for weight, tumor growth and survival. If tumors regress, mice are maintained and monitored to see if regrowth of tumors does or does not occur. Mice are euthanized once tumor sizes reach 1,500 mm3 or at the humane endpoint. Tumors are excised at the endpoint and either frozen directly in liquid nitrogen for storage or fixed in 10% formalin for histology or immunohistochemistry. In another experiment, tumors are collected before humane endpoint and split into 3 portions: formalin-fixation for immunohistochemical staining, fresh frozen for RNA and DNA extraction and deep sequencing; and single cell suspension for single cell sequencing. Based on the characterizations of all the plasmid constructs in the previous Examples, we expect to observe specific tumor cell targeting in ectopic tumors and the expression of the encoded payloads to exhibit the desired effects. Example 27. Evaluation of the CCTS strains with KillerRed in vivo KillerRed has been used in Photodynamic therapy (PDT) (258) (172). Mice carrying subcutaneous tumors are used. When tumors reach the size of around 100 mm3, tumor-bearing mice are treated with a safe dose of CCTS strains specifying synthesis of KillerRed. Fluorescence imaging of tumors are acquired daily in vivo using an IVIS-Spectrum (PerkinElmer, USA) with excitation wavelength of 570 nm and emission wavelength of 620 nm. A suitable wavelength is used for PDT. When tumors reach the size of around 100 mm3, tumor-bearing mice are treated with a safe dose of CCTS strains delivering KillerRed. Tumors are either treated with a continuous wave or pulsed laser without causing excessive temperature effect on the skin surface. A validation parameter is described by Shirmanova et .al (259). The PDT is carried at 593 nm, 150 mW/cm2, 270 J/cm2 for the continuous laser wave daily for 7 days, or at 584 nm, 225 mW/cm2, 337 J/cm2 for the pulsed laser on the days 6, 7, and 8 of tumor growth. Skin surface temperature is monitored using an Infrared thermograph. After the treatment, randomly selected tumors in treated and untreated groups is collected and split into 3 portions: formalin-fixation for immunohistochemical staining, fresh frozen for RNA and DNA extraction and deep sequencing; and single cell suspension for single cell sequencing. During these studies, tumors are monitored with the size measured with a caliper twice a week until the mice reach humane endpoint. Example 28. Universal vaccine vectors to enable expression of both bacterial and eukaryotic genes by insertion of selected nucleotide sequences after the Ptrc or Ptrc bla SSopt promoter and the PCMV promoter, respectively. A universal vaccine vector is a single vector that enables expression of genes both in prokaryotic and eukaryotic cells, even though it only specifies expression of gene only either in prokaryotic or eukaryotic cells. The regulated delayed lysis plasmids pG8R388 (Figure 19A) and pG8R389 (Figure 19B) are examples of two universal vectors to enable expression and delivery of proteins of both bacterial and eukaryotic origins. Plasmid pG8R388 has a pUC ori and can express bacterial genes in the cytosol or on the surface of Salmonella and after invasion into a eukaryotic cell and lysis can employ the Pcmv promoter to express the eukaryotic gene(s) in animal cells. Plasmid pG8R389 has a pUC ori and can express a bacterial gene fused with bla SSopt in Salmonella and secrete the synthesized protein into the periplasm and upon invasion into a eukaryotic cell and lysis can employ the Pcmv promoter to express the eukaryotic gene in animal cells. The prokaryotic promoter is not limited to Ptrc and could be any promoter that functions in prokaryotic cells. Other prokaryotic promoters can be used. Multiple prokaryotic promoters can be used to drive the expression of multiple genes. Other secretion signals can be used to replace the bla SSopt. The eukaryotic promoter is not limited to Pcmv or PEF1α and could be any promoter that functions in eukaryotic cells. Other eukaryotic promoters can be used. Multiple eukaryotic promoters can also be used to drive the expression of multiple genes. Under the control of a prokaryotic promoter, a single gene can be expressed and multiple genes can also be expressed as an operon or protein fusion with or without suitable linkers. These linkers could be flexible, rigid, cleavable or dipeptide linkers. Some examples are listed by Chen et.al (260-262). An unexhaustive list includes (GGGS)n, (GGGGS)n, (G)n, (EAAAK)n, (XP)n , GCT KESGSVSSEQLAQFRSLD, EGKSSGSGSESKST, and GSAGSAAGSGEF. Figure 17A illustrates plasmid pG8R380, in which an operon fusion links ompAΩlhrh and gfp. Figure 19A and Figure 21C illustrates plasmids pG8R385 and pG8R418, in which an operon fusion links ompAΩher2 SCFV and gfp. Under the control of a eukaryotic promoter, a single gene can be expressed and multiple genes can also be expressed separated by one or multiple 2A cleavage peptides. The 2A peptide could be P2A, E2A, T2A, F2A or other 2A-like sequences and thus form bi-, tri-, and quad-, penta- or multiple cistronic vectors (263-265). Plasmid pG8R343 (Fig.10), pG8R344 (Fig.10), pG8R383 (Fig.18), pG8R384 (Fig.18) are examples of bicistrion vectors with the P2A peptide. One or multiple IRES sequences could be used to generate multicistronic constructions for simultaneously expression of multiple genes (177, 179, 266-269). Multiple gene can be linked by a linker described above. Figure 14 and Figure 15B and 15C illustrate examples in which HAC PD1 is linked to CXCL11 through a linker. Figure 16B-D illustrate examples in which HAC PD1 is linked to EGFP through a 5A linker PPVAT.
Exemplary Sequences
Sequence of pG8R314 (SEQ ID NO: 134)
Red highlighted: ompA sequence (SEQ ID NO:1 (DNA) and SEQ ID NO: 2 (amino acid))
Green highlighted: PLZ4 peptide (SEQ ID NO:3 (DNA) and SEQ ID NO:4 (amino acid))
Cyan highlighted: Linker (SEQ. ID NO:5 (DNA) and SEQ. ID NO:6 (amino acid))
Yellow highlighted: asd sequence (SEQ. ID NO:7 (DNA) and SEQ ID NO:8 (amino acid))
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0002
Sequence of pG8R320 (SEQ ID NO: 135)
Red highlighted: ompA sequence (SEQ ID NO:1 (DNA) and SEQ ID NO: 2 (amino acid))
Green highlighted: PLZ4 peptide (SEQ ID NO:3 (DNA) and SEQ ID NO:4 (amino acid))
Cyan highlighted: Linker (SEQ ID NO:5 (DNA) and SEQ ID NO:6 (amino acid))
Yellow highlighted: asd sequence (SEQ. ID NO:7 (DNA) and SEQ ID NO:8 (amino acid))
Grey highlighted: murA sequence (SEQ ID NO: 9 (DNA) and SEQ ID NO: 10 (amino acid))
Pink highlighted: araC sequence (SEQ ID NO: 11 (DNA) and SEQ ID NO: 12 (amino acid))
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Human CXCL11 (SEQ ID NO: 13 (DNA) and SEQ ID NO:14 (amino acid))
Figure imgf000090_0002
Figure imgf000091_0001
Mouse CXCL11(SEQ ID NO: 15 (DNA) and SEQ ID NO:16(amino acid))
Figure imgf000091_0002
KillerRed-memo (SEQ ID NO: 17 (DNA) and SEQ ID NO: 18 (amino acid))
Figure imgf000091_0003
KillerRed-mlto (SEQ. ID NO: 19 (DNA) and SEQ ID NO: 20 (amino acid))
Figure imgf000091_0004
Figure imgf000092_0001
HLAB in pG8R345 (SEQ ID NO: 21 (DNA) and SEQ ID NO:22(amino acid))
Figure imgf000092_0002
OmpA-LHRH peptide (SEQ ID NO: 136) in pG8R380 and pG8R381 Green highlighted: LHRH peptide (SEQ ID NO: 23 (DNA) and SEQ ID NO:24 (amino acid))
Figure imgf000092_0003
Figure imgf000093_0001
LHRH peptide (SEQ ID NO:25)
EHWSYGLRPG
Figure imgf000093_0002
KillerRed memo-P2A-HumanCXCL11 (SEQ ID NO: 52)
Red highlighted: Neuromodulin N-terminal sequence (mem)(SEQ ID NO: SEQ ID NO: 26 (DNA) and SEQ ID NO: 27 (amino acid))
Cyan highlighted: KillerRed sequence (SEQ ID NO: 28 (DNA) and SEQ ID NO: 29 (amino acid))
Yellow highlighted: GSG P2A sequence (SEQ ID NO: 30 (DNA) and SEQ ID NO: 31 (amino acid))
Pink highlighted: Human CXCL11 sequence (SEQ ID NO: 32 (DNA) and SEQ ID NO: 33(amino acid))
Figure imgf000093_0003
Figure imgf000094_0002
KillerRed memo-P2A-Mouse CXCL11 (SEQ ID NO: 53)
Red highlighted: Neuromodulin N-terminal sequence (mem)(SEQ ID NO: 34 (DNA) and SEQ ID NO: 35(amino acid))
Cyan highlighted: KillerRed sequence (SEQ ID NO: 36 (DNA) and SEQ ID NO: 37 (amino acid))
Yellow highlighted: GSG P2A sequence (SEQ ID NO: 38 (DNA) and SEQ ID NO: 39(amino acid))
Green highlighted: Mouse CXCL11 sequence (SEQ ID NO: 40 (DNA) and SEQ ID NO: 41 (amino acid))
Figure imgf000094_0001
Figure imgf000095_0001
GSG P2A sequence (SEQ ID NO: 42 (DNA) and (SEQ iD NO: 43 (amine acid))
Figure imgf000095_0003
Her2 ScFV sequence (SEQ ID NO: 137)
Red highlighted: ompA sequence (mem)(SEQ ID NO: 44 (DNA) and SEQ ID NO: 45 (amino acid))
Green highlighted: Her2 ScFV sequence (SEQ ID NO: 46 (DNA) and SEQ ID NO: 47 (amino acid))
Figure imgf000095_0002
Figure imgf000096_0001
HAC-PD1 sequence (SEQ ID NO : 48(DNA) and SEQ ID NO: 49(amino acid)
Figure imgf000096_0002
Figure imgf000097_0001
haPD1-lgG sequence (SEQ ID NO: 50 (DNA) and SEQ ID NO: 51(arnino acid))
Figure imgf000097_0002
Sequence of pG8R388 (SEQ ID NO: 138)
Yellow highlighted: asd sequence (SEQ. ID NO:7 (DNA) and SEQ ID NO:8 (amino acid))
Grey highlighted: murA sequence (SEO. ID NO: 9 (DNA) and SEQ ID NO: 10 (amino acid))
Pink highlighted: araC sequence (SEQ ID NO: 11 (DNA) and SEQ ID NO: 12 (amino acid))
