US20020026655A1 - Genetically modified tumor-targeted bacteria with reduced virulence - Google Patents

Genetically modified tumor-targeted bacteria with reduced virulence Download PDF

Info

Publication number
US20020026655A1
US20020026655A1 US09/149,832 US14983298A US2002026655A1 US 20020026655 A1 US20020026655 A1 US 20020026655A1 US 14983298 A US14983298 A US 14983298A US 2002026655 A1 US2002026655 A1 US 2002026655A1
Authority
US
United States
Prior art keywords
salmonella
percent
mutant
msbb
gene
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.)
Granted
Application number
US09/149,832
Other versions
US6447784B1 (en
Inventor
David Bermudes
Kenneth Brooks Low
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.)
Yale University
Vion Pharmaceuticals Inc
Original Assignee
Yale University
Vion Pharmaceuticals 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
Priority to US09/149,832 priority Critical patent/US6447784B1/en
Application filed by Yale University, Vion Pharmaceuticals Inc filed Critical Yale University
Priority to DE69841261T priority patent/DE69841261D1/en
Priority to CA2302866A priority patent/CA2302866C/en
Priority to KR1020007002535A priority patent/KR20010015577A/en
Priority to BR9812079-4A priority patent/BR9812079A/en
Priority to JP2000510842A priority patent/JP2002500001A/en
Priority to AT98946891T priority patent/ATE447005T1/en
Priority to PCT/US1998/018701 priority patent/WO1999013053A1/en
Priority to CNB98811030XA priority patent/CN1253551C/en
Priority to NZ503376A priority patent/NZ503376A/en
Priority to EP98946891A priority patent/EP1012232B1/en
Priority to IL13493698A priority patent/IL134936A0/en
Priority to AU93807/98A priority patent/AU749695B2/en
Assigned to VION PHARMACEUTICALS, INC. reassignment VION PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BERMUDES, DAVID, ITTENSOHN, MARTINA
Assigned to YALE UNIVERSITY reassignment YALE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LOW, KENNETH BROOKS
Priority to HK01104342A priority patent/HK1033956A1/en
Publication of US20020026655A1 publication Critical patent/US20020026655A1/en
Priority to US10/187,278 priority patent/US6863894B2/en
Publication of US6447784B1 publication Critical patent/US6447784B1/en
Application granted granted Critical
Priority to US11/064,533 priority patent/US7354592B2/en
Assigned to YALE UNIVERSITY reassignment YALE UNIVERSITY CHANGE OF ASSIGNEE ADDRESS Assignors: YALE UNIVERSITY
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/255Salmonella (G)
    • 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
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/025Enterobacteriales, e.g. Enterobacter
    • A61K39/0275Salmonella
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • 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
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/8215Microorganisms
    • Y10S435/822Microorganisms using bacteria or actinomycetales
    • Y10S435/879Salmonella

