Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(22): 36492-36508, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28445131

ABSTRACT

The potential of bacteria-mediated tumor therapy (BMTT) is highlighted by more than a century of investigation. Attenuated Salmonella has prevailed as promising therapeutic agents. For BMTT - categorized as an immune therapy - the exact contribution of particular immune reactions to the therapeutic effect remains ambiguous. In addition, one could argue for or against the requirement of bacterial viability and tumor targeting. Herein we evaluate the isolated therapeutic efficacy of purified LPS and TNF-α, which together account for a dominant immunogenic pathway of gram negative bacteria like Salmonella. We show that therapeutic efficacy against CT26 tumors does not require bacterial viability. Analogous to viable Salmonella SL7207, tumor regression by a specific CD8+ T cell response can be induced by purified LPS or recombinant TNF-α (rTNF-α). Conversely, therapeutic effects against RenCa tumors were abrogated upon bacterial avitalization and limited using isolated adjuvants. This argues for an alternative mechanistic explanation for SL7207 against RenCa that depends on viability and persistence. Unable to boost bacterial therapies by co-injection of rTNF-α suggested therapeutic effects along this axis are exhausted by the intrinsic adjuvanticity of bacteria alone. However, the importance of TNF-α for BMTT was highlighted by its support of tumor invasion and colonization in concert with lower infective doses of Salmonella. In consideration, bacterial therapeutic effectiveness along the axis of LPS and TNF-α appears limited, and does not offer the necessary plasticity for different tumors. This emphasizes a need for recombinant strengthening and vehicular exploitation to accommodate potency, plasticity and distinctiveness in BMTT.


Subject(s)
Immunotherapy , Lipopolysaccharides/immunology , Salmonella/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Female , Humans , Immunity, Innate , Immunologic Factors/pharmacology , Immunotherapy/methods , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Neoplasm Regression, Spontaneous/immunology , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
2.
Cancer Res ; 77(7): 1553-1563, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202530

ABSTRACT

Intentional bacterial infections can produce efficacious antitumor responses in mice, rats, dogs, and humans. However, low overall success rates and intense side effects prevent such approaches from being employed clinically. In this work, we titered bacteria and/or the proinflammatory cytokine TNFα in a set of established murine models of cancer. To interpret the experiments conducted, we considered and calibrated a tumor-effector cell recruitment model under the influence of functional tumor-associated vasculature. In this model, bacterial infections and TNFα enhanced immune activity and altered vascularization in the tumor bed. Information to predict bacterial therapy outcomes was provided by pretreatment tumor size and the underlying immune recruitment dynamics. Notably, increasing bacterial loads did not necessarily produce better long-term tumor control, suggesting that tumor sizes affected optimal bacterial loads. Short-term treatment responses were favored by high concentrations of effector cells postinjection, such as induced by higher bacterial loads, but in the longer term did not correlate with an effective restoration of immune surveillance. Overall, our findings suggested that a combination of intermediate bacterial loads with low levels TNFα administration could enable more favorable outcomes elicited by bacterial infections in tumor-bearing subjects. Cancer Res; 77(7); 1553-63. ©2017 AACR.


Subject(s)
Bacterial Infections/immunology , Neoplasms/therapy , Animals , Bacterial Load , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Models, Theoretical , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/microbiology , Tumor Burden , Tumor Necrosis Factor-alpha/therapeutic use
3.
PLoS One ; 11(5): e0155338, 2016.
Article in English | MEDLINE | ID: mdl-27171245

ABSTRACT

Systemically applied Salmonella enterica spp. have been shown to invade and colonize neoplastic tissues where it retards the growth of many tumors. This offers the possibility to use the bacteria as a vehicle for the tumor specific delivery of therapeutic molecules. Specificity of such delivery is solely depending on promoter sequences that control the production of a target molecule. We have established the functional structure of bacterial promoters that are transcriptionally active exclusively in tumor tissues after systemic application. We observed that the specific transcriptional activation is accomplished by a combination of a weak basal promoter and a strong FNR binding site. This represents a minimal set of control elements required for such activation. In natural promoters, additional DNA remodeling elements are found that alter the level of transcription quantitatively. Inefficiency of the basal promoter ensures the absence of transcription outside tumors. As a proof of concept, we compiled an artificial promoter sequence from individual motifs representing FNR and basal promoter and showed specific activation in a tumor microenvironment. Our results open possibilities for the generation of promoters with an adjusted level of expression of target proteins in particular for applications in bacterial tumor therapy.