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Sequence of pG8R389 (SEQ ID NO: 139)
Red highlight: optimal bla sequence (SEQ ID NO: 127 (DNA) and SEQ ID NO: 128 (amino acid))
Yellow highlighted: asd sequence (SEQ ID NO:7 (DNA) and SEQ ID NO:8 (amino acid))
Grey highlighted: murA sequence (SEQ. ID NO: 9 (DNA) and SEQ ID NO: 10 (amino acid))
Pink highlighted: araC sequence (SEQ ID NO: 11 (DNA) and SEQ ID NO: 12 (amino acid))
Figure imgf000102_0002
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
GSG T2A sequence (derived from thoseaasigna virus 2A ) (SEQ. ID NO: 129
(amino acid))
Figure imgf000107_0003
GSG E2A sequence (derived from equine rhinitis A virus )(SEQ ID NO: 130
(amino acid))
Figure imgf000107_0002
GSG F2A sequence (derived from foot-and-mouth disease virus) (SEQ ID NO: 131. (amino acid))
Figure imgf000108_0002
Linker sequence in pG8R362-pG8R365 (SEQJD NO:132 (DNA)) (SEQ ID NO: 133 (amino acid))
Figure imgf000108_0001
References 1. Centers for Disease Control and Prevention NCfHS.2021. Underlying Cause of Death 1999-2020 on CDC WONDER Online Database, released in 2021. Data are from the Multiple Cause of Death Files, 1999-2020, as compiled from data provided by the 57 vital statistics jurisdictions through the Vital Statistics Cooperative Program. 2. Anonymous. William Coley. https://en.wikipedia.org/wiki/William_Coley. Accessed Aug 28. 3. Coley WB.1891. II. Contribution to the Knowledge of Sarcoma. Ann Surg 14:199-220. 4. Malmgren RA, Flanigan CC.1955. Localization of the vegetative form of Clostridium tetani in mouse tumors following intravenous spore administration. Cancer Res 15:473-478. 5. Forbes NS.2010. Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer 10:785-794. 6. Kohwi Y, Imai K, Tamura Z, Hashimoto Y.1978. Antitumor effect of Bifidobacterium infantis in mice. Gan 69:613-618. 7. Yu YA, Shabahang S, Timiryasova TM, Zhang Q, Beltz R, Gentschev I, Goebel W, Szalay AA.2004. Visualization of tumors and metastases in live animals with bacteria and vaccinia virus encoding light-emitting proteins. Nat Biotechnol 22:313-320. 8. Pan ZK, Weiskirch LM, Paterson Y.1999. Regression of established B16F10 melanoma with a recombinant Listeria monocytogenes vaccine. Cancer Res 59:5264-5269. 9. Wang CZ, Kazmierczak RA, Eisenstark A.2016. Strains, Mechanism, and Perspective: Salmonella-Based Cancer Therapy. Int J Microbiol 2016:5678702. 10. Toso JF, Gill VJ, Hwu P, Marincola FM, Restifo NP, Schwartzentruber DJ, Sherry RM, Topalian SL, Yang JC, Stock F, Freezer LJ, Morton KE, Seipp C, Haworth L, Mavroukakis S, White D, MacDonald S, Mao J, Sznol M, Rosenberg SA.2002. Phase I study of the intravenous administration of attenuated Salmonella typhimurium to patients with metastatic melanoma. J Clin Oncol 20:142-152. 11. Heimann DM, Rosenberg SA.2003. Continuous intravenous administration of live genetically modified Salmonella typhimurium in patients with metastatic melanoma. J Immunother 26:179-180. 12. Thamm DH, Kurzman ID, King I, Li Z, Sznol M, Dubielzig RR, Vail DM, MacEwen EG.2005. Systemic administration of an attenuated, tumor-targeting Salmonella typhimurium to dogs with spontaneous neoplasia: phase I evaluation. Clin Cancer Res 11:4827-4834. 13. Nemunaitis J, Cunningham C, Senzer N, Kuhn J, Cramm J, Litz C, Cavagnolo R, Cahill A, Clairmont C, Sznol M.2003. Pilot trial of genetically modified, attenuated Salmonella expressing the E. coli cytosine deaminase gene in refractory cancer patients. Cancer Gene Ther 10:737-744. 14. Luo Y, Kong Q, Yang J, Mitra A, Golden G, Wanda SY, Roland KL, Jensen RV, Ernst PB, Curtiss R, III.2012. Comparative genome analysis of the high pathogenicity Salmonella Typhimurium strain UK-1. PLoS One 7:e40645. 15. Sanapala S, Mosca L, Wang S, Curtiss R, III.2018. Comparative evaluation of Salmonella Typhimurium vaccines derived from UK-1 and 14028S: Importance of inherent virulence. PLoS One 13:e0203526. 16. Felgner S, Kocijancic D, Frahm M, Curtiss R, III, Erhardt M, Weiss S.2016. Optimizing Salmonella enterica serovar Typhimurium for bacteria-mediated tumor therapy. Gut Microbes 7:171-177. 17. Broadway KM, Modise T, Jensen RV, Scharf BE.2014. Complete genome sequence of Salmonella enterica serovar Typhimurium VNP20009, a strain engineered for tumor targeting. J Biotechnol 192 Pt A:177-178. 18. Broadway KM, Denson EA, Jensen RV, Scharf BE.2015. Rescuing chemotaxis of the anticancer agent Salmonella enterica serovar Typhimurium VNP20009. J Biotechnol 211:117-120. 19. Kong Q, Six DA, Liu Q, Gu L, Roland KL, Raetz CR, Curtiss R, III.2011. Palmitoylation state impacts induction of innate and acquired immunity by the Salmonella enterica serovar Typhimurium msbB mutant. Infect Immun 79:5027- 5038. 20. Kong Q, Six DA, Liu Q, Gu L, Wang S, Alamuri P, Raetz CR, Curtiss R, III. 2012. Phosphate groups of lipid A are essential for Salmonella enterica serovar Typhimurium virulence and affect innate and adaptive immunity. Infect Immun 80:3215-3224. 21. Kong Q, Six DA, Roland KL, Liu Q, Gu L, Reynolds CM, Wang X, Raetz CR, Curtiss R, III.2011. Salmonella synthesizing 1-dephosphorylated lipopolysaccharide exhibits low endotoxic activity while retaining its immunogenicity. J Immunol 187:412-423. 22. Kong Q, Yang J, Liu Q, Alamuri P, Roland KL, Curtiss R, III.2011. Effect of deletion of genes involved in lipopolysaccharide core and O-antigen synthesis on virulence and immunogenicity of Salmonella enterica serovar Typhimurium. Infect Immun 79:4227-4239. 23. Hoffman RM.2011. Tumor-seeking Salmonella amino acid auxotrophs. Curr Opin Biotechnol 22:917-923. 24. Zhao M, Yang M, Li XM, Jiang P, Baranov E, Li S, Xu M, Penman S, Hoffman RM. 2005. Tumor-targeting bacterial therapy with amino acid auxotrophs of GFP- expressing Salmonella typhimurium. Proc Natl Acad Sci U S A 102:755-760. 25. Zhao M, Yang M, Ma H, Li X, Tan X, Li S, Yang Z, Hoffman RM.2006. Targeted therapy with a Salmonella typhimurium leucine-arginine auxotroph cures orthotopic human breast tumors in nude mice. Cancer Res 66:7647-7652. 26. Nagakura C, Hayashi K, Zhao M, Yamauchi K, Yamamoto N, Tsuchiya H, Tomita K, Bouvet M, Hoffman RM.2009. Efficacy of a genetically-modified Salmonella typhimurium in an orthotopic human pancreatic cancer in nude mice. Anticancer Res 29:1873-1878. 27. Schmitz-Winnenthal FH, Hohmann N, Niethammer AG, Friedrich T, Lubenau H, Springer M, Breiner KM, Mikus G, Weitz J, Ulrich A, Buechler MW, Pianka F, Klaiber U, Diener M, Leowardi C, Schimmack S, Sisic L, Keller AV, Koc R, Springfeld C, Knebel P, Schmidt T, Ge Y, Bucur M, Stamova S, Podola L, Haefeli WE, Grenacher L, Beckhove P.2015. Anti-angiogenic activity of VXM01, an oral T-cell vaccine against VEGF receptor 2, in patients with advanced pancreatic cancer: A randomized, placebo-controlled, phase 1 trial. Oncoimmunology 4:e1001217. 28. Gniadek TJ, Augustin L, Schottel J, Leonard A, Saltzman D, Greeno E, Batist G.2020. A Phase I, Dose Escalation, Single Dose Trial of Oral Attenuated Salmonella typhimurium Containing Human IL-2 in Patients With Metastatic Gastrointestinal Cancers. J Immunother 43:217-221. 29. Selmic LE, Burton JH, Thamm DH, Withrow SJ, Lana SE.2014. Comparison of carboplatin and doxorubicin-based chemotherapy protocols in 470 dogs after amputation for treatment of appendicular osteosarcoma. J Vet Intern Med 28:554-563. 30. Fritz SE, Henson MS, Greengard E, Winter AL, Stuebner KM, Yoon U, Wilk VL, Borgatti A, Augustin LB, Modiano JF, Saltzman DA.2016. A phase I clinical study to evaluate safety of orally administered, genetically engineered Salmonella enterica serovar Typhimurium for canine osteosarcoma. Vet Med Sci 2:179-190. 31. Ganai S, Arenas RB, Forbes NS.2009. Tumour-targeted delivery of TRAIL using Salmonella typhimurium enhances breast cancer survival in mice. Br J Cancer 101:1683-1691. 32. Loeffler M, Le'Negrate G, Krajewska M, Reed JC.2008. Inhibition of tumor growth using Salmonella expressing Fas ligand. J Natl Cancer Inst 100:1113- 1116. 33. Ryan RM, Green J, Williams PJ, Tazzyman S, Hunt S, Harmey JH, Kehoe SC, Lewis CE.2009. Bacterial delivery of a novel cytolysin to hypoxic areas of solid tumors. Gene Ther 16:329-339. 34. Nguyen VH, Kim HS, Ha JM, Hong Y, Choy HE, Min JJ.2010. Genetically engineered Salmonella typhimurium as an imageable therapeutic probe for cancer. Cancer Res 70:18-23. 35. Saltzman DA, Heise CP, Hasz DE, Zebede M, Kelly SM, Curtiss R, III, Leonard AS, Anderson PM.1996. Attenuated Salmonella typhimurium containing interleukin-2 decreases MC-38 hepatic metastases: a novel anti-tumor agent. Cancer Biother Radiopharm 11:145-153. 36. Sorenson BS, Banton KL, Frykman NL, Leonard AS, Saltzman DA.2008. Attenuated Salmonella typhimurium with interleukin 2 gene prevents the establishment of pulmonary metastases in a model of osteosarcoma. J Pediatr Surg 43:1153-1158. 37. al-Ramadi BK, Fernandez-Cabezudo MJ, El-Hasasna H, Al-Salam S, Bashir G, Chouaib S.2009. Potent anti-tumor activity of systemically-administered IL2- expressing Salmonella correlates with decreased angiogenesis and enhanced tumor apoptosis. Clin Immunol 130:89-97. 38. Nagaraj S, Pisarev V, Kinarsky L, Sherman S, Muro-Cacho C, Altieri DC, Gabrilovich DI.2007. Dendritic cell-based full-length survivin vaccine in treatment of experimental tumors. J Immunother 30:169-179. 