Definitions

  • the present invention is concerned with the isolation of a gene of Salmonella which, when genetically disrupted, reduces both virulence and septic shock caused by this organism and increases sensitivity to agents which promote eradication of the bacteria, e.g., chelating agents.
  • the nucleotide sequence of this gene and the means for its genetic disruption are provided, and examples of the use of tumor-targeted bacteria which possess a disruption in this gene to inhibit growth of cancers, including, but not limited to, melanoma, colon cancer, and other solid tumors are described.
  • the present invention also provides for the genetic disruption of this gene in combination with disruption of an auxotrophic gene.
  • a major problem in the chemotherapy of solid tumor cancers is delivery of therapeutic agents, such as drugs, in sufficient concentrations to eradicate tumor cells while at the same time minimizing damage to normal cells.
  • therapeutic agents such as drugs
  • studies in many laboratories are directed toward the design of biological delivery systems, such as antibodies, cytokines, and viruses for targeted delivery of drugs, pro-drug converting enzymes, and/or genes into tumor cells.
  • biological delivery systems such as antibodies, cytokines, and viruses for targeted delivery of drugs, pro-drug converting enzymes, and/or genes into tumor cells.
  • mice injected with bacillus Calmette-Guerin (BCG) have increased serum levels of TNF and that TNF-positive serum caused necrosis of the sarcoma Meth A and other transplanted tumors in mice.
  • BCG Bacillus Calmette-Guerin
  • Klimpel et al., 1990, J. Immunol. 145:711-717 showed that fibroblasts infected in vitro with Shigella or Salmonella had increased susceptibility to TNF.
  • Pawelek et al. provided methods for isolation of genes from Rhodobacter responsible for monophosphoryl lipid A (MLA) production. MLA acts as an antagonist to septic shock. Pawelek et al.
  • Hone and Powell W097/18837 (“Hone and Powell”), disclose methods to produce gram-negative bacteria having non-pyrogenic Lipid A or LPS.
  • Hone and Powell broadly asserts that conditional mutations in a large number of genes including msbB, kdsA, kdsb, kdtA, and htrB, etc. can be introduced into a broad variety of gram-negative bacteria including E. coli, Shigella sp., Salmonella sp., etc., the only mutation exemplified is an htrB mutation introduced into E. coli.
  • Hone and Powell propose the therapeutic use of non-pyrogenic Salmonella with a mutation in the msbB gene, there is no enabling description of how to accomplish such use.
  • Hone and Powell propose using non-pyrogenic bacteria only for vaccine purposes.
  • a vaccine vector has requirements quite different from tumor-targeted vectors.
  • Vaccine vectors are intended to elicit an immune response.
  • a preferred live bacterial vaccine must be immunogenic so that it elicits protective immunity; however, the vaccine must not be capable of excessive growth in vivo which might result in adverse reactions.
  • a suitable bacterial vaccine vector is temperature sensitive having minimal replicative ability at normal physiological ranges of body temperature.
  • tumor-targeted parasitic vectors such as but not limited to Salmonella
  • preferred tumor-targeted parasitic vectors are safely tolerated by the normal tissues of the body such that pathogenesis is limited, yet the vectors target to tumors and freely replicate within them.
  • vaccine vectors which replicate minimally at normal body temperatures would not be suitable for use as tumor-targeted vectors.
  • tumor-specific Salmonella strains include 1) serum resistance, allowing the parasite to pass through the vasculature and lymphatic system in the process of seeking tumors, 2) facultative anaerobiasis, i.e., ability to grow under anaerobic or aerobic conditions allowing amplification in large necrotic tumors which are hypoxic as well as small metastatic tumors which may be more aerobic, 3) susceptibility to the host's defensive capabilities, limiting replication in normal tissues but not within tumors where the host defensive capabilities may be impaired, 4) attenuation of virulence, whereby susceptibility to the host defenses may be increased, and the parasite is tolerated by the host, but does not limit intratumoral replication, 5) invasive capacity towards tumor cells, aiding in tumor targeting and anti-tumor activity, 6) motility, aiding in permeation throughout the tumor, 7) antibiotic sensitivity for control during treatment and for post treatment elimination (e.g., sensitivity to ampicillin, chloramphenicol,
  • the present invention provides a means to enhance the safety of tumor-targeted bacteria, for example, by genetic modification of the lipid A molecule.
  • the modified tumor-targeted bacteria of the present invention induce TNF ⁇ less than the wild type bacteria and have reduced ability to directly kill normal mammalian cells or cause systemic disease compared to the wild type strain.
  • the modified tumor-targeted bacteria of the present invention have increased therapeutic efficacy, i.e., more effective dosages of bacteria can be used and for extended time periods due to the lower toxicity in the form of less induced TNF ⁇ and systemic disease.
  • the present invention provides compositions and methods for the genetic disruption of the msbB gene in bacteria, such as Salmonella, which results in bacteria, such as Salmonella, possessing a lesser ability to elicit TNF ⁇ and reduced virulence compared to the wild type.
  • bacteria such as Salmonella
  • the invention provides for improved methods for selecting genetic disruptions of the msbB gene.
  • the genetically modified bacteria have increased sensitivity to a chelating agent compared to bacteria with the wild type msbB gene.
  • Salmonella having a disrupted msbB gene which are hyperinvasive to tumor tissues, are able to replicate within the tumors, and are useful for inhibiting the growth and/or reducing the tumor volume of sarcomas, carcinomas, lymphomas or other solid tumor cancers, such as germ line tumors and tumors of the central nervous system, including, but not limited to, breast cancer, prostate cancer, cervical cancer, uterine cancer, lung cancer, ovarian cancer, testicular cancer, thyroid cancer, astrocytoma, glioma, pancreatic cancer, stomach cancer, liver cancer, colon cancer, and melanoma.
  • the bacteria are attenuated by other means, including but not limited biosynthetic pathway mutations leading to auxotrophy.
  • the biosynthetic pathway mutation is a genetic disruption of the puri gene.
  • the bacteria express pro-drug converting enzymes including but not limited to HSV-TK, cytosine deaminase (CD), and p450 oxidoreductase.
  • the present invention also provides a means for enhanced sensitivity for use in terminating therapy and for post therapy elimination.
  • the tumor-targeted bacteria having a genetically modified lipid A also have enhanced susceptibility to certain agents, e.g., chelating agents. It is a further advantage to modify tumor-targeted bacteria in this way because it increases the ability to eliminate the bacteria with agents which have an antibiotic-like effect, such as chelating agents including, but not limited to, Ethylenediaminetetraacetic Acid (EDTA), Ethylene Glycol-bis( ⁇ -aminoethyl Ether) N,N,N′,N′,-Tetraacetic Acid (EGTA), and sodium citrate. Modification to enhance the ability to eliminate the bacteria via exogenous means, such as the administration of an agent to which the genetically modified bacteria are more sensitive than their wild type counterparts, is therefore useful.
  • EDTA Ethylenediaminetetraacetic Acid
  • EGTA Ethylene Glycol-bis(
  • the present invention further provides for a Salmonella strain comprising deletion mutations in both the msbB gene as well as an auxotrophic gene.
  • the auxotrophic deletion mutation affects the purl gene.
  • these mutations lead to increased safety of the strain.
  • the strain also carries other mutations described herein which increase efficacy of the strain but are not essential for its safety.
  • Salmonella encompasses all Salmonella species, including: Salmonella typhi, Salmonella choleraesuis , and Salmonella enteritidis . Serotypes of Salmonella are also encompassed herein, for example, typhimurium, a subgroup of Salmonella enteritidis , commonly referred to as Salmonella typhimurium.
  • Attenuation is a modification so that a microorganism or vector is less pathogenic.
  • the end result of attenuation is that the risk of toxicity as well as other side-effects is decreased, when the microorganism or vector is administered to the patient.
  • Virulence is a relative term describing the general ability to cause disease, including the ability to kill normal cells or the ability to elicit septic shock (see specific definition below).
  • Septic shock is a state of internal organ failure due to a complex cytokine cascade, initiated by TNF ⁇ .
  • the relative ability of a microorganism or vector to elicit TNF ⁇ is used as one measure to indicate its relative ability to induce septic shock.
  • Chelating agent sensitivity is defined as the effective concentration at which bacteria proliferation is affected, or the concentration at which the viability of bacteria, as determined by recoverable colony forming units (c.f.u.), is reduced.
  • FIG. 1 The complete DNA sequence of the Salmonella wild type (WT) 14028 msbB gene (SEQ ID NO:1) and the deduced amino acid sequence of the encoded protein (SEQ ID NO:2).
  • FIGS. 2 A- 2 C Knockout construct generated using the cloned Salmonella WT 14028 msbb gene.
  • the cloned gene was cut with SphI and MluI thereby removing approximately half of the msbB coding sequence, and the tetracycline resistance gene (TET) from pBR322 cut with AatII and AvaI was inserted after blunt-ending using the Klenow fragment of DNA polymerase I.
  • A Knockout construct.
  • B Salmonella chromosomal copy of msbB.
  • C Salmonella disrupted chromosomal copy of msbB after homologous recombination.
  • the start codon (ATG) and stop codon (TAA) and restriction sites AseI, BamHI, SphI, MluI, and EcoRV are shown.
  • the position of two primers, P1 and P2 which generate two different sized PCR products for either wild type or disrupted msbB are shown.
  • FIGS. 3 A- 3 C Southern blot analysis of chromosomally disrupted Salmonella WT 14028 msbB.
  • the AseI enzyme cuts upstream of msbB, and the BamH1 cuts in one location in the wild type, but in a second location in the tetracycline gene which results in a higher molecular weight product.
  • Lane 1 shows the position of the band in the knockout construct, compared to the WT 14028 in lane 2 (WT). Lanes 3 and 4 show the clones YS8211 and YS861 with a higher molecular weight product.
  • C Southern blot of a similar gel probed with an 32 P-labeled mluI fragment derived from the cloned msbB. See text Section 7.2 for details.
  • FIG. 4 TNF ⁇ induction by live Salmonella WT 14028 in mice. 1 X 10 8 live bacteria in 0.1 cc phosphate buffered saline of the wild type or msbB ⁇ disrupted strains were injected i.v. in the tail vein of Balb/c mice. The bar graph indicates the TNF ⁇ induction with error bars. Clone YS8211 induces TNF ⁇ 32% compared to Salmonella WT 14028.
  • FIG. 5 TNF ⁇ response by Sinclair swine to live Salmonella WT 14028 and msbB clone YS8212. TNF ⁇ levels were measured at 1.5 and 6.0 hours following i.v. introduction of 1 ⁇ 109 c.f.u. Salmonella WT 14028 and YS8212. At 1.5 hours TNF ⁇ response was significantly lower (p ⁇ 0.011) in the msbB deletion mutant compared to the wild type.
  • FIGS. 6 A- 6 B Respiratory level changes induced by LPS from WT 14028 and msbB ⁇ clone YS8212. Sinclair swine were injected with A) 5 ⁇ g/kg purified LPS or B) 500 ⁇ g/kg purified LPS and respiration rate was determined.
  • the 500 ⁇ g/kg of LPS from Salmonella WT 14028 raised the rate of respiration to more than 4 times normal, whereas the rate of respiration in msbB ⁇ LPS-treated animals was less than doubled.
  • FIG. 7 TNF ⁇ induction by live Salmonella WT 14028 in human monocytes.
  • Human monocytes isolated from peripheral blood were exposed to increasing amounts of Salmonella c.f.u.
  • concentrations of TNF ⁇ induced by WT 14028 were more than 3 times higher than those induced by a number of msbB ⁇ clones, i.e., YS8211, YS8212, YS8658, and YS1170.
  • the concentration of TNF ⁇ induced by Salmonella WT 14028 was at least 105 times higher than concentrations of TNF ⁇ induced by the independent msbB knockouts, i.e., YS7216 and YS8211, and the derivatives, i.e., YS1170, YS8644, YS1604, YS8212, YS8658, YS1601, YS1629.
  • FIGS. 9 A- 9 B Survival of mice and Sinclair swine, injected with 2 ⁇ 10 7 or 1 ⁇ 10 9 respectively of live bacteria.
  • FIG. 10 Biodistribution of msbB ⁇ Salmonella YS8211 in B16FlO melanoma tumors. At 5 days, the ratio of msbB ⁇ Salmonella within the tumors compared to those in the liver exceeded 1000:1.
  • FIG. 11 Tumor retardation by msbB ⁇ Salmonella.
  • B16F10 melanoma tumors were implanted in the flank of C57BL/6 mice and allowed to progress to day 8. Mice either received no bacteria (control) or msbB ⁇ strains YS8211, YS8212, YS7216, YS1629. Two of the strains, YS8211 and YS1629 retarded tumor progression significantly, whereas strains YS7216 and YS8212 did not.
  • FIGS. 12 A- 12 B Sensitivity of WT 14028 and msbB disrupted bacteria to chelating agents.
  • Wild type and msbb disrupted Salmonella clone YS8211 and YS862 were grown in LB broth lacking sodium chloride (LB-zero), in the presence or absence of 1 mM EDTA (FIG. 12A) or in the presence or absence of 10 mM sodium citrate (FIG. 12B).
  • the OD 600 was determined and plotted as a function of time.
  • the msbB+ strain showed little inhibition by EDTA or sodium citrate, compared to the msbB ⁇ strains which showed near complete cessation of growth after 3 hours for EDTA or sodium citrate.
  • FIGS. 13 A- 13 B Survival of msbB ⁇ bacteria within murine macrophages.
  • Murine bone marrow-derived macrophages (FIG. 13A) and a murine macrophage cell line, J774, (FIG. 13B) were used as hosts for bacterial internalization and quantified over time. The data are presented as a percentage of initial c.f.u.
  • FIG. 14 Conversion of msbB1( ⁇ ): :tet to tet S using the positive selection suicide vector pCVD442 carrying a second version of the msbB ⁇ (msbB2 ( ⁇ ) amp R sac B+ )
  • FIG. 15 Schematic diagram of the derivation of strain YS1456 from wild type Salmonella typhimurium . See text Section 8.1 for details.
  • FIG. 16 Schematic diagram of the derivation of strain YS1646 from wild type Salmonella typhimurium . See text Section 8.2 for details.
  • FIG. 17 Effect of YS1646 dose on B16-B10 murine melanoma tumor growth.
  • FIG. 18 Antibiotic suppression of YS1646-induced mortality following lethal infection.
  • the present invention is based on the isolation of a gene of Salmonella, i.e., msbB, which, when present in its normal form, contributes to TNF ⁇ induction, general virulence, survival within macrophages, and insensitivity to certain agents which promote eradication of the bacteria.
  • the present invention is directed to the genetic modification of the gene which results in disrupting the normal function of the product of the gene, and the incorporation of the genetic modification into tumor-targeted bacteria, including Salmonella, for therapeutic use.
  • the bacteria have a genetic modification of the msbB gene as well as genetic modification of a gene in a biosynthetic pathway, such as the purI gene, resulting in an auxotrphic strain.
  • the genetically modified bacteria are used in animals, including humans, for reduction of volume and/or growth inhibition of solid tumors.
  • bacteria useful for the present invention show preference for attachment to and penetration into certain solid tumor cancer cells or have an enhanced propensity to proliferate in tumor tissues as compared to normal tissues.
  • bacteria include but are not limited to Salmonella, having a natural ability to distinguish between cancerous or neoplastic cells tissues and normal cells/tissues.
  • tumor cell-specific bacteria useful for the invention may be selected for and/or improved in tumor targeting ability using the methods described by Pawelek et al., WO 96/40238 incorporated herein by reference.
  • Pawelek et al. describe methods for isolating tumor cell-specific bacteria by cycling a microorganism through preselected target cells, preferably solid tumor cells in vitro, or through a solid tumor in vivo, using one or more cycles of infection.
  • E. coli gene has been shown to be involved in myristilization of lipid A (Somerville et al., 1996, J. Clin. Invest. 97:359-365.)
  • the chromosomal organization of the E. coli msbB gene and the DNA sequence coding for the msbB gene have been described (Engel, et al., 1992, J. Bacteriol. 174:6394-6403; Karow and Georgopoulos, 1992, J. Bacteriol. 174: 702-710; Somerville et al., 1996, J. Clin. Invest. 97: 359-365).
  • the msbB gene can be isolated from bacterial strains, other than E. coli , using low stringency DNA/DNA hybridization techniques known to those skilled in the art. (Sambrook et al., Molecular Cloning, Cold Spring Harbor Laboratory Press, 1989). For an illustrative example of isolation of a msbB gene of bacteria, including but not limited to Salmonella spp., see Section 7.1 infra. A bacterial DNA library can be probed with a 32 P-labeled msbB gene from E. coli . Hybridizing clones are determined to be correct if they contain DNA sequences similar to the known E. coli msbB gene.
  • One embodiment of the present invention provides a composition of matter which is a strain of bacteria with a genetic alteration in the msbB gene.
  • the bacteria is Salmonella sp.
  • Genetic alteration in the form of disruption or deletion can be accomplished by several means known to those skilled in the art, including homologous recombination using an antibiotic resistance marker. These methods involve disruption of the plasmid-based, cloned msbB gene using restriction endonucleases such that part or all of the gene is disrupted or eliminated or such that the normal transcription and translation are interrupted, and an antibiotic resistance marker for phenotypic selection is inserted in the region of that deletion, disruption or other alteration.
  • Linearized DNA is transformed into Salmonella, and bacteria bearing the antibiotic resistance are further examined for evidence of genetic alteration.
  • Means for examining genetic alteration include PCR analysis and Southern blotting.
  • Genetic disruption of a Salmonella msbB gene see Section 7.2.
  • the msbB ⁇ /antibiotic resistance marker can be transduced into a new bacterial strain.
  • An illustrative example is provided in Section 7.2. Bacteriophage P22 and a Salmonella msbB ⁇ clone can be grown in zero salt Luria broth and the new phages in the supernate can be used to infect a new Salmonella strain.
  • Yet another embodiment of the present invention provides Salmonella that are attenuated in more than one manner, e.g., a mutation in the pathway for lipid A production, such as the msbB mutation described herein and one or more mutations to auxotrophy for one or more nutrients or metabolites, such as uracil biosynthesis, purine biosynthesis, and arginine biosynthesis as described by Bochner, 1980, J. Bacteriol. 143:926-933 herein incorporated by reference.
  • the ability of msbB ⁇ Salmonella to accumulate within tumors is retained by msbB ⁇ 0 Salmonella having one or more mutations resulting in an auxotrophic strain.
  • the bacterial vector which selectively targets tumors and expresses a pro-drug converting enzyme is auxotrophic for uracil, aromatic amino acids, isoleucine and valine and synthesizes an altered lipid A.
  • the msbB ⁇ Salmonella also contain a genetic modification of the biosynthetic pathway gene, purl, leading to decreased virulence of the strain compared to wild type. An illustrative example is provided in Sections 7 and 8.
  • a characteristic of the msbB ⁇ Salmonella, described herein, is decreased ability to induce a TNF ⁇ response compared to the wild type bacterial vector. Both the whole bacteria and isolated or purified lipopolysaccharide (LPS) elicit a TNF ⁇ response.
  • LPS lipopolysaccharide
  • the msbB ⁇ Salmonella induce TNF ⁇ expression at about 5 percent to about 40 percent compared to the wild type Salmonella sp.
  • the msbB ⁇ Salmonella induce TNF ⁇ expression at about 5 percent to about 40 percent of the level induced by wild type Salmonella, e.g., WT 14028.
  • the msbB ⁇ Salmonella induce TNF ⁇ expression at about 10 percent to about 35 percent of that induced by a wild type Salmonella sp.
  • purified LPS from msbB ⁇ Salmonella induces TNF ⁇ expression at a level which is less than or equal to 0.001 percent of the level induced by LPS purified from wild type Salmonella sp.
  • TNF ⁇ response induced by whole bacteria or isolated or purified LPS can be assessed in vitro or in vivo using commercially available assay systems such as by enzyme linked immunoassay (ELISA).
  • ELISA enzyme linked immunoassay
  • Comparison of TNF ⁇ production on a per c.f.u. or on a pg/kg basis, is used to determine relative activity. Lower TNF ⁇ levels on a per unit basis indicate decreased induction of TNF ⁇ production.
  • Another characteristic of the msbB ⁇ Salmonella, described herein, is decreased virulence towards the host cancer patient compared to the wild type bacterial vector. Wild type Salmonella can under some circumstances exhibit the ability to cause significant progressive disease. Acute lethality can be determined for normal wild type live Salmonella and live msbB ⁇ Salmonella using animal models. For an illustrative example, see Section 7.4 and Section 9, Table III. Comparison of animal survival for a fixed inoculum is used to determine relative virulence. Strains having a higher rate of survival have decreased virulence.
  • msbB ⁇ Salmonella Another characteristic of msbB ⁇ Salmonella described herein, is decreased survival within macrophage cells as compared to survival of wild type bacteria.
  • Wild type Salmonella e.g., ATCC 14028
  • ATCC 14028 are noted for their ability to survive within macrophages (Baumler, et al., 1994, Infect. Immun. 62:1623-1630; Buchmeier and Heffron 1989, Infect. Immun. 57:1-7; Buchmeier and Heffron, 1990, Science 248:730-732; Buchmeier et al., 1993, Mol. Microbiol. 7:933-936; Fields et al., 1986, Proc. Natl. Acad. Sci.
  • a comparison of survival time in macrophages can be made using an in vitro cell culture assay. A lower number of c.f.u. over time is indicative of reduced survival within macrophages. For an illustrative example, see Section 8 infra. As shown therein, using the gentamicin-based internalization assay and bone marrow-derived murine macrophages or the murine macrophage cell line J774, a comparison of survival of WT 14028 and msbB ⁇ clone YS8211 was determined. In an embodiment of the invention, survival occurs at about 50 percent to about 30 percent; preferably at about 30 percent to about 10 percent; more preferably at about 10 percent to about 1 percent of survival of the wild type stain.
  • Another characteristic of one embodiment of the msbB ⁇ Salmonella, described herein, is increased sensitivity of the tumor-targeted bacteria to specific chemical agents which is advantageously useful to assist in the elimination of the bacteria after administration in vivo.
  • Bacteria are susceptible to a wide range of antibiotic classes.
  • certain Salmonella msbB ⁇ mutants encompassed by the present invention are sensitive to certain chemicals which are not normally considered antibacterial agents.
  • certain msbB ⁇ 0 Salmonella mutants are more sensitive than WT 14028 to chelating agents.
  • a chelating agent for example, EDTA, EGTA or sodium citrate. Comparison of growth is measured as a function of optical density, i.e., a lower optical density in the msbB ⁇ strain grown in the presence of an agent, than when the strain is grown in its absence, indicates sensitivity. Furthermore, a lower optical density in the msbB ⁇ strain grown in the presence of an agent, compared to the msbB + strain grown in its presence, indicates sensitivity specifically due to the msbB mutation. For an illustrative example, see section 7.7 infra.
  • 90 percent inhibition of growth of msbB ⁇ Salmonella occurs at about 0.25 mM EDTA to about 0.5 mM EDTA, preferably at about 99 percent inhibition at about 0.25 mM EDTA to above 0.5 mM EDTA, more preferably at greater than 99 percent inhibition at about 0.25 mM EDTA to about 0.5 mM EDTA. Similar range of growth inhibition is observed at similar concentrations of EGTA.
  • the msbB ⁇ mutants of the present invention are stable, i.e., produce few derivatives (as defined below).
  • modified LB 10 g tryptone, 5 g yeast extract, 2 ml 1N CaCl 2 , and 2 ml 1N MgSO 4 per liter, adjusted to pH 7 using 1N NaOH.
  • the msbB ⁇ mutants when grown in normal LB, the msbB ⁇ mutants may give rise to derivatives.
  • derivatives is intended to mean spontaneous variants of the msbB ⁇ mutants characterized by a different level of virulence, tumor inhibitory activity and/or sensitivity to a chelating agent when compared to the original msbB ⁇ mutant.
  • the level of virulence, tumor inhibitory activity, and sensitivity to a chelating agent of a derivative may be greater, equivalent, or less compared to the original msbB ⁇ mutant.
  • Derivatives of msbB ⁇ strains grow faster on unmodified LB than the original msbB ⁇ strains.
  • derivatives can be recognized by their ability to grow on MacConkey agar (an agar which contains bile salts) and by their resistance to chelating agents, such as EGTA and EDTA.
  • Derivatives can be stably preserved by cryopreservation at ⁇ 70° C. or lyophilization according to methods well known in the art (Cryz et al., 1990, In New Generation Vaccines, M. M. Levine (ed.), Marcel Dekker, New York pp.
  • Virulence is determined by evaluation of the administered dose at which half of the animals die (LD 50 ). Comparison of the LD 50 of the derivatives can be used to assess the comparative virulence. Decrease in the LD 50 of a spontaneous derivative as compared to its msbB ⁇ parent, indicates an increase in virulence. In an illustrative example, the faster-growing derivatives either exhibit the same level of virulence, a greater level of virulence, or a lower level of virulence compared to their respective original mutant strains (see Section 9, Table III.) In another example, the ability of a derivative to induce TNF ⁇ remains the same as the original mutant strain (see Section 7.3, FIG. 7).
  • the derivatives can either inhibit tumor growth more than or less than their respective original mutant strains (see Section 7.6, FIG. 11). It is demonstrated in Section 7.6 that the original msbB ⁇ mutant, YS8211, significantly inhibits tumor growth whereas a derivative of this clone, YS8212, has less tumor growth inhibition activity. In contrast, the derivative, YS1629, exhibits enhanced tumor growth inhibition activity compared to its parent msbB ⁇ clone, YS7216.