Subject(s)
Neoplasms/genetics , Promoter Regions, Genetic , Salmonella enterica/genetics , Animals , Base Sequence , Female , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Liver/metabolism , Mice, Inbred BALB C , Nucleotide Motifs/genetics , Spleen/metabolism , Tumor Microenvironment
4.
Oncotarget ; 7(16): 22605-22, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26981777

ABSTRACT

To date, virulent bacteria remain the basis of most bacteria mediated cancer therapies. For clinical application attenuation is required. However, this might result in a drastically lowered therapeutic capacity. Herein we argue that the E. coli probiotic Symbioflor-2, with a history of safe application may constitute a viable tumor therapeutic candidate. We demonstrate that Symbioflor-2 displays a highly specific tumor targeting ability as determined in murine CT26 and RenCa tumor models. The excellent specificity was ascribed to reduced levels of adverse colonization. A high safety standard was demonstrated in WT and Rag1-/- mice. Thus, Symbioflor-2 may represent an ideal tumor targeting delivery system for therapeutic molecules. Moreover, Symbioflor-2 was capable of inducing CT26 tumor clearance as result of an adjuvant effect on tumor specific CD8+ T cells analogous to the Salmonella variant SL7207. However, lower therapeutic efficacy against RenCa tumors suggested a generally reduced therapeutic potency for probiotics. Interestingly, concurrent depletion of Gr-1+ or Ly6G+ cells installed therapeutic efficacy equal to SL7207, thus highlighting the role of innate effector cells in restraining the anti-tumor effects of Symbioflor-2. Collectively, our findings argue for a strategy of safe strain application and a more sustainable use of bacteria as a delivery system for therapeutic molecules.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/pharmacology , Escherichia coli , Neoplasms , Probiotics/pharmacology , Animals , Drug Delivery Systems/methods , Mice , Mice, Inbred BALB C
5.
Methods Mol Biol ; 1409: 61-8, 2016.
Article in English | MEDLINE | ID: mdl-26846802

ABSTRACT

Non-invasive in vivo imaging represents a powerful tool to monitor cellular and molecular processes in the living animal. In the special case of bacteria-mediated cancer therapy using bioluminescent bacteria, it opens up the possibility to follow the course of the microorganisms into the tumor via the circulation. The mechanism by which bacteria elicit their anti-tumor potential is not completely understood. However, this knowledge is crucial to improve bacteria as an anti-cancer tool that can be introduced into the clinic. For the study of these aspects, in vivo imaging can be considered a key technology.


Subject(s)
Bacteria/metabolism , Diagnostic Imaging/methods , Neoplasms/diagnosis , Neoplasms/metabolism , Animals , Bacteria/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Genes, Reporter , Humans , Molecular Imaging/methods , Neoplasms/genetics
6.
Int J Cancer ; 137(8): 2019-28, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-25868911

ABSTRACT

Facultative anaerobic bacteria like E. coli can colonize solid tumors often resulting in tumor growth retardation or even clearance. Little mechanistic knowledge is available for this phenomenon which is however crucial for optimization and further implementation in the clinic. Here, we show that intravenous injections with E. coli TOP10 can induce clearance of CT26 tumors in BALB/c mice. Importantly, re-challenging mice which had cleared tumors showed that clearance was due to a specific immune reaction. Accordingly, lymphopenic mice never showed tumor clearance after infection. Depletion experiments revealed that during induction phase, CD8(+) T cells are the sole effectors responsible for tumor clearance while in the memory phase CD8(+) and CD4(+) T cells were involved. This was confirmed by adoptive transfer. CD4(+) and CD8(+) T cells could reject newly set tumors while CD8(+) T cells could even reject established tumors. Detailed analysis of adoptively transferred CD4(+) T cells during tumor challenge revealed expression of granzyme B, FasL, TNF-α and IFN-γ in such T cells that might be involved in the anti-tumor activity. Our findings should pave the way for further optimization steps of this promising therapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/microbiology , Colonic Neoplasms/therapy , Escherichia coli/immunology , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Fas Ligand Protein/metabolism , Female , Granzymes/metabolism , Immunotherapy, Adoptive , Injections, Intravenous , Interferon-gamma , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Tumor Necrosis Factor-alpha/metabolism
7.
PLoS Pathog ; 10(9): e1004385, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25210785

