Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Microb Biotechnol ; 17(5): e14461, 2024 May.
Article in English | MEDLINE | ID: mdl-38758181

ABSTRACT

Immunotherapies have revolutionized cancer treatment. These treatments rely on immune cell activation in tumours, which limits the number of patients that respond. Inflammatory molecules, like lipopolysaccharides (LPS), can activate innate immune cells, which convert tumour microenvironments from cold to hot, and increase therapeutic efficacy. However, systemic delivery of lipopolysaccharides (LPS) can induce cytokine storm. In this work, we developed immune-controlling Salmonella (ICS) that only produce LPS in tumours after colonization and systemic clearance. We tuned the expression of msbB, which controls production of immunogenic LPS, by optimizing its ribosomal binding sites and protein degradation tags. This genetic system induced a controllable inflammatory response and increased dendritic cell cross-presentation in vitro. The strong off state did not induce TNFα production and prevented adverse events when injected into mice. The accumulation of ICS in tumours after intravenous injection focused immune responses specifically to tumours. Tumour-specific expression of msbB increased infiltration of immune cells, activated monocytes and neutrophils, increased tumour levels of IL-6, and activated CD8 T cells in draining lymph nodes. These immune responses reduced tumour growth and increased mouse survival. By increasing the efficacy of bacterial anti-cancer therapy, localized production of LPS could provide increased options to patients with immune-resistant cancers.


Subject(s)
Lipopolysaccharides , Neoplasms , Animals , Lipopolysaccharides/immunology , Neoplasms/therapy , Neoplasms/immunology , Mice , Salmonella/immunology , Salmonella/genetics , Mice, Inbred C57BL , Disease Models, Animal , Dendritic Cells/immunology , Immunotherapy/methods , Humans
2.
iScience ; 27(6): 109813, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38799578

ABSTRACT

As therapies, oncolytic viruses regress tumors and have the potential to induce antitumor immune responses that clear hard-to-treat and late-stage cancers. Despite this promise, clearance from the blood prevents treatment of internal solid tumors. To address this issue, we developed virus-delivering Salmonella (VDS) to carry oncolytic viruses into cancer cells. The VDS strain contains the PsseJ-lysE delivery circuit and has deletions in four homologous recombination genes (ΔrecB, ΔsbcB, ΔsbcCD, and ΔrecF) to preserve essential hairpins in the viral genome required for replication and infectivity. VDS delivered the genome for minute virus of mice (MVMp) to multiple cancers, including breast, pancreatic, and osteosarcoma. Viral delivery produced functional viral particles that are cytotoxic and infective to neighboring cells. The release of mature virions initiated new rounds of infection and amplified the infection. Using Salmonella for delivery will circumvent the limitations of oncolytic viruses and will provide a new therapy for many cancers.

3.
J Avian Med Surg ; 37(3): 217-225, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37962315

ABSTRACT

Nutritional support in malnourished animals is an essential aspect of wildlife rehabilitation; this support is especially relevant when providing lifesaving nutrition to endangered species such as the African penguin (Spheniscus demersus). This study investigated the short-term effects of a commercially available, semi-elemental, critical care diet compared with a hand-made fish formula. Twenty-one African penguin chicks were selected on admission to the Southern African Foundation for the Conservation of Costal Birds in Cape Town, South Africa, in November 2015. Initial assessment included body weight, a full clinical exam, white blood cell count, packed cell volume, and total plasma protein. Ten animals received the commercial critical care diet, whereas a control group of 11 animals were fed hand-made formula for the 2-week study period. All animals were weighed daily and blood sampling was repeated after 14 days. The median weight of both groups increased significantly over 14 days (critical care diet χ2 = 10.1, P = 0.002; control χ2 = 7.4, P = 0.006). The difference was not significant between the groups for start weight (χ2 = 0.1, P = 0.725) or end weight (χ2 = 0, P = 1.000) and was not significantly different in the change over time for either absolute numbers (χ2 = 1.7, P = 0.193) or percent gain (χ2 = 0.8, P = 0.36). The values for packed cell volume, total plasma protein, and white blood cell count increased in all animals after the 14-day study period was complete. On the basis of the results of this study, it was determined that the differing diets led to similar weight gain.