  • a derivative which is more virulent than its parent mutant but which does induce TNF ⁇ at a lower level when compared to the wild type, i.e., at a level of about 5 percent to about 40 percent of that induced by the wild type Salmonella, can be further modified to contain one or more mutations to auxotrophy.
  • the YS1170 derivative is mutated such that it is auxotrophic for one or more aromatic amino acids, e.g., aroA, and thus can be made less virulent and is useful according to the methods of the present invention.
  • genetic modifications of the purl gene involved in purine biosynthesis) yeild Salmonella strains that are less virulent than the parent strain. (See Sections 7 and 8).
  • the derivative Prior to use of a derivative in the methods of the invention, the derivative is assessed to determine its level of virulence, ability to induce TNF ⁇ , ability to inhibit tumor growth, and sensitivity to a chelating agent.
  • the msbB ⁇ mutant Salmonella are advantageously used in methods to produce a tumor growth inhibitory response or a reduction of tumor volume in an animal including a human patient having a solid tumor cancer.
  • the msbB ⁇ mutant Salmonella possess tumor targeting ability or target preferably to tumor cells/tissues rather than normal cells/tissues.
  • the msbB ⁇ mutant Salmonella possess the ability to retard or reduce tumor growth and/or deliver a gene or gene product that retards or reduces tumor growth.
  • Tumor targeting ability can be assessed by a variety of methods known to those skilled in the art, including but not limited to cancer animal models.
  • Salmonella with a msbB ⁇ modification are assayed to determine if they possess tumor targeting ability using the B16F10 melanoma subcutaneous animal model.
  • a positive ratio of tumor to liver indicates that the genetically modified Salmonella possesses tumor targeting ability.
  • see Section 7.5 see Section 7.5.
  • Salmonella with the msbB ⁇ modification can be assayed to determine if they possess anti-tumor ability using any of a number of standard in vivo models, for example, the B16F10 melanoma subcutaneous animal model.
  • standard in vivo models for example, the B16F10 melanoma subcutaneous animal model.
  • tumors are implanted in the flanks of mice and staged to day 8 and then bacterial strains are injected i.p. Tumor volume is monitored over time.
  • Anti-tumor activity is determined to be present if tumors are smaller in the bacteria-containing groups than in the untreated tumor-containing animals.
  • see section 7.6 infra see section 7.6 infra.
  • the Salmonella of the present invention for in vivo treatment are genetically modified such that, when administered to a host, the bacteria is less toxic to the host and easier to eradicate from the host's system.
  • the Salmonella are super-infective, attenuated and specific for a target tumor cell.
  • the Salmonella may be sensitive to chelating agents having antibiotic-like activity.
  • the Salmonella used in the methods of the invention can encode “suicide genes”, such as pro-drug converting enzymes or other genes, which are expressed and secreted by the Salmonella in or near the target tumor.
  • “suicide genes” such as pro-drug converting enzymes or other genes, which are expressed and secreted by the Salmonella in or near the target tumor.
  • Table 2 of Pawelek et al. W096/40238 at pages 34-35 presents an illustrative list of pro-drug converting enzymes which are usefully secreted or expressed by msbB ⁇ mutant Salmonella for use in the methods of the invention. Table 2 and pages 32-35 are incorporated herein by reference.
  • the gene can be under the control of either constitutive, inducible or cell-type specific promoters. See Pawelek et al.
  • a suicide gene is expressed and secreted only when a Salmonella has invaded the cytoplasm of the target tumor cell, thereby limiting the effects due to expression of the suicide gene to the target site of the tumor.
  • the Salmonella administered to the host, expresses the HSV TK gene.
  • the ganciclovir Upon concurrent expression of the TK gene and administration of ganciclovir to the host, the ganciclovir is phosphorylated in the periplasm of the microorganism which is freely permeable to nucleotide triphosphates.
  • the phosphorylated ganciclovir a toxic false DNA precursor, readily passes out of the periplasm of the microorganism and into the cytoplasm and nucleus of the host cell where it incorporates into host cell DNA, thereby causing the death of the host cell.
  • the method of the invention for inhibiting growth or reducing volume of a solid tumor comprises administering to a patient having a solid tumor, an effective amount of an isolated mutant Salmonella sp. comprising a genetically modified msbB gene, said mutant being capable of targeting to the solid tumor when administered in vivo.
  • the msbB ⁇ mutant Salmonella may also express a suicide gene as described above.
  • the isolated Salmonella is analyzed for sensitivity to chelating agents to insure for ease in eradication of the Salmonella from the patient's body after successful treatment or if the patient experiences complications due to the administration of the isolated Salmonella.
  • Salmonella is employed which is sensitive to a chelating agent, at about 0.25 mM to about 1.0 mM of a chelating agent such as EGTA, EDTA or sodium citrate can be administered to assist in eradication of the Salmonella after the anti-tumor effects have been achieved.
  • the mutant Salmonella When administered to a patient, e.g., an animal for veterinary use or to a human for clinical use, the mutant Salmonella can be used alone or may be combined with any physiological carrier such as water, an aqueous solution, normal saline, or other physiologically acceptable excipient.
  • a physiological carrier such as water, an aqueous solution, normal saline, or other physiologically acceptable excipient.
  • the dosage ranges from about 1.0 c.f.u./kg to about 1 ⁇ 10 10 c.f.u./kg; optionally from about 1.0 c.f.u./kg to about 1 ⁇ 10 8 c.f.u./kg; optionally from about 1 ⁇ 10 2 c.f.u./kg to about 1 x 108 c.f.u./kg; optionally from about 1 ⁇ 10 4 c.f.u./kg to about 1 ⁇ 10 8 c.f.u./kg.
  • the mutant Salmonella of the present invention can be administered by a number of routes, including but not limited to: orally, topically, injection including, but limited to intravenously, intraperitoneally, subcutaneously, intramuscularly, intratumorally, i.e., direct injection into the tumor, etc.
  • the entire library was harvested from the petri dishes by flooding them with phosphate buffered saline and using a glass rod to dislodge the colonies, and the resulting bacterial population was subjected to a large-scale plasmid isolation, resulting in an amplified Salmonella library plasmid pool. This plasmid pool was then transformed to Salmonella LT2 YS5010, thereby eliminating the E. coli background.
  • a probe for msbB homologues was generated using a clone of the E. coli msbB gene (Karow and Georgopoulos 1992 J. Bacteriol. 174: 702-710) by digesting E. coli with BglII/HincII and isolating a 600 bp fragment which corresponds to a portion of the coding sequence. This fragment was labeled using ⁇ 32 P-dCTP and used to probe the Salmonella library at low-stringency conditions consisting of 6 ⁇ SSC, 0.1 % SDS, 2 ⁇ Denhardts, 0.5 % non-fat dry milk overnight at 55° C.
  • the complete nucleotide sequence of the Salmonella msbb gene (SEQ ID NO:1) and the deduced amino acid sequence of the encoded protein (SEQ ID NO:2) is shown in FIG. 1.
  • the DNA homology of the putative Salmonella msbB and the E. coli msbB is 75%.
  • the protein homology is 98%, confirming that the cloned Salmonella gene is a bona fide msbB.
  • a knockout construct was generated using the cloned Salmonella msbB gene.
  • the cloned gene was cut with SphI and MluI, thereby removing approximately half of the msbB coding sequence, and the tetracycline resistance gene from pBR322, cut with AatII and AvaI, was inserted after blunt-ending using the Klenow fragment of DNA polymerase I (FIG. 2A- 2 C).
  • the knockout disruption was accomplished by homologous recombination procedures (Russell et al., 1989, J. Bacteriol.
  • Bacteria from the transformation protocol were first selected on tetracycline plates, and subsequently examined for the presence of plasmid-containing non-chromosomal integrated contaminants by ampicillin resistance and the presence of plasmids as determined by standard plasmid mini-preps (Titus, D. E., ed. Promega Protocols and Applications Guide , Promega Corp, 1991). Bacterial colonies which were tetracycline resistant yet lacked plasmids were subjected to a PCR-based analysis of the structure of their msbB gene.
  • PCR was used with primers which generate a fragment inclusive of the region into which the tetracycline gene was inserted, where the forward primer was GTTGACTGGGAAGGTCTGGAG (SEQ ID NO:3), corresponding to bases 586 to 606, and the reverse primer was CTGACCGCGCTCTATCGCGG (SEQ ID NO:4), corresponding to bases 1465 to 1485.
  • Wild type Salmonella msbB+ results in an approximately 900 base pair product
  • the disrupted gene with the tetracycline insert results in an approximately 1850 base pair product.
  • Several clones were obtained where only the larger PCR product was produced, indicating that the disruption in the msbB gene had occurred.
  • msbB mutation was confirmed, additional strains containing the msbB ⁇ mutation were generated.
  • the Salmonella strains used included WT 14028 and YS72 (pur ⁇ xyl ⁇ hyperinvasive mutant from WT 14028; Pawelek et al., WO 96/40238).
  • P22 transduction was used to generate YS8211 (msbB::tet) using YS82 as a donor and YS861 and YS862 (msbB1::tet) using YS86 as a donor; all with WT 14028 as recipient.
  • YS7216 (msbB1::tet from YS72) was generated by transduction using YS82 as a donor.
  • Several derivatives are encompassed by the present invention, including but not limited to derivatives of YS8211 (YS8212, YS1170), YS862 (YS8644, YS8658), and YS7216 (YS1601, YS1604, YS1629).
  • spontaneous derivatives grow somewhat faster on Luria agar compared to WT 14028 or msbB ⁇ clones generated by transduction.
  • msbB + strains were grown in LB broth or on LB plates containing 1.5% agar at 37° C.
  • msbB ⁇ strains were grown in modified LB containing 10 g tryptone, 5 g yeast extract, 2 ml 1N CaCl 2 and 2 ml 1N MgSO 4 per liter, adjusted to pH 7 using 1N NaOH.
  • LB lacking NaCl was used, with 4 mg/l tetracycline. Liquid cultures were shaken at 225 rpm.
  • cells were diluted 1:100 in LB, grown to OD 600 32 0.8 to 1.0, washed in phosphate buffered saline (PBS), and resuspended in PBS.
  • PBS phosphate buffered saline
  • E. coli SM10 ⁇ pir carrying a plasmid with the msbB( ⁇ ) bla and sacB genes was used as a donor.
  • the bla gene for betalactamase confers resistance to ampicillin.
  • the donor strain was mated using standard mating procedures, with a Salmonella strain into which the plasmid with msbB( ⁇ ) bla sacb was to be introduced. Since the Salmonella strain contained a second antibiotic resistance marker (e.g., streptomycin resistance), the recombinant Salmonella clones were then selected for dual resistance to ampicillin and streptomycin.
  • a second antibiotic resistance marker e.g., streptomycin resistance
  • Colonies were then tested for sensitivity to tetracycline and ampicillin as above, and the msbB isoform was confirmed by PCR. This improved method was used to generate strains for P22 phage transduction of msbB( ⁇ ) bla sacB chromosomal element. These strains were then used to generate the YS1456 and YS1646 stains, which represent preferred embodiments of the novel msbB mutations of the present invention (see FIG. 15 and 16).
  • Results are presented in FIG. 4 and expressed as a percent of the level of TNF ⁇ induced by wild type Salmonella.
  • YS8211 induced TNF ⁇ significantly less than WT 14028.
  • the msbB ⁇ strain induced TNF ⁇ about 33% (i.e., 3 times less) of the wild type msbB + strain.
  • Results are presented in FIG. 5 and are expressed as picograms of TNF ⁇ /ml serum.
  • LPS Lipopolysaccharide
  • Salmonella WT 14028 and the msbB ⁇ clone, YS8212 was prepared using the procedure described by Galanos et al. (1969 Eur. J. Biochem. 9: 245-249). Briefly, LPS was extracted from bacteria which had been grown to OD 600 of 1.0. The bacteria were pelleted by centrifugation, washed twice with distilled water and frozen at ⁇ 20 C. LPS was purified by extraction with a mixture of 18.3 ml H20:15 ml phenol in a shaking water bath for 1 hr at 70 C.
  • the mixture was cooled on ice, centrifuged at 20,000 ⁇ g for 15 min, and the aqueous phase was removed.
  • LPS was precipitated from the aqueous phase by addition of NaCl to 0.05 M and 2 volumes ethanol and incubation on ice, followed by centrifugation of 2000 ⁇ g for 10 min. The precipitation was repeated after redissolving the pellet in 0.05 M NaCl, and the pellet lyophilized.
  • the LPS was dissolved in sterile distilled water, and either 5 ⁇ g/kg or 500 ⁇ g/kg LPS was injected into the ear vein of Sinclair swine which had been anesthetized with Isoflurane. After 1.5 and 6.0 hours, respiration rate was determined and recorded.
  • Results are presented in FIG. 6 and are expressed as a percentage of respiration at time zero (t o ).
  • respiration was significantly higher in the pigs administered wild type LPS as compared to those administered the LPS from the msbB ⁇ strain.
  • disruption of the msbb gene in Salmonella produces a modification in lipid A which results in reduced ability to increase respiration.
  • Human monocytes were prepared from peripheral blood by centrifugation through Isolymph (Pharmacia) and allowed to adhere to 24 well plates containing RPMI 1640.
  • the bacteria were added to the cell culture wells and the culture medium was harvested after 2.0 hours, centrifuged to remove the cellular content, and analyzed for TNF ⁇ using a Genzyme redicta ELISA plate, which was read using a Gilson spectrophotometer.
  • the bacteria were diluted into phosphate buffered saline (PBS) at a ratio of 1:10 and the equivalent of 2 ⁇ 10 7 c.f.u. were injected i.p. into C57BL/6 mice bearing B16F10 melanomas. Survival was determined daily, or at two to four day intervals.
  • PBS phosphate buffered saline
  • Results are presented in FIG. 9A and are expressed as percent survival.
  • the bacteria were washed in phosphate buffered saline and 1.0 ⁇ 10 9 were injected into the ear vein of Sinclair swine (n 4/strain). Survival was determined daily, or at two to four day intervals.
  • Results are presented in FIG. 9B and are expressed as percent survival.
  • Salmonella WT 14028 with the msbB ⁇ modification were assayed to determine if they possessed tumor targeting ability using the B16F10 melanoma subcutaneous animal model.
  • the msbB ⁇ clone, YS8211 was grown in LB media lacking sodium chloride at 37° C. with shaking at 250 rpm to an OD 600 of 0.8.
  • An aliquot of 2.0 ⁇ 10 6 c.f.u. was injected i.v. into C57BL/6 mice which had been implanted with 2 ⁇ 10 5 B16 melanoma cells 16 days prior to the bacterial infection.
  • mice were sacrificed and tumors and livers assayed for the presence of the bacteria by homogenization and plating of serial dilutions.
  • Results are presented in FIG. 10 and are expressed as c.f.u. bacteria/g tissue.
  • a positive ratio of tumor to liver 700:1 was found at 2 days, and increased to a positive ratio of 2000:1 at 5 days.
  • the msbB ⁇ mutant maintained the ability to target to a solid cancer tumor.
  • Salmonella typhimurium 14028 msbB ⁇ clones YS8211, YS8212, YS7216, and YS1629 and WT 14028 (control) were grown in LB media lacking sodium chloride at 37° C. with shaking at 250 rpm to an OD 600 of 0.8. An aliquot of 2.0 ⁇ 10 6 c.f.u. was injected i.p. into C57BL/6 mice which had been implanted with 2 ⁇ 10 5 B16 melanoma cells 8 days prior to the bacterial infection. Tumor volume was monitored over time.
  • Results are presented in FIG. 11. Two of the strains, YS8211 and YS1629, showed significant tumor retardation, i.e., tumor growth inhibition.
  • bacteria with or without the msbB mutation were grown in the presence or absence of 1 mM EDTA or 10 mM sodium citrate in Luria Broth (LB) lacking sodium chloride.
  • An overnight culture of each of the bacterial strains was diluted 1 to 100 in fresh media, and grown at 37° C. with shaking at 250 rpm. The effect on growth was determined by spectrophotometric readings at an OD 600 .
  • WT 14028 and msbB ⁇ clone YS8211 were grown in the presence or absence of 1 mM EDTA (FIG. 12A). EDTA did not inhibit the growth of WT 14028. In contrast, the msbB ⁇ clone showed near complete cessation of growth after 3 hours in the presence of EDTA.
  • WT 14028 and msbB ⁇ clone YS862 were grown in the presence and absence of 10 mM sodium citrate (FIG. 12B).
  • the msbB + WT 14028 strain showed little inhibition by sodium citrate compared to the msbB ⁇ strain which showed near complete cessation of growth after 3 hours in the presence of sodium citrate.
  • the msbB ⁇ Salmonella mutants exhibited sensitivity to chelating agents which promote eradication of the bacteria, a characteristic which is similar to an antibiotic effect. It is envisioned that such a characteristic would be advantageous for use of msbB ⁇ Salmonella mutants for in vivo therapy.
  • the hyperinvasive pur strain YS72, its msbB ⁇ strain, YS7216, and a derivative of YS7216, YS1629 were grown in the presence of increasing concentrations of EDTA.
  • a fresh culture of YS72, its msbB ⁇ strain YS7216 and its faster-growing derivative YS1629 were diluted 1 to 100 in fresh, zero salt LB media containing 0, 0.25, 0.5, 1.0 or 2.0 mM EDTA and grown at 37° C. with 225 RPM for 4 hours, and c.f.u. was determined by plating serial dilutions onto LB plates (Table I).
  • Spontaneous derivatives of msbB ⁇ strains YS8211 and YS7216 were selected from in vitro culture on non-modified LB medium based upon enhanced growth characteristics. These bacterial strains were grown to OD 600 of 0.8 and c.f.u. ranging from 1 ⁇ 10 2 to 1 ⁇ 10 8 were injected i.v. into the tail vein of C57BL/6 mice. Acute lethality was determined at 3 days, and the LD 50 determined as described by Welkos and O'Brien (Methods in Enzymology 235:29-39, 1994). The results are presented in Table II.
  • msbB + strains were grown in LB broth or LB plates containing 1.5% agar at 37° C.
  • msbB strains were grown in modified LB containing 10 g tryptone, 5 g yeast extract, 2 ml 1N CaCl 2 and 2 ml 1N MgSO 4 per liter, adjusted to pH 7 using 1N NaOH.
  • LB lacking NaCl was used, with 4 mg/l tetracycline.
  • Liquid cultures were shaken at 225 rpm.
  • the msbbl::tet was transduced to auxotrophic strains to generate YS1604 (msbB ⁇ , pur ⁇ , hyperinvasive), YS7232 (msbB ⁇ , purI ⁇ , hyperinvasive), YS7244 (msbB ⁇ , purI ⁇ , aroA ⁇ hyperinvasive), YS1482 (msbB ⁇ , purI ⁇ , purA ⁇ ).
  • PBS phosphate buffered saline
  • tumors were excised, weighed, homogenized, and c.f.u. determined by plating serial dilutions onto modified LB described above.
  • Results are presented in Table III and are expressed as c.f.u. per gram tumor tissue. Some of the strains, YS8211, YS1604, and YS7232 show high levels of c.f.u. within the tumors, whereas YS7244 and YS1482 are approximately 500 to 5000 times less.
  • Salmonella strain YS1456 The generation of Salmonella strain YS1456 from the wild type Salmonella typhimurium is outlined in FIG. 15.
  • the wild type Salmonella typhimurium was transduced with purl 1757::Tn10 which conferred tetracycline-resistance, resulting in strain YS1451.
  • Strain YS1451 was then subjected to a Bochner selection to render the strain tet sensitive and introduce tets gene and introduce a purl deletion (Bochner et al. 1980, J. Bacteriol. 143:926-933), yielding the strain YS1452.
  • Strain YS1452 was tet s and purI ⁇ .
  • Strain 1452 was then transduced with msbB1::tet via bacteriophage P22, using strain YS8211 (msbB::tet) as the donor.
  • YS1453 The resulting strain, YS1453, was initially sensitive to 10 mM ethylene glycol bis((b-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA), spontaneously reverted to a EGTA-resistant phenotype.
  • EGTA ethylene glycol bis((b-aminoethyl ether)-N,N,N′,N′-tetraacetic acid
  • YS1454 One such revertant, was selected by plating YS1453 on EGTA (2mM in Luria agar).
  • Strain YS1454 was then transduced with the msbB2( ⁇ ) bla sacB chromosomal element, selecting for ampicillin resistance. This transduction process brought in a second version of the disrupted msbB gene, denoted msbB2( ⁇ ) as well as the bla and sacB genes.
  • the bla gene is responsible for the transcription of the enzyme ⁇ -lactamase, which metabolizes ampicillin, and was used to select for ampicillin resistant transductants.
  • the sacB gene is responsible for the conversion of sucrose into a toxic chemical, levan, that is lethal to the host cells, and was subsequently used to select for recombinants which lose or have mutations in sacb (see Section 7.2.1 for improved pre-selection methods with sucrose).
  • the presence of the bla and sacB genes allowed the selection of the amp r and suc s strain (denoted as strain YS1455), which contained both the msbB1::tet and msbB2( ⁇ ) genes.
  • Strain YS1455 was then plated on Luria Bertani (LB) sucrose to select a suc r amp s tet s derivative to remove msbB1::tet and restore antibiotic sensitivity.
  • the derivative was denoted as strain YS1456.
  • YS1456 has deletion mutations in purl and msbB. It is also tet s amp s and EGTA r .
  • Salmonella strain YS1646 from the wild type Salmonella typhimurium (wild type strain ATCC 14028) is outlined in FIG. 16.
  • the wild type Salmonella typhimurium was mutagenized with nitrosoguanidine and ultraviolet (UV) light and selected for hyperinvasiveness in melanoma cells.
  • the resistant strain, denoted YS72, were confirmed to possess tumor-hyperinvasiveness pur ⁇ and xyl ⁇ 0 properties (Pawelek et al., 1997, Caner Res 57: 4537-4544).
  • strain YS72 was transduced with the purl 1757::Tn10 gene, which conferred tetracycline-resistance.
  • the donor for the purI 1757::Tn10 gene was Salmonella strain TT11 (purl 1757::Tn10).
  • the donor strain was originally obtained from the Salmonella Genetic Stock Center (Dept. of Biological Science, Univ. Calgary, Calgary, Alberta, Canada T2N 1N4). Transduction was performed using bacteriophage P22 (mutant HT105/1 int-201). The transductant, denoted YS1641, was isolated following selection on tetracycline.
  • Strain YS1641 was then subjected to a Bochner selection to remove the tet gene and introduce a purI gene deletion (Bochner et al., 1980, J. Bacteriol. 143:926-933), yielding strain YS1642.
  • Strain YS1642 was tet s and purI ⁇ . The selection of a tet ⁇ -deleted strain allowed further genetic modification (e.g., msbB gene disruption, see next paragraph) using tet gene transduction.
  • Strain YS1642 has a tight purine requirement due to purI( ⁇ ), and has been shown to revert to purI + at a frequency of less than 1 in 1010 cells.
  • Strain YS1642 was then transduced with msbB1::tet via bacteriophage P22, using strain YS8211 (msbB::tet) as the donor.
  • the DNA sequence for the msbB gene is shown in FIG. 1.
  • the tet gene in the msbB1::tet gene confers resistance to 5 mg/L of tetracycline.
  • the resulting strain thus obtained was YS1643.
  • Strain YS1643 was initially sensitive to 10 mM ethylene glycol bis((b-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA), spontaneously reverted to a EGTA-resistant phenotype.
  • EGTA ethylene glycol bis((b-aminoethyl ether)-N,N,N′,N′-tetraacetic acid
  • YS1644 One such revertant, was selected by plating YS1643 on EGTA (2 mM in Luria agar).
  • Strain YS1644 was then transduced with the msbB2(A) bla sacB chromosomal element. This transduction process brought in a second version of the disrupted msbB gene, denoted as msbB2( ⁇ ) as well as the bla and sacB genes.
  • the bla gene is responsible for the transcription of the enzyme ⁇ -lactamase, which metabolizes ampicillin, and was subsequently used to select transductants.
  • the sacB gene is responsible for the conversion of sucrose into a toxic chemical, levan, that is lethal to the host cells, and was used to select for recombinants.
  • the presence of the bla and sacB genes allowed the selection of the amp r and suc s strain (denoted as strain YS1645), which contained both the msbB1::tet and msbB2(A) genes.
  • Strain YS1645 was plated on Luria-Bertani (LB) sucrose to select a suc r amp s tet s derivative to remove the msbB::tet gene and restore antibiotic sensitivity (i.e., a derivative with deletion of msbb1::tet bla sacb). This derivative was denoted as strain YS1646.
  • YS1646 has deletion mutations in purI, and msbB. It is also tet s , amp s , and EGTA r .
  • IV Intravenous (IV) administration of YS1646, an attentuated strain of Salmonella typhimurium , resulted in selective replication within tumors, and concomitant inhibition of tumor growth (see FIG. 17 and Table IV).
  • YS1646 The efficacy of YS1646 was evaluated in mice previously implanted with B16-F10 melanoma. In this study a single IV dose of YS1646 at 10 4 , 10 5 or 10 6 cfu/mouse significantly reduced tumor size when compared to control treatment, and the degree of tumor size reduction was dose-related. The efficacy observed with the highest dose of YS1646 was superior to that with the positive control, CYTOXANTM (also known as cyclophosamide), whereas the efficacy with the mid-dose of YS1646 was equivalent to that with, CYTOXANTM.
  • CYTOXANTM also known as cyclophosamide
  • YS1646 efficacy induced by YS1646 was induced by a single IV dose, whereas that induced by CYTOXANTM was multiple IV doses (given weekly, for 3 weeks).
  • Each dosage group was comprised of 10 tumor-bearing animals, which were randomized prior to bacteria administration. Mice were administered bacteria on Day 7, and tumor volumes were measured on Days 10, 13, 17, 20, and 24.
  • CYTOXANTM cyclophosphamide
  • YS1646 does not cause lethality, in contrast to the parental wide type strain ATCC 14028, which causes 100% mortality at a dose of 1 ⁇ 10 2 cfu/mouse. This indicates that YS1646 is greater than 10,000-fold less virulent than the parental wild type strain.
  • the antitumor efficacy was observed at doses of 10 4 to 10 6 cfu/mouse, whereas lethality was not observed until the doses were >106 cfu/mouse.
  • the dose inducing mortality was 1 to 100-fold greater than the dose inducing anti-tumor efficacy (see FIG. 18).
  • Groups were divided into the following treatment categories: 1) untreated control, 2) ampicillin-treated, 3) ciprofloxacin-treated, and 4) ciprofloxacin and ampicillin treated.
  • Antibiotic treatment was initiated 3 days following bacteria administration and animals were observed daily for appearance and mortality for 14 days. Results presented herein demonstrate that use of antibiotic was able to supress mortality following lethal bacterial infections (see FIG. 18).