39. Bertino P, Panigada M, Soprana E, Bianchi V, Bertilaccio S, Sanvito F, Rose AH, Yang H, Gaudino G, Hoffmann PR, Siccardi A, Carbone M.2013. Fowlpox-based survivin vaccination for malignant mesothelioma therapy. Int J Cancer 133:612-623. 40. Srivastava AK, Sharma RK, Yolcu ES, Ulker V, MacLeod K, Dinc G, Shirwan H.2012. Prime-boost vaccination with SA-4-1BBL costimulatory molecule and survivin eradicates lung carcinoma in CD8+ T and NK cell dependent manner. PLoS One 7:e48463. 41. Ishizaki H, Manuel ER, Song GY, Srivastava T, Sun S, Diamond DJ, Ellenhorn JD.2011. Modified vaccinia Ankara expressing survivin combined with gemcitabine generates specific antitumor effects in a murine pancreatic carcinoma model. Cancer Immunol Immunother 60:99-109. 42. Curtiss R, III.2015. Antigen Delivery System II: Development of Live Attenuated Bacterial Vectors p1233-1269. In Mestecky J, Strober W, Russell MW, Kelsall BL, Cheroutre H, Lambrecht BN (ed), Mucosal Immunology (Fourth Edition) doi:http://dx.doi.org/10.1016/B978-0-12-415847-4.00064-1. Academic Press, Boston. 43. Curtiss R, III.2005. Antigen Delivery Systems II: Development of Live Recombinant Attenuated Bacterial Antigen and DNA Vaccine Delivery Vector Vaccines p1009-1037. In Mestecky J, Lamm ME, McGhee JR, Bienenstock J, Mayer L, Strober W (ed), Mucosal Immunology (Third Edition) doi:http://dx.doi.org/10.1016/B978-012491543-5/50060-7. Academic Press, Burlington. 44. Galen JE, Curtiss R, III.2014. The delicate balance in genetically engineering live vaccines. Vaccine 32:4376-4385. 45. Kong W, Brovold M, Koeneman BA, Clark-Curtiss J, Curtiss R, III.2012. Turning self-destructing Salmonella into a universal DNA vaccine delivery platform. Proc Natl Acad Sci U S A 109:19414-19419. 46. Curtiss R, III, Kelly SM, Tinge SA, Tacket CO, Levine MM, Srinivasan J, Koopman M.1994. Recombinant Salmonella vectors in vaccine development. Dev Biol Stand 82:23-33. 47. Curtiss R, III, Xin W, Li Y, Kong W, Wanda SY, Gunn B, Wang S.2010. New technologies in using recombinant attenuated Salmonella vaccine vectors. Crit Rev Immunol 30:255-270. 48. Kong W, Clark-Curtiss J, Curtiss R, III.2013. Utilizing Salmonella for antigen delivery: the aims and benefits of bacterial delivered vaccination. Expert Rev Vaccines 12:345-347. 49. Wang S, Kong Q, Curtiss R, III.2013. New technologies in developing recombinant attenuated Salmonella vaccine vectors. Microb Pathog 58:17-28. 50. Curtiss R, III., Wanda SY, Gunn BM, Zhang X, Tinge SA, Ananthnarayan V, Mo H, Wang S, Kong W.2009. Salmonella enterica serovar Typhimurium strains with regulated delayed attenuation in vivo. Infect Immun 77:1071-1082. 51. Li Y, Wang S, Scarpellini G, Gunn B, Xin W, Wanda SY, Roland KL, Curtiss R, III.2009. Evaluation of new generation Salmonella enterica serovar Typhimurium vaccines with regulated delayed attenuation to induce immune responses against PspA. Proc Natl Acad Sci U S A 106:593-598. 52. Juárez-Rodríguez MD, Yang J, Kader R, Alamuri P, Curtiss R, III, Clark- Curtiss JE.2012. Live attenuated Salmonella vaccines displaying regulated delayed lysis and delayed antigen synthesis to confer protection against Mycobacterium tuberculosis. Infect Immun 80:815-831. 53. Wang S, Li Y, Scarpellini G, Kong W, Shi H, Baek CH, Gunn B, Wanda SY, Roland KL, Zhang X, Senechal-Willis P, Curtiss R, III.2010. Salmonella vaccine vectors displaying delayed antigen synthesis in vivo to enhance immunogenicity. Infect Immun 78:3969-3980. 54. Wang S, Li Y, Shi H, Sun W, Roland KL, Curtiss R, III.2011. Comparison of a regulated delayed antigen synthesis system with in vivo-inducible promoters for antigen delivery by live attenuated Salmonella vaccines. Infect Immun 79:937- 949. 55. Xin W, Wanda SY, Li Y, Wang S, Mo H, Curtiss R, III.2008. Analysis of type II secretion of recombinant pneumococcal PspA and PspC in a Salmonella enterica serovar Typhimurium vaccine with regulated delayed antigen synthesis. Infect Immun 76:3241-3254. 56. Shi H, Wang S, Curtiss R, III.2013. Evaluation of regulated delayed attenuation strategies for Salmonella enterica serovar Typhi vaccine vectors in neonatal and infant mice. Clin Vaccine Immunol 20:931-944. 57. Shi H, Wang S, Roland KL, Gunn BM, Curtiss R, III.2010. Immunogenicity of a live recombinant Salmonella enterica serovar Typhimurium vaccine expressing pspA in neonates and infant mice born from naive and immunized mothers. Clin Vaccine Immunol 17:363-371. 58. Ehretsmann CP, Carpousis AJ, Krisch HM.1992. Specificity of Escherichia coli endoribonuclease RNase E: in vivo and in vitro analysis of mutants in a bacteriophage T4 mRNA processing site. Genes Dev 6:149-159. 59. McDowall KJ, Kaberdin VR, Wu SW, Cohen SN, Lin-Chao S.1995. Site- specific RNase E cleavage of oligonucleotides and inhibition by stem-loops. Nature 374:287-290. 60. Lin-Chao S, Wong TT, McDowall KJ, Cohen SN.1994. Effects of nucleotide sequence on the specificity of rne-dependent and RNase E-mediated cleavages of RNA I encoded by the pBR322 plasmid. J Biol Chem 269:10797-10803. 61. Kang HY, Curtiss R, III.2003. Immune responses dependent on antigen location in recombinant attenuated Salmonella typhimurium vaccines following oral immunization. FEMS Immunol Med Microbiol 37:99-104. 62. Kang HY, Srinivasan J, Curtiss R, III.2002. Immune responses to recombinant pneumococcal PspA antigen delivered by live attenuated Salmonella enterica serovar Typhimurium vaccine. Infect Immun 70:1739-1749. 63. Konjufca V, Jenkins M, Wang S, Juárez-Rodríguez MD, Curtiss R, III.2008. Immunogenicity of recombinant attenuated Salmonella enterica serovar Typhimurium vaccine strains carrying a gene that encodes Eimeria tenella antigen SO7. Infect Immun 76:5745-5753. 64. Muralinath M, Kuehn MJ, Roland KL, Curtiss R, III.2011. Immunization with Salmonella enterica serovar Typhimurium-derived outer membrane vesicles delivering the pneumococcal protein PspA confers protection against challenge with Streptococcus pneumoniae. Infect Immun 79:887-894. 65. Kong W, Wanda SY, Zhang X, Bollen W, Tinge SA, Roland KL, Curtiss R, III. 2008. Regulated programmed lysis of recombinant Salmonella in host tissues to release protective antigens and confer biological containment. Proc Natl Acad Sci U S A 105:9361-9366. 66. Ashraf S, Kong W, Wang S, Yang J, Curtiss R, III.2011. Protective cellular responses elicited by vaccination with influenza nucleoprotein delivered by a live recombinant attenuated Salmonella vaccine. Vaccine 29:3990-4002. 67. Ameiss K, Ashraf S, Kong W, Pekosz A, Wu WH, Milich D, Billaud JN, Curtiss R, III.2010. Delivery of woodchuck hepatitis virus-like particle presented influenza M2e by recombinant attenuated Salmonella displaying a delayed lysis phenotype. Vaccine 28:6704-6713. 68. Jiang Y, Mo H, Willingham C, Wang S, Park JY, Kong W, Roland KL, Curtiss R, III.2015. Protection Against Necrotic Enteritis in Broiler Chickens by Regulated Delayed Lysis Salmonella Vaccines. Avian Dis 59:475-485. 69. Germanier R, Fürer E.1971. Immunity in experimental salmonellosis. II. Basis for the avirulence and protective capacity of gal E mutants of Salmonella typhimurium. Infect Immun 4:663-673. 70. Collins LV, Attridge S, Hackett J.1991. Mutations at rfc or pmi attenuate Salmonella typhimurium virulence for mice. Infect Immun 59:1079-1085. 71. Frey SE, Lottenbach KR, Hill H, Blevins TP, Yu Y, Zhang Y, Brenneman KE, Kelly-Aehle SM, McDonald C, Jansen A, Curtiss R, III.2013. A Phase I, dose- escalation trial in adults of three recombinant attenuated Salmonella Typhi vaccine vectors producing Streptococcus pneumoniae surface protein antigen PspA. Vaccine 31:4874-4880. 72. Gunn BM, Wanda SY, Burshell D, Wang C, Curtiss R, III.2010. Construction of recombinant attenuated Salmonella enterica serovar Typhimurium vaccine vector strains for safety in newborn and infant mice. Clin Vaccine Immunol 17:354-362. 73. Wang S, Hofacre CL, Wanda SY, Zhou J, Callum RA, Nordgren B, Curtiss R, III.2022. A triple-sugar regulated Salmonella vaccine protects against Clostridium perfringens-induced necrotic enteritis in broiler chickens. Poult Sci 101:101592. 74. Bentley R.1990. The shikimate pathway--a metabolic tree with many branches. Crit Rev Biochem Mol Biol 25:307-384. 75. Galán JE, Nakayama K, Curtiss R, III.1990. Cloning and characterization of the asd gene of Salmonella typhimurium: use in stable maintenance of recombinant plasmids in Salmonella vaccine strains. Gene 94:29-35. 76. Black S, Wright NG.1955. Aspartic β-semialdehyde dehydrogenase and aspartic β-semialdehyde. J Biol Chem 213:39-50. 77. Nakayama K, Kelly SM, Curtiss R, III 1988. Construction of an Asd+ expression-cloning Vector: stable maintenance and high level expression of cloned genes in a Salmonella vaccine strain. Nat Biotech 6:693-697. 78. Xin W, Wanda SY, Zhang X, Santander J, Scarpellini G, Ellis K, Alamuri P, Curtiss R, III.2012. The Asd+-DadB+ dual-plasmid system offers a novel means to deliver multiple protective antigens by a recombinant attenuated Salmonella vaccine. Infect Immun 80:3621-3633. 79. Curtiss R, III. Apr.15, 20032012. Regulated attenuation of live vaccines to enhance cross-protective immunogenicity. US patent 8,133,493. 