ABSTRACT

The coordinated action of a variety of virulence factors allows Salmonella enterica to invade epithelial cells and penetrate the mucosal barrier. The influence of the age-dependent maturation of the mucosal barrier for microbial pathogenesis has not been investigated. Here, we analyzed Salmonella infection of neonate mice after oral administration. In contrast to the situation in adult animals, we observed spontaneous colonization, massive invasion of enteroabsorptive cells, intraepithelial proliferation and the formation of large intraepithelial microcolonies. Mucosal translocation was dependent on enterocyte invasion in neonates in the absence of microfold (M) cells. It further resulted in potent innate immune stimulation in the absence of pronounced neutrophil-dominated pathology. Our results identify factors of age-dependent host susceptibility and provide important insight in the early steps of Salmonella infection in vivo. We also present a new small animal model amenable to genetic manipulation of the host for the analysis of the Salmonella enterocyte interaction in vivo.


Subject(s)
Enterocytes/microbiology , Epithelial Cells/microbiology , Macrophages/microbiology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Virulence/immunology , Age Factors , Animals , Animals, Newborn , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enterocytes/immunology , Enterocytes/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation, Bacterial , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/pathology
8.
BMC Bioinformatics ; 13: 202, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22897887

ABSTRACT

BACKGROUND: Transcriptional activity of genes depends on many factors like DNA motifs, conformational characteristics of DNA, melting etc. and there are computational approaches for their identification. However, in real applications, the number of predicted, for example, DNA motifs may be considerably large. In cases when various computational programs are applied, systematic experimental knock out of each of the potential elements obviously becomes nonproductive. Hence, one needs an approach that is able to integrate many heterogeneous computational methods and upon that suggest selected regulatory elements for experimental verification. RESULTS: Here, we present an integrative bioinformatic approach aimed at the discovery of regulatory modules that can be effectively verified experimentally. It is based on combinatorial analysis of known and novel binding motifs, as well as of any other known features of promoters. The goal of this method is the identification of a collection of modules that are specific for an established dataset and at the same time are optimal for experimental verification. The method is particularly effective on small datasets, where most statistical approaches fail. We apply it to promoters that drive tumor-specific gene expression in tumor-colonizing Gram-negative bacteria. The method successfully identified a number of potential modules, which required only a few experiments to be verified. The resulting minimal functional bacterial promoter exhibited high specificity of expression in cancerous tissue. CONCLUSIONS: Experimental analysis of promoter structures guided by bioinformatics has proved to be efficient. The developed computational method is able to include heterogeneous features of promoters and suggest combinatorial modules for experimental testing. Expansibility and robustness of the methodology implemented in the approach ensures good results for a wide range of problems.


Subject(s)
Promoter Regions, Genetic , Sequence Analysis, DNA , Algorithms , Computational Biology/methods , Gene Expression , Neoplasms/genetics , Nucleotide Motifs , Salmonella/genetics
9.
Nucleic Acids Res ; 40(7): 2984-94, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22140114

ABSTRACT

Conventional cancer therapies are often limited in effectiveness and exhibit strong side effects. Therefore, alternative therapeutic strategies are demanded. The employment of tumor-colonizing bacteria that exert anticancer effects is such a novel approach that attracts increasing attention. For instance, Salmonella enterica serovar Typhimurium has been used in many animal tumor models as well as in first clinical studies. These bacteria exhibit inherent tumoricidal effects. In addition, they can be used to deliver therapeutic agents. However, bacterial expression has to be restricted to the tumor to prevent toxic substances from harming healthy tissue. Therefore, we screened an S. Typhimurium promoter-trap library to identify promoters that exclusively drive gene expression in the cancerous tissue. Twelve elements could be detected that show reporter gene expression in tumors but not in spleen and liver. In addition, a DNA motif was identified that appears to be necessary for tumor specificity. Now, such tumor-specific promoters can be used to safely express therapeutic proteins by tumor-colonizing S. Typhimurium directly in the neoplasia.