Abstract

The present invention is directed to mutant Salmonella sp. having a genetically modified msbB gene in which the mutant Salmonella is capable of targeting solid tumors. The invention is also directed to Salmonella sp. containing a genetically modified msbB gene as well as an genetic modification in a biosynthetic pathway gene such as the purl gene. The present invention further relates to the therapeutic use of the mutant Salmonella for growth inhibition and/or reduction in volume of solid tumors.

Description

  • This application is a continuation-in-part of application Ser. No. 08/926,636, filed Sep. 10, 1997, the entire disclosure of which is incorporated by reference herein in its entirety.[0001]
  • 1. FIELD OF THE INVENTION
  • The present invention is concerned with the isolation of a gene of Salmonella which, when genetically disrupted, reduces both virulence and septic shock caused by this organism and increases sensitivity to agents which promote eradication of the bacteria, e.g., chelating agents. The nucleotide sequence of this gene and the means for its genetic disruption are provided, and examples of the use of tumor-targeted bacteria which possess a disruption in this gene to inhibit growth of cancers, including, but not limited to, melanoma, colon cancer, and other solid tumors are described. The present invention also provides for the genetic disruption of this gene in combination with disruption of an auxotrophic gene. [0002]
  • 2. BACKGROUND OF THE INVENTION
  • Citation or identification of any reference in [0003] Section 2, or any section of this application shall not be construed as an admission that such reference is available as prior art to the present invention.
  • A major problem in the chemotherapy of solid tumor cancers is delivery of therapeutic agents, such as drugs, in sufficient concentrations to eradicate tumor cells while at the same time minimizing damage to normal cells. Thus, studies in many laboratories are directed toward the design of biological delivery systems, such as antibodies, cytokines, and viruses for targeted delivery of drugs, pro-drug converting enzymes, and/or genes into tumor cells. Houghton and Colt, 1993, New Perspectives in Cancer Diagnosis and Management 1: 65-70; de Palazzo, et al., 1992a, Cell. Immunol. 142:338-347; de Palazzo et al., 1992b, Cancer Res. 52: 5713-5719; Weiner, et al., 1993a, J. Immunotherapy 13:110-116; Weiner et al., 1993b, J. Immunol. 151:2877-2886; Adams et al., 1993, Cancer Res. 53:4026-4034; Fanger et al., 1990, FASEB J. 4:2846-2849; Fanger et al., 1991, Immunol. Today 12:51-54; Segal, et al., 1991, Ann N.Y. Acad. Sci. 636:288-294; Segal et al., 1992, Immunobiology 185:390-402; Wunderlich et al., 1992; Intl. J. Clin. Lab. Res. 22:17-20; George et al., 1994, J. Immunol. 152:1802-1811; Huston et al., 1993, Intl. Rev. Immunol. 10:195-217; Stafford et al., 1993, Cancer Res. 53:4026-4034; Haber et al., 1992, Ann. N.Y. Acad. Sci. 667:365-381; Haber, 1992, Ann. N.Y. Acad. Sci. 667: 365-381; Feloner and Rhodes, 1991, Nature 349:351-352; Sarver and Rossi, 1993, AIDS Research & Human Retroviruses 9:483-487; Levine and Friedmann, 1993, Am. J. Dis. Child 147:1167-1176; Friedmann, 1993, Mol. Genetic Med. 3:1-32; Gilboa and Smith, 1994, Trends in Genetics 10:139-144; Saito et al., 1994, Cancer Res. 54:3516-3520; Li et al., 1994, Blood 83:3403-3408; Vieweg et al., 1994, Cancer Res. 54:1760-1765; Lin et al., 1994, Science 265:666-669; Lu et al., 1994, Human Gene Therapy 5:203-208; Gansbacher et al., 1992, Blood 80:2817-2825; Gastl et al., 1992, Cancer Res. 52:6229-6236. [0004]
  • 2.1 Bacterial Infections and Cancer [0005]
  • Regarding bacteria and cancer, an historical review reveals a number of clinical observations in which cancers were reported to regress in patients with bacterial infections. Nauts et al., 1953, Acta Medica. Scandinavica 145:1-102, (Suppl. 276) state: [0006]
  • The treatment of cancer by injections of bacterial products is based on the fact that for over two hundred years neoplasms have been observed to regress following acute infections, principally streptococcal. If these cases were not too far advanced and the infections were of sufficient severity or duration, the tumors completely disappeared and the patients remained free from recurrence. [0007]
  • Shear, 1950, J. A.M.A. 142:383-390 (Shear), observed that 75 percent of the spontaneous remissions in untreated leukemia in the Children's Hospital in Boston occurred following an acute episode of bacterial infection. Shear questioned: [0008]
  • Are pathogenic and non-pathogenic organisms one of Nature's controls of microscopic foci of malignant disease, and in making progress in the control of infectious diseases, are we removing one of Nature's controls of cancer? [0009]
  • Subsequent evidence from a number of research laboratories indicated that at least some of the anti-cancer effects are mediated through stimulation of the host immune system, resulting in enhanced immuno-rejection of the cancer cells. For example, release of the lipopolysaccharide (LPS) endotoxin by gram-negative bacteria such as Salmonella triggers release of tumor necrosis factor, TNF, by cells of the host immune system, such as macrophages, Christ et al., 1995, Science 268:80-83. Elevated TNF levels in turn initiate a cascade of cytokine-mediated reactions which culminate in the death of tumor cells. In this regard, Carswell et al., 1975, Proc. Natl. Acad. Sci. USA 72:3666-3669, demonstrated that mice injected with bacillus Calmette-Guerin (BCG) have increased serum levels of TNF and that TNF-positive serum caused necrosis of the sarcoma Meth A and other transplanted tumors in mice. Further, Klimpel et al., 1990, J. Immunol. 145:711-717, showed that fibroblasts infected in vitro with Shigella or Salmonella had increased susceptibility to TNF. [0010]
  • As a result of such observations as described above, immunization of cancer patients with BCG injections is currently utilized in some cancer therapy protocols. See Sosnowski, 1994, Compr. Ther. 20:695-701; Barth and Morton, 1995, Cancer 75 (Suppl. 2):726-734; Friberg, 1993, Med. Ooncol. Tumor. Pharmacother. 10:31-36 for reviews of BCG therapy. [0011]
  • 2.2 Parasites and Cancer Cells [0012]
  • Although the natural biospecificity and evolutionary adaptability of parasites has been recognized for some time and the use of their specialized systems as models for new therapeutic procedures has been suggested, there are few reports of, or proposals for, the actual use of parasites as vectors. [0013]
  • Lee et al., 1992, Proc. Natl. Acad. Sci. USA 89:1847-1851 (Lee et al.) and Jones et al., 1992, Infect. Immun. 60:2475-2480 (Jones et al.) isolated mutants of Salmonella typhimurium that were able to invade HEp-2 (human epidermoid carcinoma) cells in vitro in significantly greater numbers than the wild type strain. The “hyperinvasive” mutants were isolated under conditions of aerobic growth of the bacteria that normally repress the ability of wild type strains to invade HEp-2 animal cells. However, Lee et al. and Jones et al. did not suggest the use of such mutants as therapeutic vectors, nor did they suggest the isolation of tumor-specific bacteria by selecting for mutants that show infection preference for melanoma or other cancers over normal cells of the body. Without tumor-specificity or other forms of attenuation, such hyperinvasive Salmonella typhimurium as described by Lee et al. and Jones et al. would likely be pan-invasive, causing wide-spread infection in the cancer patient. [0014]
  • 2.3 Tumor-targeted Bacteria [0015]
  • Genetically engineered Salmonella have been demonstrated to be capable of tumor targeting, possess anti-tumor activity and are useful in delivering effector genes such as the herpes simplex thymidine kinase (HSV TK) to solid tumors (Pawelek et al., WO 96/40238). Two significant considerations for the in vivo use of bacteria are their virulence and ability to induce tumor necrosis factor a (TNFα)-mediated septic shock. As TNFα-mediated septic shock is among the primary concerns associated with bacteria, modifications which reduce this form of an immune response would be useful because TNFα levels would not become toxic, and a more effective concentration and/or duration of the therapeutic vector could be used. [0016]
  • 2.4 Modified Bacterial Lipid A [0017]
  • Modifications to the lipid composition of tumor-targeted bacteria which alter the immune response as a result of decreased induction of TNFα production were suggested byPawelek et al. (Pawelek et al., WO 96/40238). Pawelek et al. provided methods for isolation of genes from Rhodobacter responsible for monophosphoryl lipid A (MLA) production. MLA acts as an antagonist to septic shock. Pawelek et al. also suggested the use of genetic modifications in the lipid A biosynthetic pathway, including the mutation firA, which codes for the third enzyme UDP-3-O (R-30 hydroxylmyristoly)glucosamine N-acyltransferase in lipid A biosynthesis (Kelley et al., 1993, J. Biol. Chem. 268: 19866-19874). Pawelek et al. showed that mutations in the firA gene induce lower levels of TNFα. However, these authors did not suggest enzymes which modify the myristate portion of the lipid A molecule. Furthermore, Pawelek et al. did not suggest that modifications to the lipid content of bacteria would alter their sensitivity to certain agents, such as chelating agents. [0018]
  • In [0019] Escherichia coli, the gene msbb (mlt) which is responsible for the terminal myristalization of lipid A has been identified (Engel, et al., 1992, J. Bacteriol. 174:6394-6403; Karow and Georgopoulos 1992, J. Bacteriol. 174: 702-710; Somerville et al., 1996, J. Clin. Invest. 97: 359-365). Genetic disruption of this gene results in a stable non-conditional mutation which lowers TNFα induction (Somerville et al., 1996, J. Clin. Invest. 97: 359-365). These references, however, do not suggest that disruption of the msbb gene in tumor-targeted Salmonella vectors would result in bacteria which are less virulent and more sensitive to chelating agents.
  • The problems associated with the use of bacteria as gene delivery vectors center on the general ability of bacteria to directly kill normal mammalian cells as well as their ability to overstimulate the immune system via TNFα which can have toxic consequences for the host (Bone, 1992 JAMA 268: 3452-3455; Dinarello et al., 1993 JAMA 269: 1829-1835). In addition to these factors, resistance to antibiotics can severely complicate coping with the presence of bacteria within the human body (Tschape, 1996, D T W Dtsch Tierarztl Wochenschr 1996 103:273-7; Ramos et al., 1996, Enferm Infec. Microbiol. Clin. 14: 345-51). [0020]
  • Hone and Powell, W097/18837 (“Hone and Powell”), disclose methods to produce gram-negative bacteria having non-pyrogenic Lipid A or LPS. Although Hone and Powell broadly asserts that conditional mutations in a large number of genes including msbB, kdsA, kdsb, kdtA, and htrB, etc. can be introduced into a broad variety of gram-negative bacteria including E. coli, Shigella sp., Salmonella sp., etc., the only mutation exemplified is an htrB mutation introduced into E. coli. Further, although Hone and Powell propose the therapeutic use of non-pyrogenic Salmonella with a mutation in the msbB gene, there is no enabling description of how to accomplish such use. Moreover, Hone and Powell propose using non-pyrogenic bacteria only for vaccine purposes. [0021]
  • The objective of a vaccine vector is significantly different from the presently claimed tumor-targeted vectors. Thus, vaccine vectors have requirements quite different from tumor-targeted vectors. Vaccine vectors are intended to elicit an immune response. A preferred live bacterial vaccine must be immunogenic so that it elicits protective immunity; however, the vaccine must not be capable of excessive growth in vivo which might result in adverse reactions. According to the teachings of Hone and Powell, a suitable bacterial vaccine vector is temperature sensitive having minimal replicative ability at normal physiological ranges of body temperature. [0022]
  • In contrast, preferred tumor-targeted parasitic vectors, such as but not limited to Salmonella, are safely tolerated by the normal tissues of the body such that pathogenesis is limited, yet the vectors target to tumors and freely replicate within them. Thus, vaccine vectors which replicate minimally at normal body temperatures, would not be suitable for use as tumor-targeted vectors. [0023]
  • The preferred properties of tumor-specific Salmonella strains include 1) serum resistance, allowing the parasite to pass through the vasculature and lymphatic system in the process of seeking tumors, 2) facultative anaerobiasis, i.e., ability to grow under anaerobic or aerobic conditions allowing amplification in large necrotic tumors which are hypoxic as well as small metastatic tumors which may be more aerobic, 3) susceptibility to the host's defensive capabilities, limiting replication in normal tissues but not within tumors where the host defensive capabilities may be impaired, 4) attenuation of virulence, whereby susceptibility to the host defenses may be increased, and the parasite is tolerated by the host, but does not limit intratumoral replication, 5) invasive capacity towards tumor cells, aiding in tumor targeting and anti-tumor activity, 6) motility, aiding in permeation throughout the tumor, 7) antibiotic sensitivity for control during treatment and for post treatment elimination (e.g., sensitivity to ampicillin, chloramphenicol, gentamicin, ciprofloxacin), and lacking antibiotic resistance markers such as those used in strain construction, and 8) low reversion rates of phenotypes aiding in the safety to the recipient individual. [0024]
  • 3. SUMMARY OF THE INVENTION
  • The present invention provides a means to enhance the safety of tumor-targeted bacteria, for example, by genetic modification of the lipid A molecule. The modified tumor-targeted bacteria of the present invention induce TNFα less than the wild type bacteria and have reduced ability to directly kill normal mammalian cells or cause systemic disease compared to the wild type strain. The modified tumor-targeted bacteria of the present invention have increased therapeutic efficacy, i.e., more effective dosages of bacteria can be used and for extended time periods due to the lower toxicity in the form of less induced TNFα and systemic disease. [0025]
  • The present invention provides compositions and methods for the genetic disruption of the msbB gene in bacteria, such as Salmonella, which results in bacteria, such as Salmonella, possessing a lesser ability to elicit TNFα and reduced virulence compared to the wild type. In one embodiment, the invention provides for improved methods for selecting genetic disruptions of the msbB gene. Additionally, the genetically modified bacteria have increased sensitivity to a chelating agent compared to bacteria with the wild type msbB gene. In a preferred embodiment, Salmonella having a disrupted msbB gene, which are hyperinvasive to tumor tissues, are able to replicate within the tumors, and are useful for inhibiting the growth and/or reducing the tumor volume of sarcomas, carcinomas, lymphomas or other solid tumor cancers, such as germ line tumors and tumors of the central nervous system, including, but not limited to, breast cancer, prostate cancer, cervical cancer, uterine cancer, lung cancer, ovarian cancer, testicular cancer, thyroid cancer, astrocytoma, glioma, pancreatic cancer, stomach cancer, liver cancer, colon cancer, and melanoma. [0026]
  • In an embodiment of the present invention, the bacteria are attenuated by other means, including but not limited biosynthetic pathway mutations leading to auxotrophy. In one specific embodiment, the biosynthetic pathway mutation is a genetic disruption of the puri gene. In another embodiment, the bacteria express pro-drug converting enzymes including but not limited to HSV-TK, cytosine deaminase (CD), and p450 oxidoreductase. [0027]
  • The present invention also provides a means for enhanced sensitivity for use in terminating therapy and for post therapy elimination. According to one embodiment of the present invention, the tumor-targeted bacteria having a genetically modified lipid A also have enhanced susceptibility to certain agents, e.g., chelating agents. It is a further advantage to modify tumor-targeted bacteria in this way because it increases the ability to eliminate the bacteria with agents which have an antibiotic-like effect, such as chelating agents including, but not limited to, Ethylenediaminetetraacetic Acid (EDTA), Ethylene Glycol-bis(β-aminoethyl Ether) N,N,N′,N′,-Tetraacetic Acid (EGTA), and sodium citrate. Modification to enhance the ability to eliminate the bacteria via exogenous means, such as the administration of an agent to which the genetically modified bacteria are more sensitive than their wild type counterparts, is therefore useful. [0028]
  • The present invention further provides for a Salmonella strain comprising deletion mutations in both the msbB gene as well as an auxotrophic gene. In a specific embodiment, the auxotrophic deletion mutation affects the purl gene. In a preferred embodiment, these mutations lead to increased safety of the strain. In another preferred embodiment, the strain also carries other mutations described herein which increase efficacy of the strain but are not essential for its safety. [0029]
  • 4. DEFINITIONS
  • As used herein, Salmonella encompasses all Salmonella species, including: [0030] Salmonella typhi, Salmonella choleraesuis, and Salmonella enteritidis. Serotypes of Salmonella are also encompassed herein, for example, typhimurium, a subgroup of Salmonella enteritidis, commonly referred to as Salmonella typhimurium.
  • Attenuation: Attenuation is a modification so that a microorganism or vector is less pathogenic. The end result of attenuation is that the risk of toxicity as well as other side-effects is decreased, when the microorganism or vector is administered to the patient. [0031]
  • Virulence: Virulence is a relative term describing the general ability to cause disease, including the ability to kill normal cells or the ability to elicit septic shock (see specific definition below). [0032]
  • Septic shock: Septic shock is a state of internal organ failure due to a complex cytokine cascade, initiated by TNFα. The relative ability of a microorganism or vector to elicit TNFα is used as one measure to indicate its relative ability to induce septic shock. [0033]
  • Chelating agent sensitivity: Chelating agent sensitivity is defined as the effective concentration at which bacteria proliferation is affected, or the concentration at which the viability of bacteria, as determined by recoverable colony forming units (c.f.u.), is reduced.[0034]
  • 5. BRIEF DESCRIPTION OF THE FIGURES
  • The present invention may be understood more fully by reference to the following detailed description, illustrative examples of specific embodiments and the appended figures. [0035]
  • FIG. 1. The complete DNA sequence of the Salmonella wild type (WT) 14028 msbB gene (SEQ ID NO:1) and the deduced amino acid sequence of the encoded protein (SEQ ID NO:2). [0036]
  • FIGS. [0037] 2A-2C. Knockout construct generated using the cloned Salmonella WT 14028 msbb gene. The cloned gene was cut with SphI and MluI thereby removing approximately half of the msbB coding sequence, and the tetracycline resistance gene (TET) from pBR322 cut with AatII and AvaI was inserted after blunt-ending using the Klenow fragment of DNA polymerase I. A=Knockout construct. B=Salmonella chromosomal copy of msbB. C=Salmonella disrupted chromosomal copy of msbB after homologous recombination. The start codon (ATG) and stop codon (TAA) and restriction sites AseI, BamHI, SphI, MluI, and EcoRV are shown. The position of two primers, P1 and P2 which generate two different sized PCR products for either wild type or disrupted msbB are shown.
  • FIGS. [0038] 3A-3C. Southern blot analysis of chromosomally disrupted Salmonella WT 14028 msbB. A) Southern blot probed with the tetracycline gene, demonstrating its presence in the plasmid construct and the two clones, and its absence in the WT 14028 bacteria. B) Southern blot of a similar gel probed with an 32P-labeled AseI/BamH1 fragment derived from the cloned msbB. The AseI enzyme cuts upstream of msbB, and the BamH1 cuts in one location in the wild type, but in a second location in the tetracycline gene which results in a higher molecular weight product. Lane 1 (KO) shows the position of the band in the knockout construct, compared to the WT 14028 in lane 2 (WT). Lanes 3 and 4 show the clones YS8211 and YS861 with a higher molecular weight product. C) Southern blot of a similar gel probed with an 32P-labeled mluI fragment derived from the cloned msbB. See text Section 7.2 for details.
  • FIG. 4. TNFα induction by [0039] live Salmonella WT 14028 in mice. 1 X 108 live bacteria in 0.1 cc phosphate buffered saline of the wild type or msbB disrupted strains were injected i.v. in the tail vein of Balb/c mice. The bar graph indicates the TNFα induction with error bars. Clone YS8211 induces TNFα 32% compared to Salmonella WT 14028.
  • FIG. 5. TNFα response by Sinclair swine to live [0040] Salmonella WT 14028 and msbB clone YS8212. TNFα levels were measured at 1.5 and 6.0 hours following i.v. introduction of 1×109 c.f.u. Salmonella WT 14028 and YS8212. At 1.5 hours TNFα response was significantly lower (p≦0.011) in the msbB deletion mutant compared to the wild type.
  • FIGS. [0041] 6A-6B. Respiratory level changes induced by LPS from WT 14028 and msbB clone YS8212. Sinclair swine were injected with A) 5 μg/kg purified LPS or B) 500 μg/kg purified LPS and respiration rate was determined. The 500 μg/kg of LPS from Salmonella WT 14028 raised the rate of respiration to more than 4 times normal, whereas the rate of respiration in msbB LPS-treated animals was less than doubled.
  • FIG. 7. TNFα induction by [0042] live Salmonella WT 14028 in human monocytes. Human monocytes isolated from peripheral blood were exposed to increasing amounts of Salmonella c.f.u. At 1.0×105 c.f.u., concentrations of TNFα induced by WT 14028 were more than 3 times higher than those induced by a number of msbB clones, i.e., YS8211, YS8212, YS8658, and YS1170.
  • FIG. 8. TNFα production by human monocytes. Human monocytes isolated from peripheral blood were exposed to increasing amounts of purified LPS. As little as 1 nanogram of LPS from wild type was sufficient to elicit a measurable TNFα response and was maximal at 10 ng. In contrast, 100 μg of LPS from each of a number of msbB[0043] clones was insufficient to generate any response. Thus, at 10 ng LPS, the concentration of TNFα induced by Salmonella WT 14028 was at least 105 times higher than concentrations of TNFα induced by the independent msbB knockouts, i.e., YS7216 and YS8211, and the derivatives, i.e., YS1170, YS8644, YS1604, YS8212, YS8658, YS1601, YS1629.
  • FIGS. [0044] 9A-9B. Survival of mice and Sinclair swine, injected with 2×107 or 1×109 respectively of live bacteria. A) WT 14028 killed all the mice in 4 days, whereas the msbB clone YS862 spared 90% of the mice past 20 days. B) Similarly, WT 14028 killed all the swine in 3 days, whereas the msbB clone YS8212 spared 100% of the swine past 20 days.
  • FIG. 10. Biodistribution of msbB[0045] Salmonella YS8211 in B16FlO melanoma tumors. At 5 days, the ratio of msbB Salmonella within the tumors compared to those in the liver exceeded 1000:1.
  • FIG. 11. Tumor retardation by msbB[0046] Salmonella. B16F10 melanoma tumors were implanted in the flank of C57BL/6 mice and allowed to progress to day 8. Mice either received no bacteria (control) or msbB strains YS8211, YS8212, YS7216, YS1629. Two of the strains, YS8211 and YS1629 retarded tumor progression significantly, whereas strains YS7216 and YS8212 did not.
  • FIGS. [0047] 12A-12B. Sensitivity of WT 14028 and msbB disrupted bacteria to chelating agents. Wild type and msbb disrupted Salmonella clone YS8211 and YS862 were grown in LB broth lacking sodium chloride (LB-zero), in the presence or absence of 1 mM EDTA (FIG. 12A) or in the presence or absence of 10 mM sodium citrate (FIG. 12B). The OD600 was determined and plotted as a function of time. The msbB+ strain showed little inhibition by EDTA or sodium citrate, compared to the msbB strains which showed near complete cessation of growth after 3 hours for EDTA or sodium citrate.
  • FIGS. [0048] 13A-13B. Survival of msbB bacteria within murine macrophages. Murine bone marrow-derived macrophages (FIG. 13A) and a murine macrophage cell line, J774, (FIG. 13B) were used as hosts for bacterial internalization and quantified over time. The data are presented as a percentage of initial c.f.u.
  • FIG. 14. Conversion of msbB1(Δ): :tet to tet[0049] S using the positive selection suicide vector pCVD442 carrying a second version of the msbB (msbB2 (Δ) ampR sacB+)
  • FIG. 15. Schematic diagram of the derivation of strain YS1456 from wild type [0050] Salmonella typhimurium. See text Section 8.1 for details.
  • FIG. 16. Schematic diagram of the derivation of strain YS1646 from wild type [0051] Salmonella typhimurium. See text Section 8.2 for details.
  • FIG. 17. Effect of YS1646 dose on B16-B10 murine melanoma tumor growth. [0052]
  • FIG. 18. Antibiotic suppression of YS1646-induced mortality following lethal infection.[0053]
  • 6. DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based on the isolation of a gene of Salmonella, i.e., msbB, which, when present in its normal form, contributes to TNFα induction, general virulence, survival within macrophages, and insensitivity to certain agents which promote eradication of the bacteria. The present invention is directed to the genetic modification of the gene which results in disrupting the normal function of the product of the gene, and the incorporation of the genetic modification into tumor-targeted bacteria, including Salmonella, for therapeutic use. In a preferred embodiment, the bacteria have a genetic modification of the msbB gene as well as genetic modification of a gene in a biosynthetic pathway, such as the purI gene, resulting in an auxotrphic strain. [0054]
  • In a preferred embodiment, the genetically modified bacteria are used in animals, including humans, for reduction of volume and/or growth inhibition of solid tumors. [0055]
  • In an additional preferred embodiment, bacteria useful for the present invention show preference for attachment to and penetration into certain solid tumor cancer cells or have an enhanced propensity to proliferate in tumor tissues as compared to normal tissues. These bacteria, include but are not limited to Salmonella, having a natural ability to distinguish between cancerous or neoplastic cells tissues and normal cells/tissues. [0056]
  • Alternatively, tumor cell-specific bacteria useful for the invention may be selected for and/or improved in tumor targeting ability using the methods described by Pawelek et al., WO 96/40238 incorporated herein by reference. Pawelek et al. describe methods for isolating tumor cell-specific bacteria by cycling a microorganism through preselected target cells, preferably solid tumor cells in vitro, or through a solid tumor in vivo, using one or more cycles of infection. [0057]
  • 6.1 Isolation/Identification of a Gene Involved in Virulence [0058]
  • The [0059] E. coli gene, msbB, has been shown to be involved in myristilization of lipid A (Somerville et al., 1996, J. Clin. Invest. 97:359-365.) The chromosomal organization of the E. coli msbB gene and the DNA sequence coding for the msbB gene have been described (Engel, et al., 1992, J. Bacteriol. 174:6394-6403; Karow and Georgopoulos, 1992, J. Bacteriol. 174: 702-710; Somerville et al., 1996, J. Clin. Invest. 97: 359-365).
  • As shown in the present invention, the msbB gene can be isolated from bacterial strains, other than [0060] E. coli, using low stringency DNA/DNA hybridization techniques known to those skilled in the art. (Sambrook et al., Molecular Cloning, Cold Spring Harbor Laboratory Press, 1989). For an illustrative example of isolation of a msbB gene of bacteria, including but not limited to Salmonella spp., see Section 7.1 infra. A bacterial DNA library can be probed with a 32P-labeled msbB gene from E. coli. Hybridizing clones are determined to be correct if they contain DNA sequences similar to the known E. coli msbB gene.
  • 6.1.1 Genetic Alteration of Salmonella msbB [0061]
  • One embodiment of the present invention provides a composition of matter which is a strain of bacteria with a genetic alteration in the msbB gene. In a preferred embodiment, the bacteria is Salmonella sp. Genetic alteration in the form of disruption or deletion can be accomplished by several means known to those skilled in the art, including homologous recombination using an antibiotic resistance marker. These methods involve disruption of the plasmid-based, cloned msbB gene using restriction endonucleases such that part or all of the gene is disrupted or eliminated or such that the normal transcription and translation are interrupted, and an antibiotic resistance marker for phenotypic selection is inserted in the region of that deletion, disruption or other alteration. Linearized DNA is transformed into Salmonella, and bacteria bearing the antibiotic resistance are further examined for evidence of genetic alteration. Means for examining genetic alteration include PCR analysis and Southern blotting. For an illustrative example of genetic disruption of a Salmonella msbB gene, see Section 7.2. [0062]
  • In another embodiment of the invention, the msbB[0063] /antibiotic resistance marker can be transduced into a new bacterial strain. An illustrative example is provided in Section 7.2. Bacteriophage P22 and a Salmonella msbB clone can be grown in zero salt Luria broth and the new phages in the supernate can be used to infect a new Salmonella strain.