80. Lin HC, Lei SP, Studnicka G, Wilcox G.1985. The araBAD operon of Salmonella typhimurium LT2. III. Nucleotide sequence of araD and its flanking regions, and primary structure of its product, L-ribulose-5-phosphate 4- epimerase. Gene 34:129-134. 81. Lin HC, Lei SP, Wilcox G.1985. The araBAD operon of Salmonella typhimurium LT2. II. Nucleotide sequence of araA and primary structure of its product, L- arabinose isomerase. Gene 34:123-128. 82. Lin HC, Lei SP, Wilcox G.1985. The araBAD operon of Salmonella typhimurium LT2. I. Nucleotide sequence of araB and primary structure of its product, ribulokinase. Gene 34:111-122. 83. Lee JH, Nishitani J, Wilcox G.1984. Genetic characterization of Salmonella typhimurium LT2 ara mutations. J Bacteriol 158:344-346. 84. Horwitz AH, Heffernan L, Morandi C, Lee JH, Timko J, Wilcox G.1981. DNA sequence of the araBAD-araC controlling region in Salmonella typhimurium LT2. Gene 14:309-319. 85. Moralejo P, Egan SM, Hidalgo E, Aguilar J.1993. Sequencing and characterization of a gene cluster encoding the enzymes for L-rhamnose metabolism in Escherichia coli. J Bacteriol 175:5585-5594. 86. Egan SM, Schleif RF.1993. A regulatory cascade in the induction of rhaBAD. J Mol Biol 234:87-98. 87. Lee H, Hsu FF, Turk J, Groisman EA.2004. The PmrA-regulated pmrC gene mediates phosphoethanolamine modification of lipid A and polymyxin resistance in Salmonella enterica. J Bacteriol 186:4124-4133. 88. Trent M, Raetz C.2002. Cloning of EptA, the lipid A phosphoethanolamine transferase associated with polymyxin resistance. J Endotoxin Res 8:159. 89. Trent MS, Ribeiro AA, Lin S, Cotter RJ, Raetz CR.2001. An inner membrane enzyme in Salmonella and Escherichia coli that transfers 4-amino-4-deoxy-L- arabinose to lipid A: induction on polymyxin-resistant mutants and role of a novel lipid-linked donor. J Biol Chem 276:43122-43131. 90. Iino T, Komeda Y, Kutsukake K, Macnab RM, Matsumura P, Parkinson JS, Simon MI, Yamaguchi S.1988. New unified nomenclature for the flagellar genes of Escherichia coli and Salmonella typhimurium. Microbiol Rev 52:533- 535. 91. Iino T.1977. Genetics of structure and function of bacterial flagella. Annu Rev Genet 11:161-182. 92. Iino T.1969. Genetics and chemistry of bacterial flagella. Bacteriol Rev 33:454- 475. 93. Liu Q, Liu Q, Yi J, Liang K, Hu B, Zhang X, Curtiss R, III, Kong Q.2016. Outer membrane vesicles from flagellin-deficient Salmonella enterica serovar Typhimurium induce cross-reactive immunity and provide cross-protection against heterologous Salmonella challenge. Sci Rep 6:34776. 94. Zieg J, Simon M.1980. Analysis of the nucleotide sequence of an invertible controlling element. Proc Natl Acad Sci U S A 77:4196-4200. 95. Silverman M, Simon M.1980. Phase variation: genetic analysis of switching mutants. Cell 19:845-854. 96. Silverman M, Zieg J, Simon M.1979. Flagellar-phase variation: isolation of the rh1 gene. J Bacteriol 137:517-523. 97. Kutsukake K, Iino T.1980. Inversions of specific DNA segments in flagellar phase variation of Salmonella and inversion systems of bacteriophages P1 and Mu. Proc Natl Acad Sci U S A 77:7338-7341. 98. Bonifield HR, Hughes KT.2003. Flagellar phase variation in Salmonella enterica is mediated by a posttranscriptional control mechanism. J Bacteriol 185:3567-3574. 99. Kutsukake K, Iino T.1980. A trans-acting factor mediates inversion of a specific DNA segment in flagellar phase variation of Salmonella. Nature 284:479-481. 100. Yamamoto S, Kutsukake K.2006. FljA-mediated posttranscriptional control of phase 1 flagellin expression in flagellar phase variation of Salmonella enterica serovar Typhimurium. J Bacteriol 188:958-967. 101. Römling U, Bian Z, Hammar M, Sierralta WD, Normark S.1998. Curli fibers are highly conserved between Salmonella typhimurium and Escherichia coli with respect to operon structure and regulation. J Bacteriol 180:722-731. 102. Łaniewski P, Baek CH, Roland KL, Curtiss R, III.2017. Analysis of Spleen- Induced Fimbria Production in Recombinant Attenuated Salmonella enterica Serovar Typhimurium Vaccine Strains. MBio 8:e01189-01117. 103. Lockman HA, Curtiss R, III.1992. Isolation and characterization of conditional adherent and non-type 1 fimbriated Salmonella typhimurium mutants. Mol Microbiol 6:933-945. 104. Thankavel K, Shah AH, Cohen MS, Ikeda T, Lorenz RG, Curtiss R, III, Abraham SN.1999. Molecular basis for the enterocyte tropism exhibited by Salmonella typhimurium type 1 fimbriae. J Biol Chem 274:5797-5809. 105. Freudl R, Cole ST.1983. Cloning and molecular characterization of the ompA gene from Salmonella typhimurium. Eur J Biochem 134:497-502. 106. Norris FA, Wilson MP, Wallis TS, Galyov EE, Majerus PW.1998. SopB, a protein required for virulence of Salmonella dublin, is an inositol phosphate phosphatase. Proc Natl Acad Sci U S A 95:14057-14059. 107. Reis BP, Zhang S, Tsolis RM, Baumler AJ, Adams LG, Santos RL.2003. The attenuated sopB mutant of Salmonella enterica serovar Typhimurium has the same tissue distribution and host chemokine response as the wild type in bovine Peyer's patches. Vet Microbiol 97:269-277. 108. Zhang S, Santos RL, Tsolis RM, Stender S, Hardt WD, Baumler AJ, Adams LG.2002. The Salmonella enterica serotype typhimurium effector proteins SipA, SopA, SopB, SopD, and SopE2 act in concert to induce diarrhea in calves. Infect Immun 70:3843-3855. 109. Goncharoff P, Nichols BP.1988. Evolution of aminobenzoate synthases: nucleotide sequences of Salmonella typhimurium and Klebsiella aerogenes pabB. Mol Biol Evol 5:531-548. 110. Kaplan JB, Merkel WK, Nichols BP.1985. Evolution of glutamine amidotransferase genes. Nucleotide sequences of the pabA genes from Salmonella typhimurium, Klebsiella aerogenes and Serratia marcescens. J Mol Biol 183:327-340. 111. Schnaitman CA, Klena JD.1993. Genetics of lipopolysaccharide biosynthesis in enteric bacteria. Microbiol Rev 57:655-682. 112. Kadrmas JL, Raetz CR.1998. Enzymatic synthesis of lipopolysaccharide in Escherichia coli. Purification and properties of heptosyltransferase i. J Biol Chem 273:2799-2807. 113. Klena JD, Ashford RS, 2nd, Schnaitman CA.1992. Role of Escherichia coli K- 12 rfa genes and the rfp gene of Shigella dysenteriae 1 in generation of lipopolysaccharide core heterogeneity and attachment of O antigen. J Bacteriol 174:7297-7307. 114. Heinrichs DE, Yethon JA, Whitfield C.1998. Molecular basis for structural diversity in the core regions of the lipopolysaccharides of Escherichia coli and Salmonella enterica. Mol Microbiol 30:221-232. 115. Stevenson G, Andrianopoulos K, Hobbs M, Reeves PR.1996. Organization of the Escherichia coli K-12 gene cluster responsible for production of the extracellular polysaccharide colanic acid. J Bacteriol 178:4885-4893. 116. Whitfield C.2006. Biosynthesis and assembly of capsular polysaccharides in Escherichia coli. Annu Rev Biochem 75:39-68. 117. Pizarro-Cerda J, Tedin K.2004. The bacterial signal molecule, ppGpp, regulates Salmonella virulence gene expression. Mol Microbiol 52:1827-1844. 118. Török I, Kari C.1980. Accumulation of ppGpp in a relA mutant of Escherichia coli during amino acid starvation. J Biol Chem 255:3838-3840. 119. Curtiss R, III, Wang S, Wanda S-Y, Kong W. Feb .10 , 20162018. Regulated expression of antigen and/or regulated attenuation to enhance vaccine immunogenicity and/or safety patent US 9 , 885 , 051 B2. 120. Ahmer BM, Tran M, Heffron F.1999. The virulence plasmid of Salmonella typhimurium is self-transmissible. J Bacteriol 181:1364-1368. 121. Na HS, Kim HJ, Lee HC, Hong Y, Rhee JH, Choy HE.2006. Immune response induced by Salmonella typhimurium defective in ppGpp synthesis. Vaccine 24:2027-2034. 122. Rudd KE, Bochner BR, Cashel M, Roth JR.1985. Mutations in the spoT gene of Salmonella typhimurium: effects on his operon expression. J Bacteriol 163:534-542. 123. Cohen A, Laban A.1983. Plasmidic recombination in Escherichia coli K-12: the role of recF gene function. Mol Gen Genet 189:471-474. 124. Kolodner R, Fishel RA, Howard M.1985. Genetic recombination of bacterial plasmid DNA: effect of RecF pathway mutations on plasmid recombination in Escherichia coli. J Bacteriol 163:1060-1066. 125. Zhang X, Wanda SY, Brenneman K, Kong W, Roland K, Curtiss R, III.2011. Improving Salmonella vector with rec mutation to stabilize the DNA cargoes. BMC Microbiol 11:31. 126. Beuzón CR, Méresse S, Unsworth KE, Ruiz-Albert J, Garvis S, Waterman SR, Ryder TA, Boucrot E, Holden DW.2000. Salmonella maintains the integrity of its intracellular vacuole through the action of SifA. EMBO J 19:3235-3249. 127. Ohlson MB, Huang Z, Alto NM, Blanc MP, Dixon JE, Chai J, Miller SI.2008. Structure and function of Salmonella SifA indicate that its interactions with SKIP, SseJ, and RhoA family GTPases induce endosomal tubulation. Cell Host Microbe 4:434-446. 128. Saltzman DA, Katsanis E, Heise CP, Hasz DE, Kelly SM, Curtiss R, III, Leonard AS, Anderson PM.1997. Patterns of hepatic and splenic colonization by an attenuated strain of Salmonella typhimurium containing the gene for human interleukin-2: a novel anti-tumor agent. Cancer Biother Radiopharm 12:37-45. 129. Saltzman DA, Katsanis E, Heise CP, Hasz DE, Vigdorovich V, Kelly SM, Curtiss R, III, Leonard AS, Anderson PM.1997. Antitumor mechanisms of attenuated Salmonella typhimurium containing the gene for human interleukin-2: a novel antitumor agent? J Pediatr Surg 32:301-306. 