Subject(s)
Gene Expression Regulation, Bacterial , Neoplasms, Experimental/microbiology , Promoter Regions, Genetic , Salmonella typhimurium/genetics , Animals , Cell Hypoxia , Cell Line, Tumor , Female , Genes, Reporter , Mice , Mice, Inbred BALB C , Nucleotide Motifs , Sequence Analysis, DNA
10.
J Mol Med (Berl) ; 88(8): 763-73, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20526574

ABSTRACT

Cancer has become the second ranking cause of death in the industrialized world. Conventional anti-cancer therapies such as surgery, radiotherapy, and chemotherapy are effective in the treatment of solid tumors only to some extent. Furthermore, they are often associated with severe side effects. Use of bacteria as alternative cancer therapeutics has sporadically been followed over more than a century. The potential to target and colonize solid tumors could be shown for many different bacteria in the meantime. Such bacteria are either obligate anaerobic bacteria like Clostridium or Bifidobacterium or facultative anaerobic like Escherichia coli or Salmonella. Here we describe bacterial strains that were successfully applied mostly in animals bearing model tumors, although first clinical trials have been reported as well. Our review mainly concentrates on Salmonella enterica serovar Typhimurium (S. Typhimurium) since these bacteria were studied most intensively thus far. Importantly, S. Typhimurium were shown not only to colonize large established tumors but also exhibit the property to invade and affect metastases. We report on a potential mechanism by which such bacteria can invade solid tumors. Furthermore, we describe several successful attempts in which the bacteria have been used as carriers for recombinant therapeutic molecules that render bacteria more powerful in eradication of the established tumor. Such attempts should be considered starting points on the way to an effective and safe tumor therapy with the help of bacteria.


Subject(s)
Neoplasms/therapy , Salmonella , Animals , Bacteria, Anaerobic/growth & development , Bacteria, Anaerobic/metabolism , Humans , Salmonella/growth & development , Salmonella/metabolism
11.
J Clin Invest ; 120(4): 1151-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20237412

ABSTRACT

Angiogenesis is a hallmark of malignant neoplasias, as the formation of new blood vessels is required for tumors to acquire oxygen and nutrients essential for their continued growth and metastasis. However, the signaling pathways leading to tumor vascularization are not fully understood. Here, using a transplantable mouse tumor model, we have demonstrated that endogenous IFN-beta inhibits tumor angiogenesis through repression of genes encoding proangiogenic and homing factors in tumor-infiltrating neutrophils. We determined that IFN-beta-deficient mice injected with B16F10 melanoma or MCA205 fibrosarcoma cells developed faster-growing tumors with better-developed blood vessels than did syngeneic control mice. These tumors displayed enhanced infiltration by CD11b+Gr1+ neutrophils expressing elevated levels of the genes encoding the proangiogenic factors VEGF and MMP9 and the homing receptor CXCR4. They also expressed higher levels of the transcription factors c-myc and STAT3, known regulators of VEGF, MMP9, and CXCR4. In vitro, treatment of these tumor-infiltrating neutrophils with low levels of IFN-beta restored expression of proangiogenic factors to control levels. Moreover, depletion of these neutrophils inhibited tumor growth in both control and IFN-beta-deficient mice. We therefore suggest that constitutively produced endogenous IFN-beta is an important mediator of innate tumor surveillance. Further, we believe our data help to explain the therapeutic effect of IFN treatment during the early stages of cancer development.