  • Yet another embodiment of the present invention provides Salmonella that are attenuated in more than one manner, e.g., a mutation in the pathway for lipid A production, such as the msbB mutation described herein and one or more mutations to auxotrophy for one or more nutrients or metabolites, such as uracil biosynthesis, purine biosynthesis, and arginine biosynthesis as described by Bochner, 1980, J. Bacteriol. 143:926-933 herein incorporated by reference. In a preferred embodiment, the ability of msbB[0064] Salmonella to accumulate within tumors is retained by msbB 0 Salmonella having one or more mutations resulting in an auxotrophic strain. In a more preferred mode of this embodiment of the invention, the bacterial vector which selectively targets tumors and expresses a pro-drug converting enzyme is auxotrophic for uracil, aromatic amino acids, isoleucine and valine and synthesizes an altered lipid A. In a specific preferred embodiment the msbB Salmonella also contain a genetic modification of the biosynthetic pathway gene, purl, leading to decreased virulence of the strain compared to wild type. An illustrative example is provided in Sections 7 and 8.
  • 6.1.2 Characteristics of Salmonella Having Disrupted msbB [0065]
  • A characteristic of the msbB[0066] Salmonella, described herein, is decreased ability to induce a TNFα response compared to the wild type bacterial vector. Both the whole bacteria and isolated or purified lipopolysaccharide (LPS) elicit a TNFα response. In an embodiment of the invention, the msbB Salmonella induce TNFα expression at about 5 percent to about 40 percent compared to the wild type Salmonella sp. (in other words, the msbB Salmonella induce TNFα expression at about 5 percent to about 40 percent of the level induced by wild type Salmonella, e.g., WT 14028.) In a preferred embodiment of the invention, the msbB Salmonella induce TNFα expression at about 10 percent to about 35 percent of that induced by a wild type Salmonella sp. In an embodiment of the invention, purified LPS from msbB Salmonella induces TNFα expression at a level which is less than or equal to 0.001 percent of the level induced by LPS purified from wild type Salmonella sp. TNFα response induced by whole bacteria or isolated or purified LPS can be assessed in vitro or in vivo using commercially available assay systems such as by enzyme linked immunoassay (ELISA). For illustrative examples, see sections 7.3.1 and 7.3.2 infra. Comparison of TNFα production on a per c.f.u. or on a pg/kg basis, is used to determine relative activity. Lower TNFα levels on a per unit basis indicate decreased induction of TNFα production.
  • Reduction of Virulence
  • Another characteristic of the msbB[0067] Salmonella, described herein, is decreased virulence towards the host cancer patient compared to the wild type bacterial vector. Wild type Salmonella can under some circumstances exhibit the ability to cause significant progressive disease. Acute lethality can be determined for normal wild type live Salmonella and live msbB Salmonella using animal models. For an illustrative example, see Section 7.4 and Section 9, Table III. Comparison of animal survival for a fixed inoculum is used to determine relative virulence. Strains having a higher rate of survival have decreased virulence.
  • Decreased Survival Within Macrophages
  • Another characteristic of msbB[0068] Salmonella described herein, is decreased survival within macrophage cells as compared to survival of wild type bacteria. Wild type Salmonella (e.g., ATCC 14028) are noted for their ability to survive within macrophages (Baumler, et al., 1994, Infect. Immun. 62:1623-1630; Buchmeier and Heffron 1989, Infect. Immun. 57:1-7; Buchmeier and Heffron, 1990, Science 248:730-732; Buchmeier et al., 1993, Mol. Microbiol. 7:933-936; Fields et al., 1986, Proc. Natl. Acad. Sci. USA 83:5189-93; Fields et al., 1989, Science 243:1059-62; Fierer et al., 1993, Infect. Immun. 61:5231-5236; Lindgren et al., 1996, Proc. Natal. Acad. Sci. USA 3197-4201; Miller et al., 1989, 30 Proc. Natl. Acad. Sci. USA 86:5054-5058; Sizemore et al., 1997, Infect. Immun. 65:309-312).
  • A comparison of survival time in macrophages can be made using an in vitro cell culture assay. A lower number of c.f.u. over time is indicative of reduced survival within macrophages. For an illustrative example, see [0069] Section 8 infra. As shown therein, using the gentamicin-based internalization assay and bone marrow-derived murine macrophages or the murine macrophage cell line J774, a comparison of survival of WT 14028 and msbB clone YS8211 was determined. In an embodiment of the invention, survival occurs at about 50 percent to about 30 percent; preferably at about 30 percent to about 10 percent; more preferably at about 10 percent to about 1 percent of survival of the wild type stain.
  • Increased Sensitivity
  • Another characteristic of one embodiment of the msbB[0070] Salmonella, described herein, is increased sensitivity of the tumor-targeted bacteria to specific chemical agents which is advantageously useful to assist in the elimination of the bacteria after administration in vivo. Bacteria are susceptible to a wide range of antibiotic classes. However, it has surprisingly been discovered that certain Salmonella msbB mutants encompassed by the present invention are sensitive to certain chemicals which are not normally considered antibacterial agents. In particular, certain msbB 0 Salmonella mutants are more sensitive than WT 14028 to chelating agents.
  • Previous descriptions of msbB[0071] E. coli have not suggested increased sensitivity to such chelating agents. To the contrary, reports have included increased resistance to detergents such as deoxycholate (Karow and Georgopoulos 1992 J. Bacteriol. 174: 702-710).
  • To determine sensitivity to chemical agents, normal wild type bacteria and msbB[0072] bacteria are compared for growth in the presence or absence of a chelating agent, for example, EDTA, EGTA or sodium citrate. Comparison of growth is measured as a function of optical density, i.e., a lower optical density in the msbB strain grown in the presence of an agent, than when the strain is grown in its absence, indicates sensitivity. Furthermore, a lower optical density in the msbB strain grown in the presence of an agent, compared to the msbB+ strain grown in its presence, indicates sensitivity specifically due to the msbB mutation. For an illustrative example, see section 7.7 infra. In an embodiment of the invention, 90 percent inhibition of growth of msbB− Salmonella (compared to growth of wild type Salmonella sp.) occurs at about 0.25 mM EDTA to about 0.5 mM EDTA, preferably at about 99 percent inhibition at about 0.25 mM EDTA to above 0.5 mM EDTA, more preferably at greater than 99 percent inhibition at about 0.25 mM EDTA to about 0.5 mM EDTA. Similar range of growth inhibition is observed at similar concentrations of EGTA.
  • Derivatives of msbB Mutants
  • When grown in Luria Broth (LB) containing zero salt, the msbB[0073] mutants of the present invention are stable, i.e., produce few derivatives (as defined below). Continued growth of the msbB mutants on modified LB (10 g tryptone, 5 g yeast extract, 2 ml 1N CaCl2, and 2 ml 1N MgSO4 per liter, adjusted to pH 7 using 1N NaOH) also maintains stable mutants.
  • In contrast, when grown in normal LB, the msbB[0074] mutants may give rise to derivatives. As used herein, “derivatives” is intended to mean spontaneous variants of the msbB mutants characterized by a different level of virulence, tumor inhibitory activity and/or sensitivity to a chelating agent when compared to the original msbB mutant. The level of virulence, tumor inhibitory activity, and sensitivity to a chelating agent of a derivative may be greater, equivalent, or less compared to the original msbB mutant.
  • Derivatives of msbB[0075] strains grow faster on unmodified LB than the original msbB strains. In addition, derivatives can be recognized by their ability to grow on MacConkey agar (an agar which contains bile salts) and by their resistance to chelating agents, such as EGTA and EDTA. Derivatives can be stably preserved by cryopreservation at −70° C. or lyophilization according to methods well known in the art (Cryz et al., 1990, In New Generation Vaccines, M. M. Levine (ed.), Marcel Dekker, New York pp. 921-932; Adams, 1996, In Methods in Molecular Medicine: Vaccine Protocols, Robinson et al. (eds), Humana Press, New Jersey, pp. 167-185; Griffiths, Id. pp. 269-288.)
  • Virulence is determined by evaluation of the administered dose at which half of the animals die (LD[0076] 50). Comparison of the LD50 of the derivatives can be used to assess the comparative virulence. Decrease in the LD50 of a spontaneous derivative as compared to its msbB parent, indicates an increase in virulence. In an illustrative example, the faster-growing derivatives either exhibit the same level of virulence, a greater level of virulence, or a lower level of virulence compared to their respective original mutant strains (see Section 9, Table III.) In another example, the ability of a derivative to induce TNFα remains the same as the original mutant strain (see Section 7.3, FIG. 7).
  • In an illustrative example, the derivatives can either inhibit tumor growth more than or less than their respective original mutant strains (see Section 7.6, FIG. 11). It is demonstrated in Section 7.6 that the original msbB[0077] mutant, YS8211, significantly inhibits tumor growth whereas a derivative of this clone, YS8212, has less tumor growth inhibition activity. In contrast, the derivative, YS1629, exhibits enhanced tumor growth inhibition activity compared to its parent msbB clone, YS7216.
  • A derivative which is more virulent than its parent mutant but which does induce TNFα at a lower level when compared to the wild type, i.e., at a level of about 5 percent to about 40 percent of that induced by the wild type Salmonella, can be further modified to contain one or more mutations to auxotrophy. In an illustrative example, the YS1170 derivative is mutated such that it is auxotrophic for one or more aromatic amino acids, e.g., aroA, and thus can be made less virulent and is useful according to the methods of the present invention. In an additional illustrative example, genetic modifications of the purl gene (involved in purine biosynthesis) yeild Salmonella strains that are less virulent than the parent strain. (See Sections 7 and 8). [0078]
  • Prior to use of a derivative in the methods of the invention, the derivative is assessed to determine its level of virulence, ability to induce TNFα, ability to inhibit tumor growth, and sensitivity to a chelating agent. [0079]
  • 6.2 Use of Salmonella with Disrupted msbB for Tumor Targeting and in Vivo Treatment of Solid Tumors [0080]
  • According to the present invention, the msbB[0081] mutant Salmonella are advantageously used in methods to produce a tumor growth inhibitory response or a reduction of tumor volume in an animal including a human patient having a solid tumor cancer. For such applications, it is advantageous that the msbB mutant Salmonella possess tumor targeting ability or target preferably to tumor cells/tissues rather than normal cells/tissues. Additionally, it is advantageous that the msbB mutant Salmonella possess the ability to retard or reduce tumor growth and/or deliver a gene or gene product that retards or reduces tumor growth. Tumor targeting ability can be assessed by a variety of methods known to those skilled in the art, including but not limited to cancer animal models.
  • For example, Salmonella with a msbB[0082] modification are assayed to determine if they possess tumor targeting ability using the B16F10 melanoma subcutaneous animal model. A positive ratio of tumor to liver indicates that the genetically modified Salmonella possesses tumor targeting ability. For an illustrative example, see Section 7.5.
  • Salmonella with the msbB[0083] modification can be assayed to determine if they possess anti-tumor ability using any of a number of standard in vivo models, for example, the B16F10 melanoma subcutaneous animal model. By way of an illustrative example, and not by way of limitation, tumors are implanted in the flanks of mice and staged to day 8 and then bacterial strains are injected i.p. Tumor volume is monitored over time. Anti-tumor activity is determined to be present if tumors are smaller in the bacteria-containing groups than in the untreated tumor-containing animals. For an illustrative example, see section 7.6 infra.
  • The Salmonella of the present invention for in vivo treatment are genetically modified such that, when administered to a host, the bacteria is less toxic to the host and easier to eradicate from the host's system. The Salmonella are super-infective, attenuated and specific for a target tumor cell. In a more preferred embodiment, the Salmonella may be sensitive to chelating agents having antibiotic-like activity. [0084]
  • In addition, the Salmonella used in the methods of the invention can encode “suicide genes”, such as pro-drug converting enzymes or other genes, which are expressed and secreted by the Salmonella in or near the target tumor. Table 2 of Pawelek et al. W096/40238 at pages 34-35 presents an illustrative list of pro-drug converting enzymes which are usefully secreted or expressed by msbB[0085] mutant Salmonella for use in the methods of the invention. Table 2 and pages 32-35 are incorporated herein by reference. The gene can be under the control of either constitutive, inducible or cell-type specific promoters. See Pawelek et al. at pages 35-43, incorporated herein by reference, for additional promoters, etc. useful for mutant Salmonella for the methods of the present invention. In a preferred embodiment, a suicide gene is expressed and secreted only when a Salmonella has invaded the cytoplasm of the target tumor cell, thereby limiting the effects due to expression of the suicide gene to the target site of the tumor.
  • In a preferred embodiment, the Salmonella, administered to the host, expresses the HSV TK gene. Upon concurrent expression of the TK gene and administration of ganciclovir to the host, the ganciclovir is phosphorylated in the periplasm of the microorganism which is freely permeable to nucleotide triphosphates. The phosphorylated ganciclovir, a toxic false DNA precursor, readily passes out of the periplasm of the microorganism and into the cytoplasm and nucleus of the host cell where it incorporates into host cell DNA, thereby causing the death of the host cell. [0086]
  • The method of the invention for inhibiting growth or reducing volume of a solid tumor comprises administering to a patient having a solid tumor, an effective amount of an isolated mutant Salmonella sp. comprising a genetically modified msbB gene, said mutant being capable of targeting to the solid tumor when administered in vivo. The msbB[0087] mutant Salmonella may also express a suicide gene as described above.
  • In addition, in one embodiment the isolated Salmonella is analyzed for sensitivity to chelating agents to insure for ease in eradication of the Salmonella from the patient's body after successful treatment or if the patient experiences complications due to the administration of the isolated Salmonella. Thus, if Salmonella is employed which is sensitive to a chelating agent, at about 0.25 mM to about 1.0 mM of a chelating agent such as EGTA, EDTA or sodium citrate can be administered to assist in eradication of the Salmonella after the anti-tumor effects have been achieved. [0088]
  • When administered to a patient, e.g., an animal for veterinary use or to a human for clinical use, the mutant Salmonella can be used alone or may be combined with any physiological carrier such as water, an aqueous solution, normal saline, or other physiologically acceptable excipient. In general, the dosage ranges from about 1.0 c.f.u./kg to about 1×10[0089] 10 c.f.u./kg; optionally from about 1.0 c.f.u./kg to about 1×108 c.f.u./kg; optionally from about 1×102 c.f.u./kg to about 1 x 108 c.f.u./kg; optionally from about 1×104 c.f.u./kg to about 1×108 c.f.u./kg.
  • The mutant Salmonella of the present invention can be administered by a number of routes, including but not limited to: orally, topically, injection including, but limited to intravenously, intraperitoneally, subcutaneously, intramuscularly, intratumorally, i.e., direct injection into the tumor, etc. [0090]
  • The following series of examples are presented by way of illustration and not by way of limitation on the scope of the invention. [0091]
  • 7. EXAMPLE Loss of Virulence, Reduced TNFα Stimulation, and Increased Chelating Agent Sensitivity, by Disruption of the Salmonella msbB
  • 7.1 Isolation and Composition of Salmonella msbB Gene [0092]
  • A Salmonella genomic DNA library was first constructed. Wild type [0093] Salmonella typhimurium (ATCC strain 14028) were grown overnight and genomic DNA extracted according to the methods of Sambrook et al. (Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Press, Cold Spring Harbor, 1989). Size-selected restriction endonuclease-digested fragments ranging from 2 to 10 kB were generated by time-limited digestion with Sau3A and selected by agarose gel electrophoresis. These fragments were ligated into pBluescript SK− and transformed to E. coli DH5α. Random analysis of clones revealed DNA inserts in ≧87%, with average size=5.1 Kb. The library consisted of 1.4×104 independent clones. In order to reduce the hybridization of the E. coli-originated msbb probe, to the 100% homologous chromosomal gene in E. coli, the entire library was harvested from the petri dishes by flooding them with phosphate buffered saline and using a glass rod to dislodge the colonies, and the resulting bacterial population was subjected to a large-scale plasmid isolation, resulting in an amplified Salmonella library plasmid pool. This plasmid pool was then transformed to Salmonella LT2 YS5010, thereby eliminating the E. coli background.
  • A probe for msbB homologues was generated using a clone of the [0094] E. coli msbB gene (Karow and Georgopoulos 1992 J. Bacteriol. 174: 702-710) by digesting E. coli with BglII/HincII and isolating a 600 bp fragment which corresponds to a portion of the coding sequence. This fragment was labeled using α32P-dCTP and used to probe the Salmonella library at low-stringency conditions consisting of 6× SSC, 0.1 % SDS, 2× Denhardts, 0.5 % non-fat dry milk overnight at 55° C. Strongly hybridizing colonies were purified, and plasmids extracted and subjected to restriction digestion and in situ gel hybridization under the same conditions used for colony hybridization (Ehtesham and Hasnain 1991 BioTechniques 11: 718-721). Further restriction digests revealed a 1.5 kB fragment of DNA which strongly hybridized with the probe and was sequenced at the Yale University Boyer Center using fluorescent dye termination thermal cycle sequencing. Sequence analysis revealed that the 1.5 kb fragment contained an msbB homologue which apparently lacked an initiating methionine corresponding to that of the E. coli gene. A probe consisting of the 5′ region of this clone was generated by performing restriction digests using EcoR1/XbaI and again hybridizing to the library. The complete nucleotide sequence of the Salmonella msbb gene (SEQ ID NO:1) and the deduced amino acid sequence of the encoded protein (SEQ ID NO:2) is shown in FIG. 1. The DNA homology of the putative Salmonella msbB and the E. coli msbB is 75%. The protein homology is 98%, confirming that the cloned Salmonella gene is a bona fide msbB.
  • 7.2 Genetic Alteration of Salmonella msbB [0095]
  • A knockout construct was generated using the cloned Salmonella msbB gene. The cloned gene was cut with SphI and MluI, thereby removing approximately half of the msbB coding sequence, and the tetracycline resistance gene from pBR322, cut with AatII and AvaI, was inserted after blunt-ending using the Klenow fragment of DNA polymerase I (FIG. 2A-[0096] 2C). The knockout disruption was accomplished by homologous recombination procedures (Russell et al., 1989, J. Bacteriol. 171:2609); the construct was linearized using SacI and KpnI, gel purified and transfected to Salmonella LT2 YS501 by electroporation. Bacteria from the transformation protocol were first selected on tetracycline plates, and subsequently examined for the presence of plasmid-containing non-chromosomal integrated contaminants by ampicillin resistance and the presence of plasmids as determined by standard plasmid mini-preps (Titus, D. E., ed. Promega Protocols and Applications Guide, Promega Corp, 1991). Bacterial colonies which were tetracycline resistant yet lacked plasmids were subjected to a PCR-based analysis of the structure of their msbB gene. PCR was used with primers which generate a fragment inclusive of the region into which the tetracycline gene was inserted, where the forward primer was GTTGACTGGGAAGGTCTGGAG (SEQ ID NO:3), corresponding to bases 586 to 606, and the reverse primer was CTGACCGCGCTCTATCGCGG (SEQ ID NO:4), corresponding to bases 1465 to 1485. Wild type Salmonella msbB+ results in an approximately 900 base pair product, whereas the disrupted gene with the tetracycline insert results in an approximately 1850 base pair product. Several clones were obtained where only the larger PCR product was produced, indicating that the disruption in the msbB gene had occurred.
  • Southern blot analysis was used to confirm the disruption of the chromosomal copy of Salmonella msbb. The plasmid-based knockout construct (KO) was compared with genomic DNA prepared from wild type and putative disrupted msbB clones, YS82, YS86, YS8211 and YS861. The DNA was double digested with AseI/BamHI and separated by agarose gel electrophoresis on 0.9% or 1.2% agarose. Results of YS8211 and YS861 are presented in FIGS. [0097] 3A-3C. Similar gels were subjected to three separate criteria: 3A) the presence of the tetracycline gene when probed with an 32P-labeled tetracycline gene fragment, 3B) Restriction fragment length when probed with an 32P-labeled AseI/BamH1 fragment derived from the cloned msbB and 3C) the presence or absence of the msbB mluI fragment removed in order to disrupt the msbB gene and insert the tetracycline gene (FIGS. 3A-3C). Since the mluI fragment was removed in order to disrupt the msbB gene and insert the tetracycline gene, it is expected that this probe would hybridize with the wild type FIG. 3C (lane 2 WT) but not the nockout construct (lane 1 KO), or the clones, ( lanes 3 and 4 YS8211 and YS821) thereby confirming the genetic alteration of the msbB gene. Each of the clones examined exhibited all of the expected criteria for an msbB gene deletion (knockout). These data further confirm that msbB exists as a single copy in the wild type Salmonella, as no other hybridizing bands were observed when probed with a labeled oligonucleotide derived from the cloned DNA.
  • After the msbB mutation was confirmed, additional strains containing the msbB[0098] mutation were generated. The Salmonella strains used included WT 14028 and YS72 (pur xyl hyperinvasive mutant from WT 14028; Pawelek et al., WO 96/40238). P22 transduction was used to generate YS8211 (msbB::tet) using YS82 as a donor and YS861 and YS862 (msbB1::tet) using YS86 as a donor; all with WT 14028 as recipient. YS7216 (msbB1::tet from YS72) was generated by transduction using YS82 as a donor. Several derivatives are encompassed by the present invention, including but not limited to derivatives of YS8211 (YS8212, YS1170), YS862 (YS8644, YS8658), and YS7216 (YS1601, YS1604, YS1629). In a preferred embodiment, spontaneous derivatives grow somewhat faster on Luria agar compared to WT 14028 or msbB clones generated by transduction. msbB+ strains were grown in LB broth or on LB plates containing 1.5% agar at 37° C. msbB strains were grown in modified LB containing 10 g tryptone, 5 g yeast extract, 2 ml 1N CaCl2 and 2 ml 1N MgSO4 per liter, adjusted to pH 7 using 1N NaOH. For transducing msbB1::tet, LB lacking NaCl was used, with 4 mg/l tetracycline. Liquid cultures were shaken at 225 rpm. For tumor targeting experiments, cells were diluted 1:100 in LB, grown to OD60032 0.8 to 1.0, washed in phosphate buffered saline (PBS), and resuspended in PBS.
  • 7.2.1 An Improved Method for Selecting msbB Genetic Alterations by Pre-selection with Sucrose [0099]
  • An improved method for selecting msbB genetic alterations by pre-selection with sucrose has been discovered. This pre-selection method is based on the selection of colonies that retain the sacB gene. The sacB gene is responsible for the conversion of sucrose into a toxic chemical, levan, that is lethal to the host cells, and can therefore be used to select for recombinants. Only those strains that undergo deletion of the sacB gene survive on medium containing sucrose and therefore have the sucrose resistance property sucr. As described below, pre-selecting of colonies that retain the sacB gene, eliminated the need for dilutions and comparison of sucrose[0100] (+) vs. sucrose(−) colonies as performed in the normal sucrose selection.
  • The Normal Selection Procedure for the Sucrase System: [0101]
  • [0102] E. coli SM10 λpir carrying a plasmid with the msbB(Δ) bla and sacB genes was used as a donor. The bla gene for betalactamase confers resistance to ampicillin. In the normal selection procedure, the donor strain was mated using standard mating procedures, with a Salmonella strain into which the plasmid with msbB(Δ) bla sacb was to be introduced. Since the Salmonella strain contained a second antibiotic resistance marker (e.g., streptomycin resistance), the recombinant Salmonella clones were then selected for dual resistance to ampicillin and streptomycin. To test for resolution of an individual clone, dilutions of each clone were plated on LB lacking sucrose, or LB containing 5% sucrose. Only those strains that underwent deletion or alteration of the sacb gene survive on sucrose. Comparison of the number of clones on sucrose(+) or sucrose(−) plates, indicates the fraction of bacterial cells that underwent resolution. Sucrose resistant colonies were then further tested for sensitivity to ampicillin and tetracycline. Tets and amps indicated excision of the sacB and bla genes during cross-over with the partial msbB gene region. PCR was then used to confirm the msbB isoform present in the tets amps clones.
  • Pre-Selection Protocol for the Sucrase System: [0103]
  • A variation in the normal sucrase protocol allowed for the screening of increased numbers of colonies, by pre-selecting colonies that retain the sacB gene. This pre-selection method eliminated the need for examination and comparison of sucrose[0104] (+) vs. sucrose(−) from a large number of colonies. After the conjugation procedure described above, the colonies (impure at this stage) were gridded directly to LB plates containing 5% sucrose and grown at 30° C. The resulting impure colonies, which continued to grow, gave rise to survivors on sucrose. Of the sucrose resistant colonies, those which displayed a phenotypic variation of “fuzzy edges” were then subjected to dilution and plated on sucrose (+) or sucrose (−) plates. Colonies were then tested for sensitivity to tetracycline and ampicillin as above, and the msbB isoform was confirmed by PCR. This improved method was used to generate strains for P22 phage transduction of msbB(Δ) bla sacB chromosomal element. These strains were then used to generate the YS1456 and YS1646 stains, which represent preferred embodiments of the novel msbB mutations of the present invention (see FIG. 15 and 16).
  • 7.3 Disruption of Salmonella msbB Reduces TNFα Induction [0105]
  • 7.3.1 TNFα Induction in Mice [0106]
  • [0107] WT 14028 and the msbB clone YS8211, were first grown to saturation in LB media at 37° C. with shaking at 225 rpm. A 1:100 dilution of these bacterial strains were then transferred to fresh LB and grown to an OD600=1.0 at 37° C. with shaking at 225 rpm. The bacteria were diluted in phosphate buffered saline and 1.0×108 c.f.u. (about 5×109 c.f.u./kg) were injected into the tail vein of Balb/C mice (n=4/strain), with PBS as a negative control. After 1.5 hours, serum was harvested in triplicate samples by cardiac puncture, centrifuged to remove the cellular content, and analyzed for TNFα using a Biosource International Cytoscreen ELISA plate, which was read on a Molecular Devices Emax microplate reader.
  • Results are presented in FIG. 4 and expressed as a percent of the level of TNFα induced by wild type Salmonella. [0108]
  • As demonstrated in FIG. 4, YS8211 induced TNFα significantly less than [0109] WT 14028. Thus, as shown in FIG. 4, the msbB strain induced TNFα about 33% (i.e., 3 times less) of the wild type msbB+ strain.
  • 7.3.2 TNFα Induction in Pigs [0110]
  • An msbB[0111] strain of Salmonella, YS8212, and WT 14028, were first grown to saturation in LB media at 37° C. with shaking at 225 rpm. A 1:100 dilution of these bacterial strains were then transferred to fresh LB and grown to an OD600=0.8 at 37° C. with 225 rpm. The bacteria were washed in phosphate buffered saline and 1.0×109 c.f.u. (about 1×108 c.f.u./kg) were injected into the ear vein of Sinclair swine (n=6/strain). After 1.5 and 6.0 hours, serum was harvested, centrifuged to remove the cellular content, and frozen for later analysis. Analysis for TNFα utilized a Genzyme Predicta ELISA plate, which was read using a Gilson spectrophotometer.
  • Results are presented in FIG. 5 and are expressed as picograms of TNFα/ml serum. [0112]
  • As demonstrated in FIG. 5, at 90 minutes the level of TNFα induced by the msbB[0113] strain was significantly lower than that induced by the Salmonella WT 14028.
  • [0114] 7.3.3 Salmonella LPS-induced Respiration in Pigs
  • Lipopolysaccharide (LPS) from [0115] Salmonella WT 14028 and the msbB clone, YS8212 was prepared using the procedure described by Galanos et al. (1969 Eur. J. Biochem. 9: 245-249). Briefly, LPS was extracted from bacteria which had been grown to OD600 of 1.0. The bacteria were pelleted by centrifugation, washed twice with distilled water and frozen at −20 C. LPS was purified by extraction with a mixture of 18.3 ml H20:15 ml phenol in a shaking water bath for 1 hr at 70 C. The mixture was cooled on ice, centrifuged at 20,000× g for 15 min, and the aqueous phase was removed. LPS was precipitated from the aqueous phase by addition of NaCl to 0.05 M and 2 volumes ethanol and incubation on ice, followed by centrifugation of 2000× g for 10 min. The precipitation was repeated after redissolving the pellet in 0.05 M NaCl, and the pellet lyophilized. The LPS was dissolved in sterile distilled water, and either 5 μg/kg or 500 μg/kg LPS was injected into the ear vein of Sinclair swine which had been anesthetized with Isoflurane. After 1.5 and 6.0 hours, respiration rate was determined and recorded.