130. Schödel F, Kelly SM, Peterson DL, Milich DR, Curtiss R, III.1994. Hybrid hepatitis B virus core-pre-S proteins synthesized in avirulent Salmonella typhimurium and Salmonella typhi for oral vaccination. Infect Immun 62:1669- 1676. 131. Augustin LB, Milbauer L, Hastings SE, Leonard AS, Saltzman DA, Schottel JL.2021. Salmonella enterica Typhimurium engineered for nontoxic systemic colonization of autochthonous tumors. J Drug Target 29:294-299. 132. Curtiss R, III., Porter SB, Munson M, Tinge SA, Hassan JO, Gentry-Weeks C, Kelly SM.1991. Nonrecombinant and recombinant avirulent Salmonella live vaccines for poultry, p 169-198. In Blankenship LC, Bailey JHS, Cox NA, Stern NJ, Meinersmann RJ (ed), Colonization control of human bacterial enteropathogens in poultry. Academic Press New York 133. Zhang X, Kelly SM, Bollen WS, Curtiss R, III.1997. Characterization and immunogenicity of Salmonella typhimurium SL1344 and UK-1 ∆crp and ∆cdt deletion mutants. Infect Immun 65:5381-5387. 134. Sanapala S, Mosca L, Wang S, Curtiss R, III 2018. Comparative evaluation of Salmonella vaccines derived from UK-1 and 14028S submitted. 135. Bertani G.1951. Studies on lysogenesis. I. The mode of phage liberation by lysogenic Escherichia coli. J Bacteriol 62:293-300. 136. Sambrook J, Russell DW.2001. Molecular cloning : a laboratory manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. 137. Schmieger H.1972. Phage P22-mutants with increased or decreased transduction abilities. Mol Gen Genet 119:75-88. 138. Schmieger H, Backhaus H.1976. Altered cotransduction frequencies exhibited by HT-mutants of Salmonella-phage P22. Mol Gen Genet 143:307-309. 139. Kang HY, Dozois CM, Tinge SA, Lee TH, Curtiss R, III.2002. Transduction- mediated transfer of unmarked deletion and point mutations through use of counterselectable suicide vectors. J Bacteriol 184:307-312. 140. Edwards RA, Keller LH, Schifferli DM.1998. Improved allelic exchange vectors and their use to analyze 987P fimbria gene expression. Gene 207:149-157. 141. Quandt J, Hynes MF.1993. Versatile suicide vectors which allow direct selection for gene replacement in gram-negative bacteria. Gene 127:15-21. 142. Roland K, Curtiss R, III., Sizemore D.1999. Construction and evaluation of a ∆cya ∆crp Salmonella typhimurium strain expressing avian pathogenic Escherichia coli O78 LPS as a vaccine to prevent airsacculitis in chickens. Avian Dis 43:429-441. 143. Shi H, Santander J, Brenneman KE, Wanda SY, Wang S, Senechal P, Sun W, Roland KL, Curtiss R, III.2010. Live recombinant Salmonella Typhi vaccines constructed to investigate the role of rpoS in eliciting immunity to a heterologous antigen. PLoS One 5:e11142. 144. Wilmes-Riesenberg MR, Bearson B, Foster JW, Curtiss R, III.1996. Role of the acid tolerance response in virulence of Salmonella typhimurium. Infect Immun 64:1085-1092. 145. Murray GL, Attridge SR, Morona R.2005. Inducible serum resistance in Salmonella typhimurium is dependent on wzzfepE-regulated very long O antigen chains. Microbes Infect 7:1296-1304. 146. Hitchcock PJ, Brown TM.1983. Morphological heterogeneity among Salmonella lipopolysaccharide chemotypes in silver-stained polyacrylamide gels. J Bacteriol 154:269-277. 147. Lin TY, Zhang H, Luo J, Li Y, Gao T, Lara PN, Jr., de Vere White R, Lam KS, Pan CX.2012. Multifunctional targeting micelle nanocarriers with both imaging and therapeutic potential for bladder cancer. Int J Nanomedicine 7:2793-2804. 148. Lin TY, Li YP, Zhang H, Luo J, Goodwin N, Gao T, White Rde V, Lam KS, Pan CX.2013. Tumor-targeting multifunctional micelles for imaging and chemotherapy of advanced bladder cancer. Nanomedicine (Lond) 8:1239-1251. 149. Li Y, Lin TY, Luo Y, Liu Q, Xiao W, Guo W, Lac D, Zhang H, Feng C, Wachsmann-Hogiu S, Walton JH, Cherry SR, Rowland DJ, Kukis D, Pan C, Lam KS.2014. A smart and versatile theranostic nanomedicine platform based on nanoporphyrin. Nat Commun 5:4712. 150. Lin TY, Li Y, Liu Q, Chen JL, Zhang H, Lac D, Zhang H, Ferrara KW, Wachsmann-Hogiu S, Li T, Airhart S, deVere White R, Lam KS, Pan CX. 2016. Novel theranostic nanoporphyrins for photodynamic diagnosis and trimodal therapy for bladder cancer. Biomaterials 104:339-351. 151. Zhang H, Aina OH, Lam KS, de Vere White R, Evans C, Henderson P, Lara PN, Wang X, Bassuk JA, Pan CX.2012. Identification of a bladder cancer- specific ligand using a combinatorial chemistry approach. Urol Oncol 30:635-645. 152. Galán JE, Curtiss R, III.1989. Cloning and molecular characterization of genes whose products allow Salmonella typhimurium to penetrate tissue culture cells. Proc Natl Acad Sci U S A 86:6383-6387. 153. Schneider CA, Rasband WS, Eliceiri KW.2012. NIH Image to ImageJ: 25 years of image analysis. Nat Methods 9:671-675. 154. Curtiss R, III. , Wang S.2021. Induction of protective immunity against antigens. US patent U.S. Patent 10,988,729. 155. Joseph M, Enting D.2019. Immune Responses in Bladder Cancer-Role of Immune Cell Populations, Prognostic Factors and Therapeutic Implications. Front Oncol 9:1270. 156. Cole KE, Strick CA, Paradis TJ, Ogborne KT, Loetscher M, Gladue RP, Lin W, Boyd JG, Moser B, Wood DE, Sahagan BG, Neote K.1998. Interferon- inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med 187:2009-2021. 157. Naumann U, Cameroni E, Pruenster M, Mahabaleshwar H, Raz E, Zerwes HG, Rot A, Thelen M.2010. CXCR7 functions as a scavenger for CXCL12 and CXCL11. PLoS One 5:e9175. 158. Colvin RA, Campanella GS, Sun J, Luster AD.2004. Intracellular domains of CXCR3 that mediate CXCL9, CXCL10, and CXCL11 function. J Biol Chem 279:30219-30227. 159. Karin N.2020. CXCR3 Ligands in Cancer and Autoimmunity, Chemoattraction of Effector T Cells, and Beyond. Front Immunol 11:976. 160. Gao Q, Zhang Y.2021. CXCL11 Signaling in the Tumor Microenvironment. Adv Exp Med Biol 1302:41-50. 161. Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. 2015. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev 26:311-327. 162. Billottet C, Quemener C, Bikfalvi A.2013. CXCR3, a double-edged sword in tumor progression and angiogenesis. Biochim Biophys Acta 1836:287-295. 163. Biber K, Dijkstra I, Trebst C, De Groot CJ, Ransohoff RM, Boddeke HW. 2002. Functional expression of CXCR3 in cultured mouse and human astrocytes and microglia. Neuroscience 112:487-497. 164. Sanchez-Martin L, Sanchez-Mateos P, Cabanas C.2013. CXCR7 impact on CXCL12 biology and disease. Trends Mol Med 19:12-22. 165. Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, McSkane M, Baba H, Lenz HJ.2018. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - A target for novel cancer therapy. Cancer Treat Rev 63:40-47. 166. Cao Y, Jiao N, Sun T, Ma Y, Zhang X, Chen H, Hong J, Zhang Y.2021. CXCL11 Correlates With Antitumor Immunity and an Improved Prognosis in Colon Cancer. Front Cell Dev Biol 9:646252. 167. Moon EK, Wang LS, Bekdache K, Lynn RC, Lo A, Thorne SH, Albelda SM. 2018. Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines. Oncoimmunology 7:e1395997. 168. Gao Q, Wang S, Chen X, Cheng S, Zhang Z, Li F, Huang L, Yang Y, Zhou B, Yue D, Wang D, Cao L, Maimela NR, Zhang B, Yu J, Wang L, Zhang Y.2019. Cancer-cell-secreted CXCL11 promoted CD8(+) T cells infiltration through docetaxel-induced-release of HMGB1 in NSCLC. J Immunother Cancer 7:42. 169. Wang P, Yang X, Xu W, Li K, Chu Y, Xiong S.2010. Integrating individual functional moieties of CXCL10 and CXCL11 into a novel chimeric chemokine leads to synergistic antitumor effects: a strategy for chemokine-based multi- target-directed cancer therapy. Cancer Immunol Immunother 59:1715-1726. 170. Bulina ME, Chudakov DM, Britanova OV, Yanushevich YG, Staroverov DB, Chepurnykh TV, Merzlyak EM, Shkrob MA, Lukyanov S, Lukyanov KA.2006. A genetically encoded photosensitizer. Nat Biotechnol 24:95-99. 171. Yuan M, Liu C, Li J, Ma W, Yu X, Zhang P, Ji Y.2019. The effects of photodynamic therapy on leukemia cells mediated by KillerRed, a genetically encoded fluorescent protein photosensitizer. BMC Cancer 19:934. 172. Liu J, Wang F, Qin Y, Feng X.2021. Advances in the Genetically Engineered KillerRed for Photodynamic Therapy Applications. Int J Mol Sci 22:10130. 173. Liao ZX, Li YC, Lu HM, Sung HW.2014. A genetically-encoded KillerRed protein as an intrinsically generated photosensitizer for photodynamic therapy. Biomaterials 35:500-508. 174. Carpentier P, Violot S, Blanchoin L, Bourgeois D.2009. Structural basis for the phototoxicity of the fluorescent protein KillerRed. FEBS Lett 583:2839-2842. 175. Bulina ME, Lukyanov KA, Britanova OV, Onichtchouk D, Lukyanov S, Chudakov DM.2006. Chromophore-assisted light inactivation (CALI) using the phototoxic fluorescent protein KillerRed. Nat Protoc 1:947-953. 176. Brosius J, Erfle M, Storella J.1985. Spacing of the -10 and -35 regions in the tac promoter. Effect on its in vivo activity. J Biol Chem 260:3539-3541. 177. Pelletier J, Sonenberg N.1988. Internal initiation of translation of eukaryotic mRNA directed by a sequence derived from poliovirus RNA. Nature 334:320- 325. 178. Kim JH, Lee SR, Li LH, Park HJ, Park JH, Lee KY, Kim MK, Shin BA, Choi SY.2011. High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice. PLoS One 6:e18556. 