Subject(s)
Interferon-beta/physiology , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/etiology , Neutrophils/physiology , Animals , CD11b Antigen/analysis , Female , Genes, myc , Killer Cells, Natural/physiology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Radiation Tolerance , STAT3 Transcription Factor/genetics
12.
PLoS One ; 4(8): e6692, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19693266

ABSTRACT

BACKGROUND: Several facultative anaerobic bacteria with potential therapeutic abilities are known to preferentially colonize solid tumors after systemic administration. How they efficiently find and invade the tumors is still unclear. However, this is an important issue to be clarified when bacteria should be tailored for application in cancer therapy. METHODOLOGY/PRINCIPAL FINDINGS: We describe the initial events of colonization of an ectopic transplantable tumor by Salmonella enterica serovar Typhimurium. Initially, after intravenous administration, bacteria were found in blood, spleen, and liver. Low numbers were also detected in tumors associated with blood vessels as could be observed by immunohistochemistry. A rapid increase of TNF-alpha in blood was observed at that time, in addition to other pro-inflammatory cytokines. This induced a tremendous influx of blood into the tumors by vascular disruption that could be visualized in H&E stainings and quantified by hemoglobin measurements of tumor homogenate. Most likely, together with the blood, bacteria were flushed into the tumor. In addition, blood influx was followed by necrosis formation, bacterial growth, and infiltration of neutrophilic granulocytes. Depletion of TNF-alpha retarded blood influx and delayed bacterial tumor-colonization. CONCLUSION: Our findings emphasize similarities between Gram-negative tumor-colonizing bacteria and tumor vascular disrupting agents and show the involvement of TNF-alpha in the initial phase of tumor-colonization by bacteria.


Subject(s)
Hemorrhage/microbiology , Neoplasms, Experimental/microbiology , Salmonella enterica/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Base Sequence , DNA Primers , Female , Hemorrhage/chemically induced , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction
13.
Microbes Infect ; 11(14-15): 1097-105, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19665575

ABSTRACT

The probiotic bacterium Escherichia coli Nissle 1917 (EcN) constitutes a prospective vector for delivering heterologous therapeutic molecules to treat several human disorders. To add versatility to this carrier system, bacteria should be equipped with expression modules that can be regulated deliberately in a temporal and quantitative manner. This approach is called in vivo remote control (IVRC) of bacterial vectors. Here, we have evaluated promoters P(araBAD), P(rhaBAD) and P(tet), which can be induced with L-arabinose, L-rhamnose or anhydrotetracycline, respectively. EcN harboring promoter constructs with luciferase as reporter gene were administered either orally to healthy mice or intravenously to tumor bearing animals. Subsequent to bacterial colonization of tissues, inducer substances were administered via the oral or systemic route. By use of in vivo bioluminescence imaging, the time course of reporter gene expression was analyzed. Each promoter displayed a specific in vivo induction profile depending on the niche of bacterial residence and the route of inducer administration. Importantly, we also observed colonization of gall bladders of mice when EcN was administered systemically at high doses. Bacteria in this anatomical compartment remained accessible to remote control of bacterial gene expression.


Subject(s)
Escherichia coli/drug effects , Gallbladder/microbiology , Gene Expression Regulation, Bacterial/drug effects , Intestines/microbiology , Neoplasms, Experimental/microbiology , Probiotics , Animals , Arabinose/administration & dosage , Arabinose/pharmacology , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Female , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Rhamnose/administration & dosage , Rhamnose/pharmacology , Skin Neoplasms/microbiology , Tetracyclines/administration & dosage , Tetracyclines/pharmacology , Tissue Distribution
14.
Cancer Res ; 68(8): 2952-60, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413765

ABSTRACT

Administration of facultative anaerobic bacteria like Salmonella typhimurium, Shigella flexneri, and Escherichia coli to tumor-bearing mice leads to a preferential accumulation and proliferation of the microorganisms within the solid tumor. Until now, all known tumor-targeting bacteria have shown poor dissemination inside the tumors. They accumulate almost exclusively in large necrotic areas and spare a rim of viable tumor cells. Interestingly, the bacteria-containing necrotic region is separated from viable tumor cells by a barrier of host neutrophils that have immigrated into the tumor. We here report that depletion of host neutrophils results in a noticeably higher total number of bacteria in the tumor and that bacteria were now also able to migrate into vital tumor tissue. Most remarkably, an increase in the size of the necrosis was observed, and complete eradication of established tumors could be observed under these conditions. Thus, bacteria-mediated tumor therapy can be amplified by depletion of host neutrophils.