  • Results are presented in FIG. 6 and are expressed as a percentage of respiration at time zero (t[0116] o).
  • As demonstrated in FIG. 6, respiration was significantly higher in the pigs administered wild type LPS as compared to those administered the LPS from the msbB[0117] strain. Thus, disruption of the msbb gene in Salmonella, produces a modification in lipid A which results in reduced ability to increase respiration.
  • 7.3.4 TNFα Induction in Human Monocytes [0118]
  • Human monocytes were prepared from peripheral blood by centrifugation through Isolymph (Pharmacia) and allowed to adhere to 24 well plates containing RPMI 1640. [0119] Salmonella WT 14028 and several of the msbB 14028 strains (YS8211, YS8212, YS8658, and YS1170) were first grown to saturation in LB media at 37° C. with shaking at 225 rpm. A 1:100 dilution of these bacterial strains was then transferred to fresh LB and grown to an OD600=0.8 at 37° C. with 225 rpm. The bacteria were added to the cell culture wells and the culture medium was harvested after 2.0 hours, centrifuged to remove the cellular content, and analyzed for TNFα using a Genzyme redicta ELISA plate, which was read using a Gilson spectrophotometer.
  • The data are presented in FIG. 7 and expressed as picograms of TNFα/ml serum. [0120]
  • As demonstrated in FIG. 7, the msbB[0121] strains induced TNFα significantly less than did the wild type strain.
  • 7.3.5 msbB[0122] Salmonella LPS TNFα Induction in Human Monocytes
  • Human monocytes were prepared from peripheral blood by centrifugation through Isolymph (Pharmacia) and allowed to adhere to 24 well plates containing RPMI 1640. Lipopolysaccharide (LPS) of wild type and of a number of msbB[0123] mutant Salmonella, (i.e., YS8211, YS8212, YS8658 and YS1170) was prepared using the procedure described by Galanos et al. (1969 Eur. J. Biochem. 9: 245-249) (see Section 7.3.3 for a brief description). The LPS was dissolved in sterile distilled water, and quantities ranging from 0.001 to 100 ng/ml LPS were added to the cell culture wells. After 15 hours the culture medium was harvested, centrifuged to remove the cellular content, and analyzed for TNFα using a Genzyme Predicta ELISA plate, which was read using a Gilson spectrophotometer.
  • The data are presented in FIG. 8 and are expressed as picograms of TNFa/ml serum. [0124]
  • As demonstrated in FIG. 8, LPS purified from the msbB[0125] strains induced TNFα significantly less than did the LPS from the wild type strain.
  • 7.4 Disruption of Salmonella msbB Reduces Virulence [0126]
  • 7.4.1 In Mice [0127]
  • A culture of [0128] wild type Salmonella 14028 and one of its msbB Salmonella clones, YS862, were grown in LB medium lacking sodium chloride at 37° C. with shaking at 250 rpm until the cultures reached an OD600 of 0.8. The bacteria were diluted into phosphate buffered saline (PBS) at a ratio of 1:10 and the equivalent of 2×107 c.f.u. were injected i.p. into C57BL/6 mice bearing B16F10 melanomas. Survival was determined daily, or at two to four day intervals.
  • Results are presented in FIG. 9A and are expressed as percent survival. [0129]
  • As shown in FIG. 9A, [0130] WT 14028 killed all the mice in 4 days, whereas the msbB mutant spared 90% of the mice past 20 days, demonstrating a significant reduction in virulence by the msbB mutant.
  • 7.4.2 In Pigs [0131]
  • A culture of [0132] WT 14028 and one of its msbB Salmonella clones, YS8212, were grown in LB medium lacking sodium chloride at 37° C. with shaking of 250 RPM until the cultures reached an OD600 of 0.8. The bacteria were washed in phosphate buffered saline and 1.0×109 were injected into the ear vein of Sinclair swine (n=4/strain). Survival was determined daily, or at two to four day intervals.
  • Results are presented in FIG. 9B and are expressed as percent survival. [0133]
  • As shown in FIG. 9B, [0134] WT 14028 killed all the swine in 3 days, whereas the msbB mutant spared 100% of the mice past 20 days, demonstrating a significant reduction in virulence.
  • 7.5 Tumor Targeting of msbB[0135] Clones
  • [0136] Salmonella WT 14028 with the msbB modification, were assayed to determine if they possessed tumor targeting ability using the B16F10 melanoma subcutaneous animal model. The msbB clone, YS8211, was grown in LB media lacking sodium chloride at 37° C. with shaking at 250 rpm to an OD600 of 0.8. An aliquot of 2.0×106 c.f.u. was injected i.v. into C57BL/6 mice which had been implanted with 2×105 B16 melanoma cells 16 days prior to the bacterial infection. At two days and five days post bacterial infection, mice were sacrificed and tumors and livers assayed for the presence of the bacteria by homogenization and plating of serial dilutions.
  • Results are presented in FIG. 10 and are expressed as c.f.u. bacteria/g tissue. As demonstrated in FIG. 10, a positive ratio of tumor to liver (700:1) was found at 2 days, and increased to a positive ratio of 2000:1 at 5 days. Thus, the msbB[0137] mutant maintained the ability to target to a solid cancer tumor.
  • 7.6 Use of Salmonella with Disrupted msbB for Anti-tumor Activity in Vivo [0138]
  • [0139] Salmonella typhimurium 14028 msbB clones YS8211, YS8212, YS7216, and YS1629 and WT 14028 (control) were grown in LB media lacking sodium chloride at 37° C. with shaking at 250 rpm to an OD600 of 0.8. An aliquot of 2.0×106 c.f.u. was injected i.p. into C57BL/6 mice which had been implanted with 2×105 B16 melanoma cells 8 days prior to the bacterial infection. Tumor volume was monitored over time.
  • Results are presented in FIG. 11. Two of the strains, YS8211 and YS1629, showed significant tumor retardation, i.e., tumor growth inhibition. [0140]
  • 7.7 Increased Sensitivity to Chelating Agents [0141]
  • In order to assess the sensitivity of bacterial strains to chelating agents, bacteria with or without the msbB mutation were grown in the presence or absence of 1 mM EDTA or 10 mM sodium citrate in Luria Broth (LB) lacking sodium chloride. An overnight culture of each of the bacterial strains was diluted 1 to 100 in fresh media, and grown at 37° C. with shaking at 250 rpm. The effect on growth was determined by spectrophotometric readings at an OD[0142] 600.
  • [0143] WT 14028 and msbB clone YS8211 were grown in the presence or absence of 1 mM EDTA (FIG. 12A). EDTA did not inhibit the growth of WT 14028. In contrast, the msbB clone showed near complete cessation of growth after 3 hours in the presence of EDTA.
  • [0144] WT 14028 and msbB clone YS862 were grown in the presence and absence of 10 mM sodium citrate (FIG. 12B). The msbB+ WT 14028 strain showed little inhibition by sodium citrate compared to the msbB strain which showed near complete cessation of growth after 3 hours in the presence of sodium citrate.
  • Thus, the msbB[0145] Salmonella mutants exhibited sensitivity to chelating agents which promote eradication of the bacteria, a characteristic which is similar to an antibiotic effect. It is envisioned that such a characteristic would be advantageous for use of msbB Salmonella mutants for in vivo therapy.
  • In order to further assess the sensitivity of Salmonella strains to chelating agents, the hyperinvasive pur strain YS72, its msbB[0146] strain, YS7216, and a derivative of YS7216, YS1629, were grown in the presence of increasing concentrations of EDTA. A fresh culture of YS72, its msbB strain YS7216 and its faster-growing derivative YS1629 were diluted 1 to 100 in fresh, zero salt LB media containing 0, 0.25, 0.5, 1.0 or 2.0 mM EDTA and grown at 37° C. with 225 RPM for 4 hours, and c.f.u. was determined by plating serial dilutions onto LB plates (Table I). Greater than 99% inhibition was achieved for the msbB strain YS7216 at concentrations of EDTA greater than 0.25 mM and its derivative YS1629 was inhibited greater than 90% at 0.5 mM and greater than 99% at 2.0 mM. In contrast, although the YS72 clone exhibited some sensitivity to EDTA it was not inhibited at the 90% level even at 2.0 mM.
    TABLE I
    c.f.u. + EDTA {% inhibition}
    Strain c.f.u. no EDTA [0.25 mM] [0.5 mM] [1.0 mM] [2.0 mM]
    YS72 3.0 × 109 2.4 × 109 1.5 × 109 7.3 × 108 4.8 × 108
    {20%} {50%} {75%} {84%}  
    YS7216 6.3 × 108 2.1 × 106 1.1 × 106 3.2 × 106 4.3 × 106
      {99.6%}   {99.8%}   {99.4%} {99.3%}
    YS1629 1.3 × 109 6.0 × 108 1.0 × 108 2.9 × 107 7.5 × 106
    {54%} {92%} {97%} {99.4%}
  • 7.8 Bacterial Survival Within Macrophages [0147]
  • In order to determine the sensitivity of msbB[0148] Salmonella to macrophages, two types of macrophages were used: (A) bone marrow-derived macrophages obtained from the femurs and tibias of C57BL/6 mice, which were allowed to replicate by addition of supernatant from the LADMAC cell line which secretes macrophage colony stimulating factor (Sklar et al., 1985. J. Cell Physiol. 125:403-412) and (B) J774 cells (a murine macrophage cell line) obtained from America Type Culture Collection (ATCC). Salmonella strains used were WT 14028 and its msbB derivatives YS8211 and YS1170. Bacteria were grown to late log phase OD600=0.8 and 1×106 were allowed to infect a confluent layer of mammalian cells within a 24 well dish for 30 min, after which the extracellular bacteria were removed by washing with culture medium and the addition of 50 μg/ml gentamicin (Elsinghorst, 1994, Methods Enzymol. 236:405-420). Bacteria were counted by plating serial dilutions of the cell layer removed using 0.01% deoxycholate, and expressed as the percent initial c.f.u. over time.
  • The results are presented in FIG. 13 and expressed as percent c.f.u. per time. The msbB[0149] strain shows significantly less survival in macrophages.
  • 7.9 LD50 OF msbB Derivatives [0150]
  • Spontaneous derivatives of msbB[0151] strains YS8211 and YS7216 were selected from in vitro culture on non-modified LB medium based upon enhanced growth characteristics. These bacterial strains were grown to OD600 of 0.8 and c.f.u. ranging from 1×102 to 1×108 were injected i.v. into the tail vein of C57BL/6 mice. Acute lethality was determined at 3 days, and the LD50 determined as described by Welkos and O'Brien (Methods in Enzymology 235:29-39, 1994). The results are presented in Table II. Thus, although all the msbB strains have a reduced ability to induce TNFα (See Section 7.3.5), the results demonstrate that strain YS1170 is significantly less attenuated than other msbB strains and therefore not all msbB strains are useful for providing both reduced TNFα induction and reduced virulence.
    TABLE II
    Strain LD50
    WT 14028 1 × 103
    YS8211 4 × 106
    YS8212 3.9 × 107  
    YS1629 1 × 107
    YS1170 1 × 106
  • 8. msbB MUTATION IN COMBINATION WITH A BIOSYNTHETIC PATHWAY MUTATION
  • In order to assess compatibility with auxotrophic mutations, as measured by retention of the ability to target and replicate within tumors, combinations of the msbB mutation with auxotrophic mutations were generated. msbB[0152] + strains were grown in LB broth or LB plates containing 1.5% agar at 37° C. msbB strains were grown in modified LB containing 10 g tryptone, 5 g yeast extract, 2 ml 1N CaCl2 and 2 ml 1N MgSO4 per liter, adjusted to pH 7 using 1N NaOH. For transducing msbB1::tet, LB lacking NaCl was used, with 4 mg/l tetracycline. Liquid cultures were shaken at 225 rpm. The msbbl::tet was transduced to auxotrophic strains to generate YS1604 (msbB, pur, hyperinvasive), YS7232 (msbB, purI, hyperinvasive), YS7244 (msbB, purI, aroA hyperinvasive), YS1482 (msbB, purI, purA). For tumor targeting experiments, cells were diluted 1:100 into LB, grown to OD600=0.8 to 1.0, washed in phosphate buffered saline (PBS), resuspended in PBS, and 2×106 were injected into the tail vein of C57BL/6 mice. At day 7, tumors were excised, weighed, homogenized, and c.f.u. determined by plating serial dilutions onto modified LB described above.
  • Results are presented in Table III and are expressed as c.f.u. per gram tumor tissue. Some of the strains, YS8211, YS1604, and YS7232 show high levels of c.f.u. within the tumors, whereas YS7244 and YS1482 are approximately 500 to 5000 times less. [0153]
    TABLE III
    Strain genetic marker c.f.u./gram tumor tissue
    YS8211 msbB
    3 × 109
    YS1604 msbB, pur, hyperinvasive 9 × 109
    YS7232 msbB, purI, hyperinvasive 9 × 109
    YS7244 msbB, purI, aroAhyperinvasive 5 × 105
    YS1482 msbB, purI, purA 6 × 106
  • 8.1 Generation of the YS1456 Strain Containing Deletions in msbB AND purI [0154]
  • The generation of Salmonella strain YS1456 from the wild type Salmonella typhimurium is outlined in FIG. 15. The wild type Salmonella typhimurium was transduced with purl 1757::Tn10 which conferred tetracycline-resistance, resulting in strain YS1451. [0155]
  • Strain YS1451 was then subjected to a Bochner selection to render the strain tet sensitive and introduce tets gene and introduce a purl deletion (Bochner et al. 1980, J. Bacteriol. 143:926-933), yielding the strain YS1452. Strain YS1452 was tet[0156] s and purI. Strain 1452 was then transduced with msbB1::tet via bacteriophage P22, using strain YS8211 (msbB::tet) as the donor. The resulting strain, YS1453, was initially sensitive to 10 mM ethylene glycol bis((b-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA), spontaneously reverted to a EGTA-resistant phenotype. One such revertant, denoted YS1454, was selected by plating YS1453 on EGTA (2mM in Luria agar).
  • Strain YS1454 was then transduced with the msbB2(Δ) bla sacB chromosomal element, selecting for ampicillin resistance. This transduction process brought in a second version of the disrupted msbB gene, denoted msbB2(Δ) as well as the bla and sacB genes. The bla gene is responsible for the transcription of the enzyme β-lactamase, which metabolizes ampicillin, and was used to select for ampicillin resistant transductants. The sacB gene is responsible for the conversion of sucrose into a toxic chemical, levan, that is lethal to the host cells, and was subsequently used to select for recombinants which lose or have mutations in sacb (see Section 7.2.1 for improved pre-selection methods with sucrose). The presence of the bla and sacB genes allowed the selection of the amp[0157] r and sucs strain (denoted as strain YS1455), which contained both the msbB1::tet and msbB2(Δ) genes.
  • Strain YS1455 was then plated on Luria Bertani (LB) sucrose to select a suc[0158] r amps tets derivative to remove msbB1::tet and restore antibiotic sensitivity. The derivative was denoted as strain YS1456. In summary YS1456 has deletion mutations in purl and msbB. It is also tets amps and EGTAr.
  • 8.2 Generation of the YS1646 Strain Containing Deletions in msbB and purI [0159]
  • The generation of Salmonella strain YS1646 from the wild type [0160] Salmonella typhimurium (wild type strain ATCC 14028) is outlined in FIG. 16. The wild type Salmonella typhimurium was mutagenized with nitrosoguanidine and ultraviolet (UV) light and selected for hyperinvasiveness in melanoma cells. The resistant strain, denoted YS72, were confirmed to possess tumor-hyperinvasiveness pur and xyl 0 properties (Pawelek et al., 1997, Caner Res 57: 4537-4544).
  • To replace the chromosomal purl gene in strain YS72 with a purl deletion, strain YS72 was transduced with the purl 1757::Tn10 gene, which conferred tetracycline-resistance. The donor for the purI 1757::Tn10 gene was Salmonella strain TT11 (purl 1757::Tn10). The donor strain was originally obtained from the Salmonella Genetic Stock Center (Dept. of Biological Science, Univ. Calgary, Calgary, Alberta, Canada T2N 1N4). Transduction was performed using bacteriophage P22 (mutant HT105/1 int-201). The transductant, denoted YS1641, was isolated following selection on tetracycline. [0161]
  • Strain YS1641 was then subjected to a Bochner selection to remove the tet gene and introduce a purI gene deletion (Bochner et al., 1980, J. Bacteriol. 143:926-933), yielding strain YS1642. Strain YS1642 was tet[0162] s and purI. The selection of a tet-deleted strain allowed further genetic modification (e.g., msbB gene disruption, see next paragraph) using tet gene transduction. Strain YS1642 has a tight purine requirement due to purI(Δ), and has been shown to revert to purI+ at a frequency of less than 1 in 1010 cells.
  • Strain YS1642 was then transduced with msbB1::tet via bacteriophage P22, using strain YS8211 (msbB::tet) as the donor. The DNA sequence for the msbB gene is shown in FIG. 1. The tet gene in the msbB1::tet gene confers resistance to 5 mg/L of tetracycline. The resulting strain thus obtained was YS1643. [0163]
  • Strain YS1643 was initially sensitive to 10 mM ethylene glycol bis((b-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA), spontaneously reverted to a EGTA-resistant phenotype. One such revertant, denoted YS1644, was selected by plating YS1643 on EGTA (2 mM in Luria agar). [0164]
  • Strain YS1644 was then transduced with the msbB2(A) bla sacB chromosomal element. This transduction process brought in a second version of the disrupted msbB gene, denoted as msbB2(Δ) as well as the bla and sacB genes. The bla gene is responsible for the transcription of the enzyme β-lactamase, which metabolizes ampicillin, and was subsequently used to select transductants. The sacB gene is responsible for the conversion of sucrose into a toxic chemical, levan, that is lethal to the host cells, and was used to select for recombinants. The presence of the bla and sacB genes allowed the selection of the amp[0165] r and sucs strain (denoted as strain YS1645), which contained both the msbB1::tet and msbB2(A) genes.
  • Strain YS1645 was plated on Luria-Bertani (LB) sucrose to select a suc[0166] r amps tets derivative to remove the msbB::tet gene and restore antibiotic sensitivity (i.e., a derivative with deletion of msbb1::tet bla sacb). This derivative was denoted as strain YS1646.
  • In summary YS1646 has deletion mutations in purI, and msbB. It is also tet[0167] s, amps, and EGTAr.
  • 8.3 Inhibition of Tumor Growth with YS1646 Strain [0168]
  • Intravenous (IV) administration of YS1646, an attentuated strain of [0169] Salmonella typhimurium, resulted in selective replication within tumors, and concomitant inhibition of tumor growth (see FIG. 17 and Table IV).
  • In all instances, a staged tumor model was used in which tumors were allowed to become established following tumor cell inoculation and prior to YS1646 administration. As a result of the ability of YS1646 to replicate within the tumor, a shallow dose-response relationship over the effective dose range was determined whereby the extent of tumor inhibition, exerted by low doses of YS1646, approached the level of tumor inhibition achieved at higher doses. This suggested that, even at low doses, significant clinical efficacy could be achieved as long as the bacteria reached the tumor and accumulated within the tumor. Doses below 1×10[0170] 2 cfu/mouse gave inconsistent results, possibly due to competition between the ability of YS1646 to reach and colonize the tumor vs. the ability of the animals to clear YS1646.
  • The efficacy of YS1646 was evaluated in mice previously implanted with B16-F10 melanoma. In this study a single IV dose of YS1646 at 10[0171] 4, 105 or 106 cfu/mouse significantly reduced tumor size when compared to control treatment, and the degree of tumor size reduction was dose-related. The efficacy observed with the highest dose of YS1646 was superior to that with the positive control, CYTOXAN™ (also known as cyclophosamide), whereas the efficacy with the mid-dose of YS1646 was equivalent to that with, CYTOXAN™. it is important to note that the efficacy induced by YS1646 was induced by a single IV dose, whereas that induced by CYTOXAN™ was multiple IV doses (given weekly, for 3 weeks). The ability of YS1646 to inhibit tumor growth, as a function of dose, was examined over an administered dose range of 1×104 to 1×106 cfu/mouse. Each dosage group was comprised of 10 tumor-bearing animals, which were randomized prior to bacteria administration. Mice were administered bacteria on Day 7, and tumor volumes were measured on Days 10, 13, 17, 20, and 24. For comparison, CYTOXAN™ (cyclophosphamide) was administered once per week at a dose of 200 mg/kg, beginning on Day 7 as well. Mean tumor volumes of each group on Day 24 are presented in Table IV.
    TABLE IV
    Mean Tumor
    Inoculum Dose Volume (mm3) ± Percent
    (cfu/mouse) S.D. T/C Inhibition
     0 4728 ± 804  0
    104 1011 ± 375 0.214 78
    105  560 ± 176 0.118 88
    106 279 ± 91 0.059 94
  • The differences observed between individual groups were deemed significant when analyzed either by the Wilcoxon signed rank test analysis, or by a two-tailed t-test. As indicated in Table IV, increasing tumor inhibition was observed with increasing dose of YS1646. All doses were found to give significant antitumor activity (T/C of less than an equal to 42%), as defined by the Drug Evaluation Branch of the Division of Cancer Treatment, National Cancer Institute (Bethesda, Md.) (Vendetti, J. M., Preclinical drug evaluation: rationale and methods, Semin. Oncol. 8:349-361; 1981), and doses of 1×10[0172] 5 cfu/mouse gave results equivalent to or better than cyclophosphamide. A linear correlation between YS1646 dose and tumor inhibition was not observed due to the ability of YS1646 to replicate preferentially within the tumor, which led to greater than expected potency at lower doses. Intravenous adminstration of YS1646, an attentuated strain of Salmonella typhimurium, resulted in selective replication within tumors, and concomitant inhibition of tumor growth. Between inoculum doses of 1×104 to 1×106 cfu/mouse, a dose-response for inhibition of tumor growth was obtained, ranging from 78% to 94% inhibition of tumor growth. At the two highest inoculum doses, the level of tumor growth inhibition was comparable to or better than that achieved by optimal treatment with cyclophosphamide.
  • 8.4 Virulence [0173]
  • At a dose of 1×10[0174] 6 cfu/mouse, YS1646 does not cause lethality, in contrast to the parental wide type strain ATCC 14028, which causes 100% mortality at a dose of 1×102 cfu/mouse. This indicates that YS1646 is greater than 10,000-fold less virulent than the parental wild type strain. The antitumor efficacy was observed at doses of 104 to 106 cfu/mouse, whereas lethality was not observed until the doses were >106 cfu/mouse. The dose inducing mortality was 1 to 100-fold greater than the dose inducing anti-tumor efficacy (see FIG. 18).
  • 8.5 Antibiotic Suppression of YS1646 Induced Mortality Following Lethal Infection [0175]
  • The ability of ampicillin and ciprofoxacin to suppress infection by YS1646 was evaluated by determining the ability of antibiotics to prevent mortality in C57BL/6 mice inoculated with 5×10[0176] 6 cfu (LD50 equivalent).
  • Groups were divided into the following treatment categories: 1) untreated control, 2) ampicillin-treated, 3) ciprofloxacin-treated, and 4) ciprofloxacin and ampicillin treated. Antibiotic treatment was initiated 3 days following bacteria administration and animals were observed daily for appearance and mortality for 14 days. Results presented herein demonstrate that use of antibiotic was able to supress mortality following lethal bacterial infections (see FIG. 18). [0177]
  • 9. DEPOSIT OF MICROORGANISMS
  • The following microorganisms were deposited with the American Type Culture Collection (ATCC), 10801 University Blvd., Manassas, VA 20110-2209, on Sep. 9, 1997, and have been assigned the indicated Accession numbers: [0178]
    Microorganism ATCC Accession No.
    YS8211 202026
    YS1629 202025
    YS1170 202024
  • The following microorganisms were deposited with the American Type Culture Collection (ATCC), 10801 University Blvd., Manassas, Va. 20110-2209, on Aug. 25, 1998, and have been assigned the indicated Accession numbers: [0179]
    Microorganism ATCC Accession No.
    YS1646 202165
    YS1456 202164
  • The invention claimed and described herein is not to be limited in scope by the specific embodiments, including but not limited to the deposited microorganism embodiments, herein disclosed since these embodiments are intended as illustrations of several aspects of the invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. [0180]
  • A number of references are cited herein, the entire disclosures of which are incorporated herein, in their entirety, by reference. [0181]
  • 1 4 1 2019 DNA SALMONELLA CDS (244)..(1212) 1 gatcaaccag caagccgtta accctctgac agcaaaattg ccgcgcacgg aaggtctgac 60 ggggtcagat cgtcgtgaat acctggcaca ggtgaaagag gttctgccgc aactgcgctt 120 cgattaacaa atgcgctgac agagccggta cgcgatgtgt gccggctttt ttgttttgtg 180 tgagacgcag acgtcgctac actattcaca attccttttc gcgtcagcag accctggaaa 240 agc atg gaa acc aaa aaa aat aat agt gag tat atc cct gaa ttc gaa 288 Met Glu Thr Lys Lys Asn Asn Ser Glu Tyr Ile Pro Glu Phe Glu 1 5 10 15 aaa tcc ttt cgc tat cca cag tat tgg ggc gcc tgg ttg ggc gcg gcg 336 Lys Ser Phe Arg Tyr Pro Gln Tyr Trp Gly Ala Trp Leu Gly Ala Ala 20 25 30 gca atg gcg ggg atc gca tta aca ccg gca tca ttc cgc gac cct ttg 384 Ala Met Ala Gly Ile Ala Leu Thr Pro Ala Ser Phe Arg Asp Pro Leu 35 40 45 ctg gcg acg ctg ggg cgt ttt gcc gga cgg ctg ggg aag agt tct cgt 432 Leu Ala Thr Leu Gly Arg Phe Ala Gly Arg Leu Gly Lys Ser Ser Arg 50 55 60 cgc cgg gcg cta att aat ctg tcg ttg tgc ttt ccg cag cgt agc gaa 480 Arg Arg Ala Leu Ile Asn Leu Ser Leu Cys Phe Pro Gln Arg Ser Glu 65 70 75 gct gag cgc gaa gcg att gtc gat gag atg ttc gcc acc gcg cca cag 528 Ala Glu Arg Glu Ala Ile Val Asp Glu Met Phe Ala Thr Ala Pro Gln 80 85 90 95 gca atg gcg atg atg gct gag ttg gcg atg cgc ggt ccg aaa aaa att 576 Ala Met Ala Met Met Ala Glu Leu Ala Met Arg Gly Pro Lys Lys Ile 100 105 110 caa cag cgt gtt gac tgg gaa ggt ctg gag att atc gag gag atg cgt 624 Gln Gln Arg Val Asp Trp Glu Gly Leu Glu Ile Ile Glu Glu Met Arg 115 120 125 cgt aac gac gaa aaa gtc att ttt ctc gta ccg cat ggc tgg ggc gtc 672 Arg Asn Asp Glu Lys Val Ile Phe Leu Val Pro His Gly Trp Gly Val 130 135 140 gac att cca gcc atg ctg atg gcc tct cag ggg caa aaa atg gcg gcg 720 Asp Ile Pro Ala Met Leu Met Ala Ser Gln Gly Gln Lys Met Ala Ala 145 150 155 atg ttt cat aat cag ggt aat ccg gtt ttt gac tat atc tgg aac aca 768 Met Phe His Asn Gln Gly Asn Pro Val Phe Asp Tyr Ile Trp Asn Thr 160 165 170 175 gtg cgt cgg cgt ttc ggc gga cgt ttg cat gcg cgt aat gac ggg att 816 Val Arg Arg Arg Phe Gly Gly Arg Leu His Ala Arg Asn Asp Gly Ile 180 185 190 aaa ccc ttt att cag tct gtt cgt cag ggc tac tgg ggt tac tac ctg 864 Lys Pro Phe Ile Gln Ser Val Arg Gln Gly Tyr Trp Gly Tyr Tyr Leu 195 200 205 ccg gac cag gat cac ggc ccg gag cat agt gaa ttc gtt gat ttc ttt 912 Pro Asp Gln Asp His Gly Pro Glu His Ser Glu Phe Val Asp Phe Phe 210 215 220 gcg aca tac aaa gcg acg ctg cct gca att ggt cgg ctg atg aaa gtg 960 Ala Thr Tyr Lys Ala Thr Leu Pro Ala Ile Gly Arg Leu Met Lys Val 225 230 235 tgc cgc gca cgc gtg ata ccg ctt ttc ccg gtg tat aat ggt aaa acg 1008 Cys Arg Ala Arg Val Ile Pro Leu Phe Pro Val Tyr Asn Gly Lys Thr 240 245 250 255 cat cgc ctg act atc cag att cgc ccg cca atg gac gat ctg ctc acg 1056 His Arg Leu Thr Ile Gln Ile Arg Pro Pro Met Asp Asp Leu Leu Thr 260 265 270 gct gac gac cac act atc gcc aga cgg atg aac gaa gag gtc gaa att 1104 Ala Asp Asp His Thr Ile Ala Arg Arg Met Asn Glu Glu Val Glu Ile 275 280 285 ttt gtc ggc ccg cat ccg gaa cag tac acc tgg atc ctg aag ctg ctc 1152 Phe Val Gly Pro His Pro Glu Gln Tyr Thr Trp Ile Leu Lys Leu Leu 290 295 300 aaa acc cgc aag cca ggc gag att cag ccg tat aag cgt aaa gat ctt 1200 Lys Thr Arg Lys Pro Gly Glu Ile Gln Pro Tyr Lys Arg Lys Asp Leu 305 310 315 tat ccc atc aaa taaataaagc ctctcgtaag agaggcttta tgctgacaaa 1252 Tyr Pro Ile Lys 320 ccctgtacta cctgatgaac aggcgtgggg gagttttact caacggtcaa aatacgcgtg 1312 gtattggttg aaccgacggt gctcatgaca tcgccctggg tcacgataac caggtcgccg 1372 gaaaccagat accctttatc gcgcagcaga ttaacagctt catgtgccgc gacaacgcca 1432 tcagccgcgc tatcaaaatg caccggcgtt actccgcgat agagcgcggt caggttcagc 1492 gtgcgttcat ggcgcgacat ggcgaaaatc ggcaggccgg agctgatacg ggaagtcatt 1552 agcgcggtac gaccggattc cgtcatggtg atgatcgcgg taacgccttt cagatggttt 1612 gccgcataca ctgcagacat ggcaatggct tcttcaacgt tgtcgaactg cacgtcgaga 1672 cggtgtttag acacattgat gctggggatt ttttctgcgc ccaggcacac gcgcgccatt 1732 gcggcaacgg tttcagaagg atactgaccg gctgcggttt cggcagacag cataaccgca 1792 tccgtgccat ccaggacggc gttcgccacg tccatcactt ccgcacgggt cggcatcggg 1852 ttggtgatca tcgactccat catttgcgtt gcggtgatga ctgcgcggtt tagctgacgc 1912 gcacggcgaa tcagcgcttt ctggatacca accagctccg gatcgccgat ttcaacgccc 1972 agatcgccac gtgcgaccat cacaacgtca gaggccagaa tgatatc 2019 2 323 PRT SALMONELLA 2 Met Glu Thr Lys Lys Asn Asn Ser Glu Tyr Ile Pro Glu Phe Glu Lys 1 5 10 15 Ser Phe Arg Tyr Pro Gln Tyr Trp Gly Ala Trp Leu Gly Ala Ala Ala 20 25 30 Met Ala Gly Ile Ala Leu Thr Pro Ala Ser Phe Arg Asp Pro Leu Leu 35 40 45 Ala Thr Leu Gly Arg Phe Ala Gly Arg Leu Gly Lys Ser Ser Arg Arg 50 55 60 Arg Ala Leu Ile Asn Leu Ser Leu Cys Phe Pro Gln Arg Ser Glu Ala 65 70 75 80 Glu Arg Glu Ala Ile Val Asp Glu Met Phe Ala Thr Ala Pro Gln Ala 85 90 95 Met Ala Met Met Ala Glu Leu Ala Met Arg Gly Pro Lys Lys Ile Gln 100 105 110 Gln Arg Val Asp Trp Glu Gly Leu Glu Ile Ile Glu Glu Met Arg Arg 115 120 125 Asn Asp Glu Lys Val Ile Phe Leu Val Pro His Gly Trp Gly Val Asp 130 135 140 Ile Pro Ala Met Leu Met Ala Ser Gln Gly Gln Lys Met Ala Ala Met 145 150 155 160 Phe His Asn Gln Gly Asn Pro Val Phe Asp Tyr Ile Trp Asn Thr Val 165 170 175 Arg Arg Arg Phe Gly Gly Arg Leu His Ala Arg Asn Asp Gly Ile Lys 180 185 190 Pro Phe Ile Gln Ser Val Arg Gln Gly Tyr Trp Gly Tyr Tyr Leu Pro 195 200 205 Asp Gln Asp His Gly Pro Glu His Ser Glu Phe Val Asp Phe Phe Ala 210 215 220 Thr Tyr Lys Ala Thr Leu Pro Ala Ile Gly Arg Leu Met Lys Val Cys 225 230 235 240 Arg Ala Arg Val Ile Pro Leu Phe Pro Val Tyr Asn Gly Lys Thr His 245 250 255 Arg Leu Thr Ile Gln Ile Arg Pro Pro Met Asp Asp Leu Leu Thr Ala 260 265 270 Asp Asp His Thr Ile Ala Arg Arg Met Asn Glu Glu Val Glu Ile Phe 275 280 285 Val Gly Pro His Pro Glu Gln Tyr Thr Trp Ile Leu Lys Leu Leu Lys 290 295 300 Thr Arg Lys Pro Gly Glu Ile Gln Pro Tyr Lys Arg Lys Asp Leu Tyr 305 310 315 320 Pro Ile Lys 3 21 DNA Artificial Sequence Description of Artificial Sequenceprimer 3 gttgactggg aaggtctgga g 21 4 20 DNA Artificial Sequence Description of Artificial Sequenceprimer 4 ctgaccgcgc tctatcgcgg 20