179. Jang SK, Krausslich HG, Nicklin MJ, Duke GM, Palmenberg AC, Wimmer E. 1988. A segment of the 5' nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation. J Virol 62:2636-2643. 180. Kreiter S, Selmi A, Diken M, Sebastian M, Osterloh P, Schild H, Huber C, Tureci O, Sahin U.2008. Increased antigen presentation efficiency by coupling antigens to MHC class I trafficking signals. J Immunol 180:309-318. 181. Hinnebusch AG, Ivanov IP, Sonenberg N.2016. Translational control by 5'- untranslated regions of eukaryotic mRNAs. Science 352:1413-1416. 182. Sonenberg N.1994. mRNA translation: influence of the 5' and 3' untranslated regions. Curr Opin Genet Dev 4:310-315. 183. Yamashita A, Takeuchi O.2017. Translational control of mRNAs by 3'- Untranslated region binding proteins. BMB Rep 50:194-200. 184. Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, Tureci O, Sahin U.2006. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood 108:4009-4017. 185. Castle JC, Kreiter S, Diekmann J, Lower M, van de Roemer N, de Graaf J, Selmi A, Diken M, Boegel S, Paret C, Koslowski M, Kuhn AN, Britten CM, Huber C, Tureci O, Sahin U.2012. Exploiting the mutanome for tumor vaccination. Cancer Res 72:1081-1091. 186. Kreiter S, Vormehr M, van de Roemer N, Diken M, Lower M, Diekmann J, Boegel S, Schrors B, Vascotto F, Castle JC, Tadmor AD, Schoenberger SP, Huber C, Tureci O, Sahin U.2015. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520:692-696. 187. Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, Grunwitz C, Vormehr M, Husemann Y, Selmi A, Kuhn AN, Buck J, Derhovanessian E, Rae R, Attig S, Diekmann J, Jabulowsky RA, Heesch S, Hassel J, Langguth P, Grabbe S, Huber C, Tureci O, Sahin U.2016. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534:396-401. 188. Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, Chen C, Olive O, Carter TA, Li S, Lieb DJ, Eisenhaure T, Gjini E, Stevens J, Lane WJ, Javeri I, Nellaiappan K, Salazar AM, Daley H, Seaman M, Buchbinder EI, Yoon CH, Harden M, Lennon N, Gabriel S, Rodig SJ, Barouch DH, Aster JC, Getz G, Wucherpfennig K, Neuberg D, Ritz J, Lander ES, Fritsch EF, Hacohen N, Wu CJ.2017. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547:217-221. 189. Gubin MM, Artyomov MN, Mardis ER, Schreiber RD.2015. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest 125:3413-3421. 190. Schumacher TN, Schreiber RD.2015. Neoantigens in cancer immunotherapy. Science 348:69-74. 191. Ott PA, Hu-Lieskovan S, Chmielowski B, Govindan R, Naing A, Bhardwaj N, Margolin K, Awad MM, Hellmann MD, Lin JJ, Friedlander T, Bushway ME, Balogh KN, Sciuto TE, Kohler V, Turnbull SJ, Besada R, Curran RR, Trapp B, Scherer J, Poran A, Harjanto D, Barthelme D, Ting YS, Dong JZ, Ware Y, Huang Y, Huang Z, Wanamaker A, Cleary LD, Moles MA, Manson K, Greshock J, Khondker ZS, Fritsch E, Rooney MS, DeMario M, Gaynor RB, Srinivasan L.2020. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 183:347-362 e324. 192. Desrichard A, Snyder A, Chan TA.2016. Cancer Neoantigens and Applications for Immunotherapy. Clin Cancer Res 22:807-812. 193. Capietto AH, Jhunjhunwala S, Delamarre L.2017. Characterizing neoantigens for personalized cancer immunotherapy. Curr Opin Immunol 46:58-65. 194. Zhu Y, Liu J.2021. The Role of Neoantigens in Cancer Immunotherapy. Front Oncol 11:682325. 195. Pearlman AH, Hwang MS, Konig MF, Hsiue EH, Douglass J, DiNapoli SR, Mog BJ, Bettegowda C, Pardoll DM, Gabelli SB, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S.2021. Targeting public neoantigens for cancer immunotherapy. Nat Cancer 2:487-497. 196. Ma W, Pham B, Li T.2022. Cancer neoantigens as potential targets for immunotherapy. Clin Exp Metastasis 39:51-60. 197. Castle JC, Uduman M, Pabla S, Stein RB, Buell JS.2019. Mutation-Derived Neoantigens for Cancer Immunotherapy. Front Immunol 10:1856. 198. Hu Z, Leet DE, Allesoe RL, Oliveira G, Li S, Luoma AM, Liu J, Forman J, Huang T, Iorgulescu JB, Holden R, Sarkizova S, Gohil SH, Redd RA, Sun J, Elagina L, Giobbie-Hurder A, Zhang W, Peter L, Ciantra Z, Rodig S, Olive O, Shetty K, Pyrdol J, Uduman M, Lee PC, Bachireddy P, Buchbinder EI, Yoon CH, Neuberg D, Pentelute BL, Hacohen N, Livak KJ, Shukla SA, Olsen LR, Barouch DH, Wucherpfennig KW, Fritsch EF, Keskin DB, Wu CJ, Ott PA. 2021. Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat Med 27:515-525. 199. Vasconcelos-Nobrega C, Colaco A, Lopes C, Oliveira PA.2012. Review: BBN as an urothelial carcinogen. In Vivo 26:727-739. 200. Summerhayes IC, Franks LM.1979. Effects of donor age on neoplastic transformation of adult mouse bladder epithelium in vitro. J Natl Cancer Inst 62:1017-1023. 201. Lattime EC, Gomella LG, McCue PA.1992. Murine bladder carcinoma cells present antigen to BCG-specific CD4+ T-cells. Cancer Res 52:4286-4290. 202. Saito R, Smith CC, Utsumi T, Bixby LM, Kardos J, Wobker SE, Stewart KG, Chai S, Manocha U, Byrd KM, Damrauer JS, Williams SE, Vincent BG, Kim WY. 2018. Molecular Subtype-Specific Immunocompetent Models of High-Grade Urothelial Carcinoma Reveal Differential Neoantigen Expression and Response to Immunotherapy. Cancer Res 78:3954-3968. 203. Prosch S, Stein J, Staak K, Liebenthal C, Volk HD, Kruger DH.1996. Inactivation of the very strong HCMV immediate early promoter by DNA CpG methylation in vitro. Biol Chem Hoppe Seyler 377:195-201. 204. Brooks AR, Harkins RN, Wang P, Qian HS, Liu P, Rubanyi GM.2004. Transcriptional silencing is associated with extensive methylation of the CMV promoter following adenoviral gene delivery to muscle. J Gene Med 6:395-404. 205. Uetsuki T, Naito A, Nagata S, Kaziro Y.1989. Isolation and characterization of the human chromosomal gene for polypeptide chain elongation factor-1α. J Biol Chem 264:5791-5798. 206. Kim DW, Uetsuki T, Kaziro Y, Yamaguchi N, Sugano S.1990. Use of the human elongation factor 1α promoter as a versatile and efficient expression system. Gene 91:217-223. 207. Kaufman WL, Kocman I, Agrawal V, Rahn HP, Besser D, Gossen M.2008. Homogeneity and persistence of transgene expression by omitting antibiotic selection in cell line isolation. Nucleic Acids Res 36:e111. 208. Teschendorf C, Warrington KH, Jr., Siemann DW, Muzyczka N.2002. Comparison of the EF-1α and the CMV promoter for engineering stable tumor cell lines using recombinant adeno-associated virus. Anticancer Res 22:3325- 3330. 209. Gopalkrishnan RV, Christiansen KA, Goldstein NI, DePinho RA, Fisher PB. 1999. Use of the human EF-1α promoter for expression can significantly increase success in establishing stable cell lines with consistent expression: a study using the tetracycline-inducible system in human cancer cells. Nucleic Acids Res 27:4775-4782. 210. Byun HM, Suh D, Jeong Y, Wee HS, Kim JM, Kim WK, Ko JJ, Kim JS, Lee YB, Oh YK.2005. Plasmid vectors harboring cellular promoters can induce prolonged gene expression in hematopoietic and mesenchymal progenitor cells. Biochem Biophys Res Commun 332:518-523. 211. Wang X, Xu Z, Tian Z, Zhang X, Xu D, Li Q, Zhang J, Wang T.2017. The EF- 1α promoter maintains high-level transgene expression from episomal vectors in transfected CHO-K1 cells. J Cell Mol Med 21:3044-3054. 212. Gill DR, Smyth SE, Goddard CA, Pringle IA, Higgins CF, Colledge WH, Hyde SC.2001. Increased persistence of lung gene expression using plasmids containing the ubiquitin C or elongation factor 1α promoter. Gene Ther 8:1539- 1546. 213. Pardoll DM.2012. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12:252-264. 214. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM.2012. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366:2455-2465. 215. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M.2012. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366:2443-2454. 216. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A, Mateus C, Boasberg P, Tumeh PC, Chmielowski B, Ebbinghaus SW, Li XN, Kang SP, Ribas A.2013. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369:134-144. 217. Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, Vaishampayan UN, Drabkin HA, George S, Logan TF, Margolin KA, Plimack ER, Lambert AM, Waxman IM, Hammers HJ.2015. Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. J Clin Oncol 33:1430-1437. 218. Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, Horn L, Lena H, Minenza E, Mennecier B, Otterson GA, Campos LT, Gandara DR, Levy BP, Nair SG, Zalcman G, Wolf J, Souquet PJ, Baldini E, Cappuzzo F, Chouaid C, Dowlati A, Sanborn R, Lopez-Chavez A, Grohe C, Huber RM, Harbison CT, Baudelet C, Lestini BJ, Ramalingam SS.2015. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol 16:257-265. 219. Tobias J, Steinberger P, Drinic M, Wiedermann U.2021. Emerging targets for anticancer vaccination: PD-1. ESMO Open 6:100278. 220. Lee CM, Tannock IF.2010. The distribution of the therapeutic monoclonal antibodies cetuximab and trastuzumab within solid tumors. BMC Cancer 10:255. 221. Sharkey RM, Gold DV, Aninipot R, Vagg R, Ballance C, Newman ES, Ostella F, Hansen HJ, Goldenberg DM.1990. Comparison of tumor targeting in nude mice by murine monoclonal antibodies directed against different human colorectal cancer antigens. Cancer Res 50:828s-834s. 222. Scott AM, Wolchok JD, Old LJ.2012. Antibody therapy of cancer. Nat Rev Cancer 12:278-287. 