Subject(s)
Bacteria, Anaerobic/growth & development , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Neutrophils/microbiology , Animals , Bacteria, Anaerobic/isolation & purification , Cell Division , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Female , Mice , Mice, Inbred BALB C , Necrosis , Salmonella typhimurium/growth & development , Salmonella typhimurium/isolation & purification , Shigella flexneri/growth & development , Shigella flexneri/isolation & purification
15.
Int J Med Microbiol ; 298(1-2): 21-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17702649

ABSTRACT

Live attenuated bacteria are well established as vaccines. Thus, their use as carriers for prophylactic and therapeutic macromolecules is a logical consequence. Here we describe several experimental applications of bacteria to carry heterologous macromolecules into the murine host. First, Listeria monocytogenes are described that are able to transfer eukaryotic expression plasmids into host cells for gene therapy. High multiplicities of infection are still required for efficient gene transfer and we point out some of the bottlenecks that counteract a more efficient transfer and application in vivo. Then, we describe Salmonella enterica serovar Typhimurium (S. typhimurium) as an expression plasmid transfer vehicle for oral DNA vaccination of mice. We demonstrate that the stabilization of the plasmid transformants results in an improved immune response. Stabilization was achieved by replacing the origin of replication of the original high-copy-number plasmid by a low-copy-number origin. Finally, we describe Salmonella carriers for the improved expression of heterologous proteins. We introduce a system in which the plasmid is carried as a single copy during cultivation but is amplified several fold upon infection of the host. Using the same in vivo inducible promoter for both protein expression and plasmid amplification, a substantial increase in antigen expression in vivo can be achieved. A modification of this approach is the introduction of inducible gene expression in vivo with a low-molecular-weight compound. Using P(BAD) promoter and L-arabinose as inducer we were able to deliberately activate genes in the bacterial carrier. No background activity could be observed with P(BAD) such that an inducible suicide gene could be introduced. This is adding an important safety feature to such live attenuated carrier bacteria.


Subject(s)
Bacterial Vaccines/immunology , Listeria monocytogenes/immunology , Salmonella typhimurium/immunology , Vaccines, DNA/immunology , Animals , Arabinose/pharmacology , Mice , Plasmids/immunology , Transformation, Genetic/immunology , Vaccines, Attenuated/immunology
16.
Cell Microbiol ; 9(6): 1529-37, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17298393

ABSTRACT

We have used Salmonella enterica serovar Typhimurium (S. typhimurium) which are able to colonize tumours besides spleen and liver. Bacteria were equipped with constructs encoding green fluorescent protein or luciferase as reporters under control of the promoter PBAD that is inducible with L-arabinose. Reporter genes could be induced in culture but also when the bacteria resided within the mouse macrophages J774A.1. More important, strong expression of reporters by the bacteria could be detected in mice after administration of L-arabinose. This was especially pronounced in bacteria colonizing tumours. Histology demonstrated that the bacteria had accumulated in and close to necrotic areas of tumours. Bacterial gene induction was observed in both regions. PBAD is tightly controlled also in vivo because gene E of bacteriophage PhiX174 could be introduced as inducible suicide gene. The possibility to deliberately induce genes in bacterial carriers within the host should render them extremely powerful tools for tumour therapy.


Subject(s)
Arabinose/metabolism , Gene Expression Regulation, Bacterial , Neoplasms/microbiology , Neoplasms/therapy , Salmonella typhimurium/genetics , Animals , Arabinose/administration & dosage , Bacteriolysis , Cell Line, Tumor , Genes, Reporter , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Salmonella typhimurium/metabolism , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...