Claims (37)

What is claimed is:
1. A mutant Salmonella sp. comprising a genetically modified msbB gene in which the mutant Salmonella is capable of targeting a solid tumor when administered in vivo.
2. The mutant Salmonella of claim 1 which is designated YS1629 and having ATCC Accession No. 202025 or is designated YS1170 and having ATCC Accession No. 202024 or is designated YS8211 and having ATCC Accession No. 202026.
3. The mutant Salmonella of claim 1 which is selected from the group consisting of Salmonella typhi, Salmonella choleraesuis, and Salmonella enteritidis.
4. The mutant Salmonella of claim 1 which expresses an altered lipid A molecule.
5. The mutant Salmonella of claim 1 which induces TNFα expression at about 5 percent to about 40 percent of that induced by a wild type Salmonella sp.
6. The mutant Salmonella of claim 1 which induces TNFα expression at about 10 percent to about 35 percent of that induced by a wild type Salmonella sp.
7. Lipopolysaccharide purified from the mutant Salmonella of claim 1 which induces TNFα expression at less than or equal to 0.001 percent of that induced by a wild type Salmonella sp.
8. The mutant Salmonella of claim 1 in which a chelating agent inhibits growth by about 90 percent compared 35 to the growth of a wild type Salmonella sp.
9. The mutant Salmonella of claim 1 in which a chelating agent inhibits growth by about 99 percent compared to the growth of a wild type Salmonella sp.
10. The mutant Salmonella of claim 1 in which a chelating agent inhibits growth greater than 99 percent compared to the growth of a wild type Salmonella sp.
11. The mutant Salmonella of claim 8, 9, or 10 in which the chelating agent is selected from the group consisting of Ethylenediaminetetraacetic Acid (EDTA), Ethylene Glycol-bis(β-aminoethyl Ether) N,N,N′,N′,-Tetraacetic Acid (EGTA) and sodium citrate.
12. The mutant Salmonella of claim 1 which survives in macrophages at about 50 percent to about 30 percent of the level of survival of a wild type Salmonella Sp.
13. The mutant Salmonella of claim 1 which survives in macrophages at about 30 percent to about 10 percent of the level of survival of a wild type Salmonella sp.
14. The mutant Salmonella of claim 1 which survives in macrophages at about 10 percent to about 1 percent of the level of survival of a wild type Salmonella sp.
15. A method of inhibiting growth or reducing volume of a solid tumor cancer, comprising administering an effective amount of the mutant Salmonella sp. of claim 1 to a patient having a solid tumor cancer.
16. The method according to claim 15 in which the mutant Salmonella is selected from the group consisting of Salmonella typhi, Salmonella choleraesuis, and Salmonella enteritidis.
17. The method according to claim 15 in which the mutant Salmonella expresses an altered lipid A molecule.
18. The method according to claim 15 in which the mutant Salmonella induces TNFα expression at about 5 percent to about 40 percent of that induced by a wild-type Salmonella sp.
19. The method according to claim 15 in which the mutant Salmonella induces TNFα expression at about 10 percent to about 35 percent of that induced by a wild-type Salmonella sp.
20. The method according to claim 15 in which lipopolysaccharide purified from the mutant Salmonella induces TNFα expression at less than or equal to 0.001 percent of that induced by a wild type Salmonella sp.
21. The method according to claim 15 in which a chelating agent inhibits growth of the mutant Salmonella by about 90 percent compared to the growth of a wild-type Salmonella sp.
22. The method according to claim 15 in which a chelating agent inhibits growth of the mutant Salmonella by about 99 percent compared to the growth of a wild-type Salmonella sp.
23. The method according to claim 15 in which a chelating agent inhibits growth of the mutant Salmonella by greater than 99 percent compared to the growth of a wild-type Salmonella sp.
24. The method according to claim 21, 22 or 23 in which the chelating agent is selected from the group consisting of EDTA, EGTA and sodium citrate.
25. The method according to claim 15 in which the mutant Salmonella survives in macrophages at about 50 percent to about 30 percent of the level of survival of a wild-type Salmonella sp.
26. The method according to claim 15 in which the mutant Salmonella survives in macrophages at about 30 percent to about 10 percent of the level of survival of a wild-type Salmonella sp.
27. The method according to claim 15 in which the mutant Salmonella survives in macrophages at about 10 percent to about 1 percent of the level of survival of a wild-type Salmonella sp.
28. The method according to claim 15 in which the solid tumor cancer is melanoma.
29. The method according to claim 15 in which the solid tumor cancer is colon carcinoma.
30. The method according to claim 15 in which the solid tumor cancer is selected from the group consisting of lung cancer, liver cancer, kidney cancer, prostate cancer, and breast cancer.
31. A pharmaceutical composition comprising an amount of the mutant Salmonella of claim 1 effective to inhibit growth or reduce volume of a solid tumor cancer; and a pharmaceutically acceptable carrier.
32. A mutant Salmonella sp. comprising a genetically modified msbb gene and a genetically modified purI gene in which the mutant Salmonella sp. is capable of targeting a solid tumor when administered in vivo.
33. A mutant Salmonella sp. of claim 32 in which the genetic modifications are deletion mutations.
34. The mutant Salmonella sp. of claim 32 which is designated YS1646 and having ATCC Accession No. 202165 or is designated YS1456 and having the ATCC Accession No. 202164.
35. A mutant Salmonella sp. comprising a genetically modified msbB gene and a genetically modified biosynthetic pathway gene in which the biosynthetic pathway mutation confers attenuated virulence.
36. A method of inhibiting growth or reducing volume of a solid tumor cancer, comprising administering an effective amount of the mutant Salmonella sp. of claim 32 to a patient having a solid tumor cancer.
37. An improved method for selecting genetic alterations of a bacterium, wherein the improvement comprises selecting a phenotypic variant which, when grown on a medium containing sucrose, produces colonies in which the edges of the mutant colonies are fuzzy or rough.
US09/149,832 1997-09-10 1998-09-08 Genetically modified tumor-targeted bacteria with reduced virulence Expired - Lifetime US6447784B1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
US09/149,832 US6447784B1 (en) 1997-09-10 1998-09-08 Genetically modified tumor-targeted bacteria with reduced virulence
CNB98811030XA CN1253551C (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
IL13493698A IL134936A0 (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
BR9812079-4A BR9812079A (en) 1997-09-10 1998-09-09 Salmonella sp. mutant, lipopolysaccharide, process to inhibit the growth or reduce the volume of a solid tumor cancer, pharmaceutical composition, and, improved process to select genetic alterations in a bacterium.
JP2000510842A JP2002500001A (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeting bacteria with reduced toxicity
AT98946891T ATE447005T1 (en) 1997-09-10 1998-09-09 GENETICALLY MODIFIED TUMOR TARGETED BACTERIA WITH REDUCED VIRULENCE
PCT/US1998/018701 WO1999013053A1 (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
CA2302866A CA2302866C (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
NZ503376A NZ503376A (en) 1997-09-10 1998-09-09 Genetically modified Salmonella sp having a modified msbB gene useful for treating tumours
AU93807/98A AU749695B2 (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
DE69841261T DE69841261D1 (en) 1997-09-10 1998-09-09 GENETICALLY MODIFIED, TUMORED TARGET BACTERIA WITH REDUCED VIRULENCE
KR1020007002535A KR20010015577A (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
EP98946891A EP1012232B1 (en) 1997-09-10 1998-09-09 Genetically modified tumor-targeted bacteria with reduced virulence
HK01104342A HK1033956A1 (en) 1997-09-10 2001-06-21 Genetically modified tumor-targeted bacteria with reduced virulence
US10/187,278 US6863894B2 (en) 1997-09-10 2002-06-27 Genetically modified tumor-targeted bacteria with reduced virulence
US11/064,533 US7354592B2 (en) 1997-09-10 2005-02-23 Genetically modified tumor-targeted bacteria with reduced virulence

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/926,636 US6080849A (en) 1997-09-10 1997-09-10 Genetically modified tumor-targeted bacteria with reduced virulence
US09/149,832 US6447784B1 (en) 1997-09-10 1998-09-08 Genetically modified tumor-targeted bacteria with reduced virulence

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/926,636 Continuation-In-Part US6080849A (en) 1997-09-10 1997-09-10 Genetically modified tumor-targeted bacteria with reduced virulence

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/187,278 Continuation US6863894B2 (en) 1997-09-10 2002-06-27 Genetically modified tumor-targeted bacteria with reduced virulence

Publications (2)

Publication Number Publication Date
US20020026655A1 true US20020026655A1 (en) 2002-02-28
US6447784B1 US6447784B1 (en) 2002-09-10

Family

ID=25453492

Family Applications (5)

Application Number Title Priority Date Filing Date
US08/926,636 Expired - Lifetime US6080849A (en) 1997-09-10 1997-09-10 Genetically modified tumor-targeted bacteria with reduced virulence
US09/149,832 Expired - Lifetime US6447784B1 (en) 1997-09-10 1998-09-08 Genetically modified tumor-targeted bacteria with reduced virulence
US09/337,689 Expired - Fee Related US6475482B1 (en) 1997-09-10 1999-06-22 Genetically modified tumor-targeted bacteria with reduce virulence
US10/125,328 Expired - Fee Related US6923972B2 (en) 1997-09-10 2002-04-18 Methods for use of genetically modified tumor-targeted bacteria with reduced virulence
US11/117,085 Expired - Fee Related US7514089B2 (en) 1997-09-10 2005-04-28 Genetically modified tumor-targeted bacteria with reduced virulence

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/926,636 Expired - Lifetime US6080849A (en) 1997-09-10 1997-09-10 Genetically modified tumor-targeted bacteria with reduced virulence

Family Applications After (3)

Application Number Title Priority Date Filing Date
US09/337,689 Expired - Fee Related US6475482B1 (en) 1997-09-10 1999-06-22 Genetically modified tumor-targeted bacteria with reduce virulence
US10/125,328 Expired - Fee Related US6923972B2 (en) 1997-09-10 2002-04-18 Methods for use of genetically modified tumor-targeted bacteria with reduced virulence
US11/117,085 Expired - Fee Related US7514089B2 (en) 1997-09-10 2005-04-28 Genetically modified tumor-targeted bacteria with reduced virulence

Country Status (5)