223. Baldo BA.2022. Immune- and Non-Immune-Mediated Adverse Effects of Monoclonal Antibody Therapy: A Survey of 110 Approved Antibodies. Antibodies (Basel) 11:17. 224. Weiner GJ.2015. Building better monoclonal antibody-based therapeutics. Nat Rev Cancer 15:361-370. 225. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, Gilson MM, Wang C, Selby M, Taube JM, Anders R, Chen L, Korman AJ, Pardoll DM, Lowy I, Topalian SL. 2010. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 28:3167-3175. 226. Garcia-Marquez MA, Thelen M, Reinke S, Keller D, Wennhold K, Lehmann J, Veldman J, Borchmann S, Rosenwald A, Sasse S, Diepstra A, Borchmann P, Engert A, Klapper W, von Bergwelt-Baildon M, Brockelmann PJ, Schlosser HA.2022. Reverted exhaustion phenotype of circulating lymphocytes as immune correlate of anti-PD1 first-line treatment in Hodgkin lymphoma. Leukemia 36:760-771. 227. Mor A, Strazza M.2021. Bridging the Gap: Connecting the Mechanisms of Immune-Related Adverse Events and Autoimmunity Through PD-1. Front Cell Dev Biol 9:790386. 228. Pascolutti R, Sun X, Kao J, Maute RL, Ring AM, Bowman GR, Kruse AC. 2016. Structure and Dynamics of PD-L1 and an Ultra-High-Affinity PD-1 Receptor Mutant. Structure 24:1719-1728. 229. Maute RL, Gordon SR, Mayer AT, McCracken MN, Natarajan A, Ring NG, Kimura R, Tsai JM, Manglik A, Kruse AC, Gambhir SS, Weissman IL, Ring AM. 2015. Engineering high-affinity PD-1 variants for optimized immunotherapy and immuno-PET imaging. Proc Natl Acad Sci U S A 112:E6506-6514. 230. Tan S, Liu K, Chai Y, Zhang CW, Gao S, Gao GF, Qi J.2018. Distinct PD-L1 binding characteristics of therapeutic monoclonal antibody durvalumab. Protein Cell 9:135-139. 231. Li Y, Cong Y, Jia M, He Q, Zhong H, Zhao Y, Li H, Yan M, You J, Liu J, Chen L, Hang H, Wang S.2021. Targeting IL-21 to tumor-reactive T cells enhances memory T cell responses and anti-PD-1 antibody therapy. Nat Commun 12:951. 232. Sasada R, Onda H, Igarashi K.1987. The establishment of IL-2 producing cells by genetic engineering. Cell Struct Funct 12:205-217. 233. Sasada R, Marumoto R, Igarashi K.1988. Secretion of human EGF and IgE in mammalian cells by recombinant DNA techniques; use of a IL-2 leader sequence. Cell Struct Funct 13:129-141. 234. Taniguchi T, Matsui H, Fujita T, Takaoka C, Kashima N, Yoshimoto R, Hamuro J.1983. Structure and expression of a cloned cDNA for human interleukin-2. Nature 302:305-310. 235. Engel JB, Tinneberg HR, Rick FG, Berkes E, Schally AV.2016. Targeting of Peptide Cytotoxins to LHRH Receptors For Treatment of Cancer. Curr Drug Targets 17:488-494. 236. Minko T, Dharap SS, Pakunlu RI, Wang Y.2004. Molecular targeting of drug delivery systems to cancer. Curr Drug Targets 5:389-406. 237. Dharap SS, Qiu B, Williams GC, Sinko P, Stein S, Minko T.2003. Molecular targeting of drug delivery systems to ovarian cancer by BH3 and LHRH peptides. J Control Release 91:61-73. 238. Dharap SS, Minko T.2003. Targeted proapoptotic LHRH-BH3 peptide. Pharm Res 20:889-896. 239. Szabo Z, Dezso B, Fodor K, Szegedi K, Flasko T, Szabo E, Olah G, Sipos E, Dobos N, Gardi J, Schally AV, Halmos G.2021. Expression of Luteinizing Hormone-Releasing Hormone (LHRH) and Type-I LHRH Receptor in Transitional Cell Carcinoma Type of Human Bladder Cancer. Molecules 26:1253. 240. Lae M, Couturier J, Oudard S, Radvanyi F, Beuzeboc P, Vieillefond A.2010. Assessing HER2 gene amplification as a potential target for therapy in invasive urothelial bladder cancer with a standardized methodology: results in 1005 patients. Ann Oncol 21:815-819. 241. Dharap SS, Wang Y, Chandna P, Khandare JJ, Qiu B, Gunaseelan S, Sinko PJ, Stein S, Farmanfarmaian A, Minko T.2005. Tumor-specific targeting of an anticancer drug delivery system by LHRH peptide. Proc Natl Acad Sci U S A 102:12962-12967. 242. Iqbal N, Iqbal N.2014. Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications. Mol Biol Int 2014:852748. 243. Li X, Taratula O, Taratula O, Schumann C, Minko T.2017. LHRH-Targeted Drug Delivery Systems for Cancer Therapy. Mini Rev Med Chem 17:258-267. 244. Nagy A, Schally AV.2005. Targeting of cytotoxic luteinizing hormone-releasing hormone analogs to breast, ovarian, endometrial, and prostate cancers. Biol Reprod 73:851-859. 245. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL.1987. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177-182. 246. Yokota T, Milenic DE, Whitlow M, Schlom J.1992. Rapid tumor penetration of a single-chain Fv and comparison with other immunoglobulin forms. Cancer Res 52:3402-3408. 247. Adams GP, McCartney JE, Tai MS, Oppermann H, Huston JS, Stafford WF, 3rd, Bookman MA, Fand I, Houston LL, Weiner LM.1993. Highly specific in vivo tumor targeting by monovalent and divalent forms of 741F8 anti-c-erbB-2 single-chain Fv. Cancer Res 53:4026-4034. 248. Milenic DE, Yokota T, Filpula DR, Finkelman MA, Dodd SW, Wood JF, Whitlow M, Snoy P, Schlom J.1991. Construction, binding properties, metabolism, and tumor targeting of a single-chain Fv derived from the pancarcinoma monoclonal antibody CC49. Cancer Res 51:6363-6371. 249. Li X, Stuckert P, Bosch I, Marks JD, Marasco WA.2001. Single-chain antibody-mediated gene delivery into ErbB2-positive human breast cancer cells. Cancer Gene Ther 8:555-565. 250. Schier R, McCall A, Adams GP, Marshall KW, Merritt H, Yim M, Crawford RS, Weiner LM, Marks C, Marks JD.1996. Isolation of picomolar affinity anti-c- erbB-2 single-chain Fv by molecular evolution of the complementarity determining regions in the center of the antibody binding site. J Mol Biol 263:551- 567. 251. Song E, Zhu P, Lee SK, Chowdhury D, Kussman S, Dykxhoorn DM, Feng Y, Palliser D, Weiner DB, Shankar P, Marasco WA, Lieberman J.2005. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat Biotechnol 23:709-717. 252. Raetz CR, Whitfield C.2002. Lipopolysaccharide endotoxins. Annu Rev Biochem 71:635-700. 253. Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, Xing L, Hung MC, Bonfiglio T, Hicks DG, Tang P.2010. The Expression Patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by Immunohistochemical Analysis in Breast Cancer Cell Lines. Breast Cancer (Auckl) 4:35-41. 254. Blankenstein MA, Henkelman MS, Klijn JG.1985. Direct inhibitory effect of a luteinizing hormone-releasing hormone agonist on MCF-7 human breast cancer cells. Eur J Cancer Clin Oncol 21:1493-1499. 255. Meng J, Lu X, Zhou Y, Zhang M, Ge Q, Zhou J, Hao Z, Gao S, Yan F, Liang C.2021. Tumor immune microenvironment-based classifications of bladder cancer for enhancing the response rate of immunotherapy. Mol Ther Oncolytics 20:410-421. 256. Berencsi K, Meropol NJ, Hoffman JP, Sigurdson E, Giles L, Rani P, Somasundaram R, Zhang T, Kalabis J, Caputo L, Furth E, Swoboda R, Marincola F, Herlyn D.2007. Colon carcinoma cells induce CXCL11-dependent migration of CXCR3-expressing cytotoxic T lymphocytes in organotypic culture. Cancer Immunol Immunother 56:359-370. 257. Nazari A, Ahmadi Z, Hassanshahi G, Abbasifard M, Taghipour Z, Falahati- Pour SK, Khorramdelazad H.2020. Effective Treatments for Bladder Cancer Affecting CXCL9/CXCL10/CXCL11/CXCR3 Axis: A Review. Oman Med J 35:e103. 258. Yan L, Kanada M, Zhang J, Okazaki S, Terakawa S.2015. Photodynamic Treatment of Tumor with Bacteria Expressing KillerRed. PLoS One 10:e0131518. 259. Shirmanova M, Yuzhakova D, Snopova L, Perelman G, Serebrovskaya E, Lukyanov K, Turchin I, Subochev P, Lukyanov S, Kamensky V, Zagaynova E.2015. Towards PDT with Genetically Encoded Photosensitizer KillerRed: A Comparison of Continuous and Pulsed Laser Regimens in an Animal Tumor Model. PLoS One 10:e0144617. 260. Chen X, Zaro JL, Shen WC.2013. Fusion protein linkers: property, design and functionality. Adv Drug Deliv Rev 65:1357-1369. 261. Reddy Chichili VP, Kumar V, Sivaraman J.2013. Linkers in the structural biology of protein-protein interactions. Protein Sci 22:153-167. 262. Patel DK, Menon DV, Patel DH, Dave G.2022. Linkers: A synergistic way for the synthesis of chimeric proteins. Protein Expr Purif 191:106012. 263. Liu Z, Chen O, Wall JBJ, Zheng M, Zhou Y, Wang L, Vaseghi HR, Qian L, Liu J.2017. Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci Rep 7:2193. 264. Luke GA, de Felipe P, Lukashev A, Kallioinen SE, Bruno EA, Ryan MD. 2008. Occurrence, function and evolutionary origins of '2A-like' sequences in virus genomes. J Gen Virol 89:1036-1042. 265. Szymczak-Workman AL, Vignali KM, Vignali DA.2012. Design and construction of 2A peptide-linked multicistronic vectors. Cold Spring Harb Protoc 2012:199-204. 266. Hellen CU, Sarnow P.2001. Internal ribosome entry sites in eukaryotic mRNA molecules. Genes Dev 15:1593-1612. 267. Kozak M.2005. A second look at cellular mRNA sequences said to function as internal ribosome entry sites. Nucleic Acids Res 33:6593-6602. 268. Shaimardanova AA, Kitaeva KV, Abdrakhmanova, II, Chernov VM, Rutland CS, Rizvanov AA, Chulpanova DS, Solovyeva VV.2019. Production and Application of Multicistronic Constructs for Various Human Disease Therapies. Pharmaceutics 11. 269. Kaufman RJ, Davies MV, Wasley LC, Michnick D.1991. Improved vectors for stable expression of foreign genes in mammalian cells by use of the untranslated leader sequence from EMC virus. Nucleic Acids Res 19:4485-4490.