Country Link
US (5) US6080849A (en)
KR (1) KR20010015577A (en)
AT (1) ATE447005T1 (en)
DE (1) DE69841261D1 (en)
ZA (1) ZA988289B (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070134214A1 (en) * 2003-08-13 2007-06-14 Feng Xu Inactivated host cell delivery of polynucleotides encoding immunogens
WO2010033798A2 (en) * 2008-09-18 2010-03-25 Aviex Technologies Llc Live bacterial vaccines resistant to carbon dioxide (co2), acidic ph and/or osmolarity for viral infection prophylaxis or treatment
US9486513B1 (en) 2010-02-09 2016-11-08 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US9593339B1 (en) 2013-02-14 2017-03-14 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US9657085B1 (en) 2009-02-09 2017-05-23 David Gordon Bermudes Protease inhibitor: protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US9737592B1 (en) 2014-02-14 2017-08-22 David Gordon Bermudes Topical and orally administered protease inhibitors and bacterial vectors for the treatment of disorders and methods of treatment
US9878023B1 (en) 2010-02-09 2018-01-30 David Gordon Bermudes Protease inhibitor: protease sensitive expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
US10087451B2 (en) 2006-09-22 2018-10-02 Aviex Technologies Llc Live bacterial vectors for prophylaxis or treatment
US10676723B2 (en) 2015-05-11 2020-06-09 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US10857233B1 (en) 2010-02-09 2020-12-08 David Gordon Bermudes Protease inhibitor combination with therapeutic proteins including antibodies
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11168326B2 (en) 2017-07-11 2021-11-09 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11471497B1 (en) 2019-03-13 2022-10-18 David Gordon Bermudes Copper chelation therapeutics
US11779612B2 (en) 2019-01-08 2023-10-10 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5997881A (en) * 1995-11-22 1999-12-07 University Of Maryland, Baltimore Method of making non-pyrogenic lipopolysaccharide or A
US6207648B1 (en) * 1997-07-24 2001-03-27 Trustees Of Boston University Methods of using cytochrome P450 reductase for the enhancement of P450-based anti-cancer gene therapy
JP2002500001A (en) * 1997-09-10 2002-01-08 ヴァイオン ファーマシューティカルズ、インコーポレーテッド Genetically modified tumor-targeting bacteria with reduced toxicity
US6080849A (en) * 1997-09-10 2000-06-27 Vion Pharmaceuticals, Inc. Genetically modified tumor-targeted bacteria with reduced virulence
US6962696B1 (en) 1999-10-04 2005-11-08 Vion Pharmaceuticals Inc. Compositions and methods for tumor-targeted delivery of effector molecules
CN1420783A (en) * 1999-10-04 2003-05-28 维昂药品公司 Compositions and methods for tumor-targeted delivery of effector molecules
US20020054865A1 (en) * 2000-09-21 2002-05-09 Minoru Fujimori Anaerobic bacterium as a drug for cancer gene therapy
US7396822B2 (en) 2001-05-24 2008-07-08 Vaxiion Therapeutics, Inc. Immunogenic minicells and methods of use
US20030194798A1 (en) 2001-05-24 2003-10-16 Surber Mark W. Minicell compositions and methods
EP1281767A3 (en) 2001-07-31 2003-05-28 Aladar A. Szalay Light emitting microorganisms and cells for diagnosis and therapy of tumors
US7015027B1 (en) 2001-11-20 2006-03-21 Mds (Canada) Inc. Radiation therapy by accumulation of therapeutic radionuclides in tumor-targeting bacteria
WO2003063593A1 (en) * 2002-01-28 2003-08-07 Vion Pharmaceuticals, Inc. Methods for treating cancer by administering tumor-targetted bacteria and an immunomodulatory agent
US20030207833A1 (en) * 2002-02-25 2003-11-06 Neil Berkley Pharmaceutical compositions with minicells
US20030224369A1 (en) * 2002-02-25 2003-12-04 Surber Mark W. Reverse screening and target identification with minicells
US20030224444A1 (en) * 2002-02-25 2003-12-04 Sabbadini Roger A. Antibodies to native conformations of membrane proteins
US20040005700A1 (en) * 2002-05-28 2004-01-08 Surber Mark W. Poroplasts
US20030228261A1 (en) * 2002-06-05 2003-12-11 Aladar Szalay Light emitting microorganisms and cells for diagnosis and therapy of diseases associated with wounded or inflamed tissue
EP1369491A1 (en) * 2002-06-05 2003-12-10 Aladar A. Szalay Light emitting microorganisms and cells for diagnosis and therapy of diseases associated with wounded or inflamed tissue
AU2004289953B2 (en) 2003-06-18 2008-09-25 Genelux Corporation Modified recombinant vaccina viruses and other microorganisms, uses thereof
US7390646B2 (en) * 2003-09-17 2008-06-24 The Regents Of The University Of California Bacterial vectors and methods of use thereof
US20050112097A1 (en) * 2003-11-26 2005-05-26 Hinton Golden S. Method for the treatment of cancer
US20070298012A1 (en) * 2003-12-16 2007-12-27 Ivan King Compositions and Methods for Tumor-Targeted Delivery of Effector Molecules
WO2006055024A2 (en) * 2004-04-05 2006-05-26 Vaxiion Therapeutics, Inc. Minicells as vaccines
US20050244375A1 (en) * 2004-04-29 2005-11-03 Leonard Arnold S Composition and method of cancer treatment
EP2365090A1 (en) 2004-05-21 2011-09-14 The Regents of The University of California Method for enhancing production of isoprenoid compounds
US20060003454A1 (en) * 2004-07-02 2006-01-05 Conjugon, Inc. Non-dividing donor cells for gene transfer
CA2586126C (en) 2004-11-24 2016-06-28 Yasunobu Kano Novel shuttle vector
US8734779B2 (en) * 2005-04-08 2014-05-27 Anaeropharma Science Inc. 5-fluorouracil-resistant bacteria and method for production thereof
US20060270040A1 (en) * 2005-05-26 2006-11-30 Conjugon, Inc. Compositions and methods for treating tissue
KR101444090B1 (en) 2005-07-05 2014-09-26 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 Polynucleotides encoding isoprenoid modifying enzymes and methods of use thereof
US7935505B2 (en) * 2006-05-24 2011-05-03 Scarab Genomics, Llc Plasmid DNA preparations and methods for producing same
MX284139B (en) 2006-05-26 2011-02-18 Amyris Biotechnologies Inc Production of isoprenoids.
TW200819540A (en) 2006-07-11 2008-05-01 Genelux Corp Methods and compositions for detection of microorganisms and cells and treatment of diseases and disorders
BRPI0716954A2 (en) 2006-09-26 2013-10-29 Univ California ISOPRENOID PRODUCTION AND ISOPRENOID PRECURSORS
US7998461B2 (en) * 2007-11-15 2011-08-16 University Of Massachusetts Salmonella cancer therapeutics and related therapeutic methods
EP2242516A1 (en) 2008-01-11 2010-10-27 Genelux Corporation Methods and compositions for detection of bacteria and treatment of diseases and disorders
WO2010151609A1 (en) * 2009-06-24 2010-12-29 Photoswitch Biosciences Inc. Photoswitch-enabled ion channel assay system
EP2569425B1 (en) 2010-05-10 2016-07-06 The Regents of The University of California Endoribonuclease compositions and methods of use thereof
WO2012008860A2 (en) 2010-07-16 2012-01-19 Auckland Uniservices Limited Bacterial nitroreductase enzymes and methods relating thereto
WO2012112696A1 (en) 2011-02-15 2012-08-23 Vaxiion Therapeutics, Inc. Therapeutic compositions and methods for antibody and fc-containing targeting molecule-based targeted delivery of bioactive molecules by bacterial minicells
US8859256B2 (en) 2011-10-05 2014-10-14 Genelux Corporation Method for detecting replication or colonization of a biological therapeutic
EP2791333B1 (en) 2011-12-16 2019-03-06 The Board of Trustees of the Leland Stanford Junior University Opsin polypeptides and methods of use thereof
WO2013128288A1 (en) * 2012-02-27 2013-09-06 Thelial Technologies S.A. Monomeric bacterial actin adp-ribosyltransferases as cancer chemotherapeutics
US20130280170A1 (en) 2012-04-20 2013-10-24 Aladar A. Szalay Imaging methods for oncolytic virus therapy
US9127284B2 (en) 2012-05-04 2015-09-08 The University Of Hong Kong Modified bacteria and their uses thereof for the treatment of cancer or tumor
AU2013290424B2 (en) 2012-07-19 2018-01-25 Redwood Bioscience, Inc. Antibody specific for CD22 and methods of use thereof
BR112015003326A2 (en) 2012-08-16 2017-07-04 Ipierian Inc methods of treatment of a tauopathy
CN110818798A (en) 2012-10-25 2020-02-21 美国比奥维拉迪维股份有限公司 Anti-complement C1s antibodies and uses thereof
EP2914291B1 (en) 2012-11-02 2022-02-23 Bioverativ USA Inc. Anti-complement c1s antibodies and uses thereof
CA2914768A1 (en) 2013-06-10 2014-12-18 Ipierian, Inc. Methods of treating a tauopathy
EP3581580A1 (en) 2014-03-28 2019-12-18 The Board of Trustees of the Leland Stanford Junior University Engineered light-activated anion channel proteins and methods of use thereof
US20180080051A1 (en) 2015-03-31 2018-03-22 Exeligen Scientific, Inc. Cas 9 retroviral integrase and cas 9 recombinase systems for targeted incorporation of a dna sequence into a genome of a cell or organism
CN108348598B (en) 2015-04-06 2023-09-01 比奥贝拉蒂美国公司 Humanized anti-C1 s antibodies and methods of use thereof
RU2760582C2 (en) 2015-09-02 2021-11-29 Иммутеп С.А.С. Anti-lag-3 antibodies
EP3452510A4 (en) 2016-04-04 2020-01-15 Bioverativ USA Inc. Anti-complement factor bb antibodies and uses thereof
SG11201903012RA (en) 2016-10-12 2019-05-30 Bioverativ Usa Inc Anti-c1s antibodies and methods of use thereof
BR112019022500A2 (en) 2017-04-27 2020-06-16 Regents Of The University Of California MICRO-ORGANISMS AND METHODS TO PRODUCE CANABINOIDS AND CANABINOID DERIVATIVES
WO2019075220A1 (en) 2017-10-11 2019-04-18 Bioverativ Usa Inc. Methods of inducing complement activity
KR20210032928A (en) 2018-03-08 2021-03-25 뉴 아틀라스 바이오테크놀로지스 엘엘씨 How to produce tryptamine
WO2020047161A2 (en) 2018-08-28 2020-03-05 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof
WO2020176809A1 (en) 2019-02-27 2020-09-03 Actym Therapeutics, Inc. Immunostimulatory bacteria engineered to colonize tumors, tumor-resident immune cells, and the tumor microenvironment
WO2020206033A1 (en) 2019-04-02 2020-10-08 Kenjockety Biotechnology, Inc. Efflux pump-cancer antigen multi-specific antibodies and compositions, reagents, kits and methods related thereto
JP2023501539A (en) 2019-11-12 2023-01-18 アクティム・セラピューティクス・インコーポレイテッド An immunostimulatory bacterial delivery platform and its use for delivery of therapeutic products
WO2021113736A1 (en) 2019-12-05 2021-06-10 Massachusetts Institute Of Technology Single-domain antibody to chloramphenicol
AU2021283080A1 (en) 2020-06-04 2022-12-15 Kenjockety Biotechnology, Inc. ABCG2 efflux pump-cancer antigen multi-specific antibodies and compositions, reagents, kits and methods related thereto
CA3191433A1 (en) 2020-08-12 2022-02-17 Actym Therapeutics, Inc. Immunostimulatory bacteria-based vaccines, therapeutics, and rna delivery platforms
WO2023086796A2 (en) 2021-11-09 2023-05-19 Actym Therapeutics, Inc. Immunostimulatory bacteria for converting macrophages into a phenotype amenable to treatment, and companion diagnostic for identifying subjects for treatment
WO2023114658A1 (en) 2021-12-13 2023-06-22 Kenjockety Biotechnology, Inc. Anti-abcb1 antibodies
WO2023159220A1 (en) 2022-02-18 2023-08-24 Kenjockety Biotechnology, Inc. Anti-cd47 antibodies
WO2023192869A1 (en) 2022-03-28 2023-10-05 University Of Massachusetts Antigen delivering salmonella for use as a tumor homing beacon to refocus preexisting, vaccine generated t cells to combat cancer

Family Cites Families (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US29043A (en) * 1860-07-10 Improvement in cultivators
JPS57142256A (en) 1981-02-25 1982-09-02 Kao Corp Sanitary napkin
US4436727A (en) * 1982-05-26 1984-03-13 Ribi Immunochem Research, Inc. Refined detoxified endotoxin product
ATE159858T1 (en) * 1983-09-26 1997-11-15 Ehrenfeld Udo AGENT AND PRODUCT FOR THE DIAGNOSIS AND THERAPY OF TUMORS AND FOR THE TREATMENT OF WEAKNESSES OF THE CELLULAR AND HUMORAL IMMUNE DEFENSE
CA1321962C (en) 1985-03-20 1993-09-07 Aizo Matsushiro Dental caries preventive preparations and method for preparing said preparations
JPH0696538B2 (en) 1985-12-19 1994-11-30 株式会社アドバンス Anti-carcinogen
JPH0761950B2 (en) 1986-10-17 1995-07-05 塩野義製薬株式会社 Antitumor agent
DE3735381A1 (en) 1987-03-31 1989-05-03 Boehringer Mannheim Gmbh RECOMBINANT DNA FOR REPRESSIBLE AND INDUCIBLE EXPRESSION OF FOREIGN GENES
GB8730037D0 (en) 1987-12-23 1988-02-03 Wellcome Found Vaccines
JPH01180830A (en) 1988-01-11 1989-07-18 Kayaku:Kk Antitumor agent
EP0338679A3 (en) 1988-03-24 1991-03-06 Genentech, Inc. Tumour necrosis factor in the treatment of bladder cancer
JPH0284172A (en) 1988-07-21 1990-03-26 Smithkline Beckman Corp Salmonella tansformant having manifestation capacity as different kind of gene and useful for recombinant vaccine
GB8912330D0 (en) 1989-05-30 1989-07-12 Wellcome Found Live vaccines
JPH0376580A (en) * 1989-08-17 1991-04-02 Japan Tobacco Inc Escherichia coli manifestation vector and production of antiviral protein using the same
ATE167061T1 (en) 1989-11-03 1998-06-15 Univ Washington CROSS-PROTECTIVE SALMONELLA VACCINES
US5830702A (en) * 1990-10-31 1998-11-03 The Trustees Of The University Of Pennsylvania Live, recombinant listeria monocytogenes and production of cytotoxic T-cell response
WO1992011361A1 (en) * 1990-12-18 1992-07-09 The General Hospital Corporation Improved vaccines
US5695983A (en) * 1990-12-18 1997-12-09 The General Hospital Corporation Salmonella vaccines
AU664360B2 (en) 1991-03-05 1995-11-16 Wellcome Foundation Limited, The Expression of recombinant proteins in attenuated bacteria
IL101409A0 (en) 1992-03-29 1992-11-15 Era Masis Ltd Method for the early diagnosis of cancer
IL101410A0 (en) 1992-03-29 1992-11-15 Era Masis Ltd Formulation for the treatment of cancer
JPH06298657A (en) 1993-04-15 1994-10-25 Nippon Shoji Kk Immunopotentiation agent
CA2146242A1 (en) 1993-08-25 1995-03-02 Kazuo Ichihara Heart function restorative
US5631236A (en) 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
JPH09504518A (en) 1993-10-06 1997-05-06 アメリカ合衆国 Treatment of tumors by gene transfer of tumor cells with genes encoding negative selectable markers and cytokines
IT1270123B (en) * 1994-10-05 1997-04-28 Dompe Spa PHARMACEUTICAL COMPOSITIONS CONTAINING ENGINEERED MICROORGANISMS AND THEIR USE FOR THERAPY
US6051237A (en) * 1994-11-08 2000-04-18 The Trustees Of The University Of Pennsylvania Specific immunotherapy of cancer using a live recombinant bacterial vaccine vector
US6150170A (en) * 1998-05-03 2000-11-21 University Of Maryland At Baltimore Method for introducing and expressing genes in animal cells, and live invasive bacterial vectors for use in the same
US5877159A (en) 1995-05-03 1999-03-02 University Of Maryland At Baltimore Method for introducing and expressing genes in animal cells and live invasive bacterial vectors for use in the same
US5705151A (en) * 1995-05-18 1998-01-06 National Jewish Center For Immunology & Respiratory Medicine Gene therapy for T cell regulation
US6190657B1 (en) 1995-06-07 2001-02-20 Yale University Vectors for the diagnosis and treatment of solid tumors including melanoma
AU731061B2 (en) 1995-09-06 2001-03-22 Department Of The Army, U.S. Government Bacterial delivery system
US5824538A (en) 1995-09-06 1998-10-20 The United States Of America As Represented By The Secretary Of The Army Shigella vector for delivering DNA to a mammalian cell
GB9521568D0 (en) 1995-10-20 1995-12-20 Lynxvale Ltd Delivery of biologically active polypeptides
WO1997018225A1 (en) 1995-11-14 1997-05-22 The General Hospital Corporation Salmonella secreted proteins and uses thereof
US5997881A (en) * 1995-11-22 1999-12-07 University Of Maryland, Baltimore Method of making non-pyrogenic lipopolysaccharide or A
US6887483B2 (en) * 1995-12-01 2005-05-03 University Of Iowa Research Foundation Non-toxic mutants of pathogenic gram-negative bacteria
WO1997025061A1 (en) * 1996-01-09 1997-07-17 Bristol-Myers Squibb Company De-myristolated lipopolysaccharide of gram-negative bacteria
PT884997E (en) 1996-02-16 2002-11-29 Janssen Pharmaceutica Nv COMPOSITIONS CONTAINING A FUNGICIDE AND A SULFURED COMPOUND
AU721819B2 (en) 1996-05-10 2000-07-13 Pharmacia & Upjohn Company Topical administration of antimicrobial agents for the treatment of systemic bacterial diseases
GB9621091D0 (en) 1996-10-09 1996-11-27 Fondation Pour Le Perfectionem Attenuated microorganisms strains and their uses
TW373083B (en) 1996-12-20 1999-11-01 Corning Inc Reflective coupling array for optical waveguide
WO1998033923A1 (en) 1997-01-30 1998-08-06 Imperial College Of Science, Technology & Medicine MUTANT msbB or htrB GENES
WO1998044131A1 (en) * 1997-03-28 1998-10-08 Walter Reed Army Institute Of Research Antimicrobial mediated bacterial dna delivery
US6537558B2 (en) * 1997-03-31 2003-03-25 Megan Health, Inc. Methods of producing and using virulence attenuated poxR mutant bacteria
US6080849A (en) * 1997-09-10 2000-06-27 Vion Pharmaceuticals, Inc. Genetically modified tumor-targeted bacteria with reduced virulence
JP2002500001A (en) * 1997-09-10 2002-01-08 ヴァイオン ファーマシューティカルズ、インコーポレーテッド Genetically modified tumor-targeting bacteria with reduced toxicity
US6143551A (en) * 1997-12-29 2000-11-07 Schering Aktiengesellschaft Delivery of polypeptide-encoding plasmid DNA into the cytosol of macrophages by attenuated listeria suicide bacteria
US6593592B1 (en) * 1999-01-29 2003-07-15 Semiconductor Energy Laboratory Co., Ltd. Semiconductor device having thin film transistors
US6962696B1 (en) * 1999-10-04 2005-11-08 Vion Pharmaceuticals Inc. Compositions and methods for tumor-targeted delivery of effector molecules
CN1420783A (en) 1999-10-04 2003-05-28 维昂药品公司 Compositions and methods for tumor-targeted delivery of effector molecules
EP1281767A3 (en) * 2001-07-31 2003-05-28 Aladar A. Szalay Light emitting microorganisms and cells for diagnosis and therapy of tumors
US20070298012A1 (en) * 2003-12-16 2007-12-27 Ivan King Compositions and Methods for Tumor-Targeted Delivery of Effector Molecules

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070134214A1 (en) * 2003-08-13 2007-06-14 Feng Xu Inactivated host cell delivery of polynucleotides encoding immunogens
US10087451B2 (en) 2006-09-22 2018-10-02 Aviex Technologies Llc Live bacterial vectors for prophylaxis or treatment
WO2010033798A2 (en) * 2008-09-18 2010-03-25 Aviex Technologies Llc Live bacterial vaccines resistant to carbon dioxide (co2), acidic ph and/or osmolarity for viral infection prophylaxis or treatment
WO2010033798A3 (en) * 2008-09-18 2010-05-14 Aviex Technologies Llc Live bacterial vaccines resistant to carbon dioxide (co2), acidic ph and/or osmolarity for viral infection prophylaxis or treatment
US11485773B1 (en) 2009-02-09 2022-11-01 David Gordon Bermudes Protease inhibitor:protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US10590185B1 (en) 2009-02-09 2020-03-17 David Gordon Bermudes Protease inhibitor: protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US9657085B1 (en) 2009-02-09 2017-05-23 David Gordon Bermudes Protease inhibitor: protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US11219671B1 (en) 2010-02-09 2022-01-11 David Gordon Bermudes Protease inhibitor:protease sensitive expression system, composition and methods for improving the therapeutic activity and specificity of proteins delivered by bacteria
US10857233B1 (en) 2010-02-09 2020-12-08 David Gordon Bermudes Protease inhibitor combination with therapeutic proteins including antibodies
US10364435B1 (en) 2010-02-09 2019-07-30 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US9486513B1 (en) 2010-02-09 2016-11-08 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US9878023B1 (en) 2010-02-09 2018-01-30 David Gordon Bermudes Protease inhibitor: protease sensitive expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
US10954521B1 (en) 2010-02-09 2021-03-23 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US10501746B1 (en) 2013-02-14 2019-12-10 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US9593339B1 (en) 2013-02-14 2017-03-14 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US11827890B1 (en) 2013-02-14 2023-11-28 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US9737592B1 (en) 2014-02-14 2017-08-22 David Gordon Bermudes Topical and orally administered protease inhibitors and bacterial vectors for the treatment of disorders and methods of treatment
US10828350B1 (en) 2014-02-14 2020-11-10 David Gordon Bermudes Topical and orally administered protease inhibitors and bacterial vectors for the treatment of disorders and methods of treatment
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US10828356B1 (en) 2014-09-18 2020-11-10 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US10729731B1 (en) 2014-09-18 2020-08-04 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US11813295B1 (en) 2014-09-18 2023-11-14 Theobald Therapeutics LLC Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US10449237B1 (en) 2014-09-18 2019-10-22 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US11633435B1 (en) 2014-09-18 2023-04-25 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US10676723B2 (en) 2015-05-11 2020-06-09 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11168326B2 (en) 2017-07-11 2021-11-09 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof
US11779612B2 (en) 2019-01-08 2023-10-10 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof
US11471497B1 (en) 2019-03-13 2022-10-18 David Gordon Bermudes Copper chelation therapeutics
US11406702B1 (en) 2020-05-14 2022-08-09 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated Salmonella as a vaccine
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine

Also Published As

Publication number Publication date
US6447784B1 (en) 2002-09-10
US7514089B2 (en) 2009-04-07
DE69841261D1 (en) 2009-12-10
KR20010015577A (en) 2001-02-26
US20030170276A1 (en) 2003-09-11
ATE447005T1 (en) 2009-11-15
US6080849A (en) 2000-06-27
US6923972B2 (en) 2005-08-02
US6475482B1 (en) 2002-11-05
US20070009489A1 (en) 2007-01-11
ZA988289B (en) 1999-03-22

Similar Documents

Publication Publication Date Title
US6447784B1 (en) Genetically modified tumor-targeted bacteria with reduced virulence
US7354592B2 (en) Genetically modified tumor-targeted bacteria with reduced virulence
US7887816B2 (en) Attenuated microorganisms for the treatment of infection
US5527529A (en) Vaccines comprising attenuated salmonella having mutations in the ompr genes
EP0717777B1 (en) Salmonella vaccines
US4735801A (en) Novel non-reverting salmonella live vaccines
US4550081A (en) Non-reverting salmonella
US6010901A (en) Salmonella virulence genes
AU666108B2 (en) CDT mutants of salmonella typhi
EP1129196B1 (en) Virulence genes and proteins, and their use
Bermudes et al. Tumour-selective Salmonella-based cancer therapy
MXPA00002355A (en) Genetically modified tumor-targeted bacteria with reduced virulence
US20030059442A1 (en) Attenuated microorganisms for the treatment of infection
EP0249449A1 (en) Bacterial strain for live vaccines
JPH09511139A (en) Vaccine composition
EP1037664A1 (en) Vaccines containing attenuated bacteria

Legal Events

Date Code Title Description
AS Assignment

Owner name: YALE UNIVERSITY, CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LOW, KENNETH BROOKS;REEL/FRAME:009608/0411

Effective date: 19981019

Owner name: VION PHARMACEUTICALS, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BERMUDES, DAVID;ITTENSOHN, MARTINA;REEL/FRAME:009608/0409

Effective date: 19981019

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: YALE UNIVERSITY, CONNECTICUT

Free format text: CHANGE OF ASSIGNEE ADDRESS;ASSIGNOR:YALE UNIVERSITY;REEL/FRAME:022939/0542

Effective date: 19980930

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12