Claims

Claims What is claimed is: 1. A genetically modified Salmonella cell (GMSC) engineered to exhibit regulated delayed lysis in vivo, the GMSC comprising a first heterologous nucleic acid that encodes a first gene product that causes the GMSC to be selectively localized to and/or internalized by target cells in vivo and a second heterologous nucleic acid that encodes a second gene product that facilitates killing of the target cells following internalization.
2. The GMSC of claim 1, wherein the first heterologous nucleic acid and the second heterologous nucleic acid are present on a plasmid in a balanced-lethal vector-host.
3. The GMSC of claims 1 or 2, wherein the GMSC is of a cancer cell targeting Salmonella (CCTS) strain.
4. The GMSC of any of claims 1-3 comprising one or more mutations set forth in Table 1.
5. The GMSC of any of claims 1-4 comprising a genotype as set forth in Table 3.
6. The GMSC of any of claims 1-5, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding OmpA operably linked to a nucleic acid sequence encoding PLZ4.
7. The GMSC of any of claims 1-6, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding CXCL11 or KillerRed, or both.
8. The GMSC of claim 7, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding KillerRed.
9. The GMSC of claim 8, wherein the second heterologous nucleic acid comprises a sequence encoding SEQ ID NO:18 and SEQ ID NO:20; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
10. The GMSC of claim 9, wherein CXCL11 is fused to KillerRed.
11. The GMSC of claim 10, wherein a P2A peptide is situated between CXCL11 and KillerRed.
12. The GMSC of claim 11, wherein the second heterologous nucleic acid comprises a sequence encoding SEQ ID NO: 52 or SEQ ID NO: 53; or an amino sequence comprising at least 90% or 95% sequence identity therewith
13. The GMSC of any of claims 1-6, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding KillerRed, KillerRed fused to neuromodulin N-terminal sequence, or KillerRed fused to a mitochondrial targeting sequence.
14. The GMSC of claim 13, wherein the second heterologous nucleic acid comprises a sequence encoding SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO: 52 and SEQ ID NO: 53; or an amino sequence comprising at least 90% or 95% sequence identity therewith
15. The GMSC of any of claims 1-5 and 11, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding a LHRH peptide or HER2ScFv, or both.
16. The GMSC of claim 15, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding at least one selected from the group consisting of SEQ ID NO: 24, SEQ ID NO:25, and SEQ ID NO: 47; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
17. The GMSC of any of claims 1-6, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding a HAC-PD1 or HaPD1-IgG, or both.
18. The GMSC of claim 17, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding at least one selected from the group consisting of SEQ ID NO: 49, and SEQ ID NO: 51; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
19. The GMSC of any of claims 1-6, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding an HLA peptide.
20. The GMSC of claim 19, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding SEQ ID NO: 21; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
21. The GMSC of claim 6, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding SEQ ID NO: 2, or an amino sequence comprising at least 90% or 95% sequence identity therewith.
22. The GMSC of claim 7, wherein the second heterologous nucleic acid sequence encodes SEQ ID NO: 14 or SEQ ID NO: 16, or an amino acid sequence comprising at least 90% or 95% sequence identity therewith.
23. The GMSC of claim 7, wherein the second heterologous nucleic acid sequence encodes SEQ ID NO: 18 or SEQ ID NO: 20, or an amino acid sequence comprising at least 90% or 95% sequence identity therewith.
24. The GMSC of any of claims 1-23, wherein the GMSC is of a Cancer Cell Targeting Salmonella (CCTS) strain, engineered to possess the following functionalities in vivo: (i) directly kill cancer cells; (ii) deliver gene products that cause cancer cells to self-destruct, (iii) deliver gene products that enhance ability to treat cancer cells, and/or (iv) directly or indirectly stimulate immune responses against cancer cells.
25. A genetically modified Salmonella cell (GMSC) engineered to exhibit regulated delayed lysis in vivo, the GMSC comprising a first heterologous nucleic acid that encodes a first gene product that causes the GMSC to be selectively localized to and/or internalized by target cells in vivo and a second heterologous nucleic acid that encodes a second gene product that facilitates killing of the target cells following internalization; wherein expression of the first heterologous nucleic acid is controlled by a bacterial promoter, optionally further comprising a sequence for secretion of the first gene product; and wherein expression of the second heterologous nucleic acid is controlled by a eukaryotic promoter for delivery to the nucleus of the target cells, wherein the bacterial promoter optionally comprises Ptrc, Ptac, Plac, or Plpp, wherein the sequence for secretion optionally comprises bla SSopt and wherein the eukaryotic promoter optionally comprises PCMV or PEF1α.
26. A composition comprising a GMSC of any of claims 1-25 and a pharmaceutically acceptable carrier.
27. A method for treating cancer comprising administering a therapeutically effective amount of genetically modified Salmonella cells (GMSCs) of claims 1-25 or composition of claim 26 to a subject who has cancer.
28. The method of claim 27, wherein the cancer is of a type selected from the group consisting of bone cancer, bladder cancer, brain cancer, breast cancer, cancer of the urinary tract, carcinoma, cervical cancer, colon cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, liver cancer, lung cancer, lymphoma and leukemia, melanoma, ovarian cancer, pancreatic cancer, pituitary cancer, prostate cancer, rectal cancer, renal cancer, sarcoma, testicular cancer, thyroid cancer, and uterine cancer. In addition, the methods may be used to treat tumors that are malignant (e.g., primary or metastatic cancers) and benign (e.g., hyperplasia, cyst, pseudocyst, hematoma, and benign neoplasm).
29. The method of claim 27, wherein the cancer is bladder cancer.
30. A plasmid comprising a first heterologous nucleic acid that encodes a first gene product whose expression in a GMSC causes the GMSC to be localized to and/or internalized by a target cell in vivo, wherein the first heterologous nucleic acid is operably linked to a first promoter that controls expression of the first heterologous nucleic acid in a Salmonella cell; and a second heterologous nucleic acid that encodes a second gene product whose expression facilitates killing of target cells following internalization by the target cell, wherein the second heterologous nucleic acid is operably linked to (i) a second promoter that controls expression of the second heterologous nucleic acid in the target cell and (ii) to a nucleus targeting sequence that directs the plasmid to a nucleus of target cell.
31. The plasmid of claim 30, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding OmpA operably linked to a nucleic acid sequence encoding PLZ4.
32. The plasmid of claim 31, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding SEQ ID NO: 2, or an amino sequence comprising at least 90% or 95% sequence identity therewith.
33. The plasmid of any of claims 30-32, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding CXCL11 or KillerRed, or both.
34. The plasmid of claim 33, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding KillerRed.
35. The plasmid of claims 33, wherein CXCL11 is fused to KillerRed.
36. The plasmid of claim 35, further comprising a P2A peptide is situated between CXCL11 and KillerRed.
37. The plasmid of claims 30-36, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding KillerRed fused to neuromodulin N-terminal sequence, or KillerRed fused to a mitochondrial targeting sequence.
38. The plasmid of claim 37, wherein the second heterologous nucleic acid comprises a sequence encoding SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO: 52 and SEQ ID NO: 53; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
39. The plasmid of claim 30, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding a LHRH peptide or HER2ScFv, or both.
40. The plasmid of claim 39, wherein the first heterologous nucleic acid comprises a nucleic acid sequence encoding at least one selected from the group consisting of SEQ ID NO: 24, SEQ ID NO:25, and SEQ ID NO: 47; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
41. The plasmid of any of claims 30-40, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding a HAC-PD1 or HaPD1-IgG, or both.
42. The plasmid of claim 41, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding at least one selected from the group consisting of SEQ ID NO: 49, and SEQ ID NO: 51; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
43. The plasmid of claim 30, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding an HLA peptide.
44. The plasmid of claim 43, wherein the second heterologous nucleic acid comprises a nucleic acid sequence encoding SEQ ID NO: 21; or an amino sequence comprising at least 90% or 95% sequence identity therewith.
45. The plasmid of any of claims 30-44, wherein the first promoter comprises a bacterial promoter, optionally further comprising a sequence such as bla SSopt for secretion of the first gene product, and the second promoter a eukaryotic promoter for delivery to the nucleus of the target cells wherein the bacterial promoter optionally comprises Ptrc, Ptac, Plac, or Plpp, wherein the sequence for secretion optionally comprises bla SSopt and wherein the eukaryotic promoter optionally comprises PCMV or PEF1α.
PCT/US2023/024484 2022-06-03 2023-06-05 Improved salmonella vectored therapies for treatment of cancer Ceased WO2023235633A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP23816835.5A EP4531880A2 (en) 2022-06-03 2023-06-05 Improved salmonella vectored therapies for treatment of cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263348492P 2022-06-03 2022-06-03
US63/348,492 2022-06-03

Publications (2)

Publication Number Publication Date
WO2023235633A2 true WO2023235633A2 (en) 2023-12-07
WO2023235633A3 WO2023235633A3 (en) 2024-02-08

Family

ID=89025643

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/024484 Ceased WO2023235633A2 (en) 2022-06-03 2023-06-05 Improved salmonella vectored therapies for treatment of cancer

Country Status (2)

Country Link
EP (1) EP4531880A2 (en)
WO (1) WO2023235633A2 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9598697B2 (en) * 2010-05-28 2017-03-21 The Arizona Board Of Regents For And On Behalf Of Arizona State University Recombinant bacterium to decrease tumor growth
WO2014043593A2 (en) * 2012-09-13 2014-03-20 Massachusetts Institute Of Technology Programmable drug delivery profiles of tumor-targeted bacteria
US11519007B2 (en) * 2019-02-22 2022-12-06 Arizona Board Of Regents On Behalf Of Arizona State University Tumor-navigating, self-eradicating, trail-armed salmonella for precision cancer therapy

Also Published As

Publication number Publication date
WO2023235633A3 (en) 2024-02-08
EP4531880A2 (en) 2025-04-09

Similar Documents

Publication Publication Date Title
EP2155243B1 (en) Compositions and methods comprising klk3, psca, or folh1 antigen
JP5347135B2 (en) Genetically engineered LISTERIA and methods of use thereof
EP2853269B1 (en) Dual delivery system for heterologous antigens comprising a recombinant Listeria strain attenuated by mutation of dal/dat and deletion of ActA comprising a nucleic acid molecule encoding an listeriolysin O - prostate specific anigen fusion protein
JP5530065B2 (en) How to make a vaccine lacking antibiotic resistance
US7855064B2 (en) Antibiotic resistance free vaccines and methods for constructing and using same
US20120135033A1 (en) Multiple delivery system for heterologous antigens
JP2021176322A (en) Listeria-based compositions comprising peptide minigene expression system, and methods of use thereof
AU2021261945B2 (en) Compositions and methods for generating an immune response to a tumor associated antigen
KR20180100443A (en) Personalized delivery vector-based immunotherapy and its use
KR20170063505A (en) Listeria-based immunogenic compositions for eliciting anti-tumor responses
WO2016183361A1 (en) Immunogenic listeria-based compositions comprising truncated acta-antigen fusions and methods of use thereof
CN107073094A (en) The recombinant listeria bacterium bacterial strain and its application method of expressing heterologous antigen coalescence protein
AU2013232291A1 (en) Suppressor cell function inhibition following listeria vaccine treatment
KR20160130774A (en) Biomarker directed multi-target immunotherapy
TW201833323A (en) Recombinant listeria vaccine strains and methods of using the same in cancer immunotherapy
CN116916944A (en) Vaccines, therapeutics and RNA delivery platforms based on immunostimulatory bacteria
EP4531880A2 (en) Improved salmonella vectored therapies for treatment of cancer
CN111334521B (en) Method for improving expression of non-integrated attenuated Listeria exogenous antigen protein
JP2023516420A (en) Live Salmonella typhi vectors engineered to express cancer protein antigens and methods of use thereof
TW202506174A (en) An mrna tumor vaccine targeting mica/b
Roeske et al. Delivery of Chicken Egg Ovalbumin to Dendritic Cells by Listeriolysin O-Secreting Vegetative Bacillus subtilis
HK40027157A (en) Microorganisms programmed to produce immune modulators and anti-cancer therapeutics in tumor cells
HK1138314B (en) Microorganisms as carriers of nucleotide sequences coding for antigens and protein toxins, process of manufacturing and uses thereof
HK1207315B (en) Dual delivery system for heterologous antigens comprising a recombinant listeria strain attenuated by mutation of dal/dat and deletion of acta comprising a nucleic acid molecule encoding an listeriolysin o - prostate specific anigen fusion protein
TW201437370A (en) Suppressor cell function inhibition following Listeria vaccine treatment

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: 23816835

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2023816835

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2023816835

Country of ref document: EP

Effective date: 20250103

WWP Wipo information: published in national office

Ref document number: 2023816835

Country of ref document: EP