Subject(s)
Spheniscidae , Animals , South Africa , Animals, Wild , Endangered Species , Chickens , Blood Proteins
4.
Int J Pharm ; 648: 123568, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37925042

ABSTRACT

Subunit vaccines that have weak immunogenic activity require adjuvant systems for enhancedcellular and long-acting humoral immune responses. Both lipid-based and polymeric-based particulate adjuvants have been widely investigated to induce the desired immune responses against the subunit vaccines. The adjuvant efficacy of these particulate adjuvants depends upon their physicochemical properties such as particle size, surface charge, shape and their composition. Previously, we showed in vitro effect of adjuvant systems based on combination of chitosan and Salmonella Typhi porins in microparticle or nanoparticle form, which were spherical with positive surface charge. In the present study, we have further developed an adjuvant system based on combination of porins with liposomes (cationic and neutral) and investigated the adjuvant effect of both the liposomal and polymeric systems in BALB/c mice using a model antigen, ovalbumin. Humoral immune responses were determined following priming and booster dose at 15-day intervals. In overall, IgM and IgG levels were induced in the presence of both the liposomal and polymeric adjuvant systems indicating the positive impact of combination with porins. The highest IgM levels were obtained on Day 8, and liposomal adjuvant systems were found to elicit significantly higher IgM levels compared to polymeric systems. IgG levels were increased significantly after booster, particularly more profound with the micro-sized polymeric system when compared to cationic liposomal system with nano-size. Our results demonstrated that the developed particulate systems are promising both as an adjuvant and delivery system, providing enhanced immune responses against subunit antigens, and have the potential for long-term protection.


Subject(s)
Liposomes , Salmonella typhi , Mice , Animals , Liposomes/chemistry , Porins , Adjuvants, Immunologic/chemistry , Adjuvants, Pharmaceutic , Antigens , Vaccines, Subunit , Immunoglobulin G , Immunoglobulin M
5.
Front Immunol ; 14: 1228532, 2023.
Article in English | MEDLINE | ID: mdl-37868996

ABSTRACT

Introduction: Immunotherapies have shown great promise, but are not effective for all tumors types and are effective in less than 3% of patients with pancreatic ductal adenocarcinomas (PDAC). To make an immune treatment that is effective for more cancer patients and those with PDAC specifically, we genetically engineered Salmonella to deliver exogenous antigens directly into the cytoplasm of tumor cells. We hypothesized that intracellular delivery of an exogenous immunization antigen would activate antigen-specific CD8 T cells and reduce tumors in immunized mice. Methods: To test this hypothesis, we administered intracellular delivering (ID) Salmonella that deliver ovalbumin as a model antigen into tumor-bearing, ovalbumin-vaccinated mice. ID Salmonella delivers antigens by autonomously lysing in cells after the induction of cell invasion. Results: We showed that the delivered ovalbumin disperses throughout the cytoplasm of cells in culture and in tumors. This delivery into the cytoplasm is essential for antigen cross-presentation. We showed that co-culture of ovalbumin-recipient cancer cells with ovalbumin-specific CD8 T cells triggered a cytotoxic T cell response. After the adoptive transfer of OT-I CD8 T cells, intracellular delivery of ovalbumin reduced tumor growth and eliminated tumors. This effect was dependent on the presence of the ovalbumin-specific T cells. Following vaccination with the exogenous antigen in mice, intracellular delivery of the antigen cleared 43% of established KPC pancreatic tumors, increased survival, and prevented tumor re-implantation. Discussion: This response in the immunosuppressive KPC model demonstrates the potential to treat tumors that do not respond to checkpoint inhibitors, and the response to re-challenge indicates that new immunity was established against intrinsic tumor antigens. In the clinic, ID Salmonella could be used to deliver a protein antigen from a childhood immunization to refocus pre-existing T cell immunity against tumors. As an off-the-shelf immunotherapy, this bacterial system has the potential to be effective in a broad range of cancer patients.


Subject(s)
CD8-Positive T-Lymphocytes , Pancreatic Neoplasms , Humans , Mice , Animals , Child , Ovalbumin , Mice, Inbred C57BL , Antigens, Neoplasm/metabolism , Vaccination , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/metabolism , Salmonella/genetics
6.
Cell Host Microbe ; 31(10): 1574-1592, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37827116

ABSTRACT

Many systemically administered cancer therapies exhibit dose-limiting toxicities that reduce their effectiveness. To increase efficacy, bacterial delivery platforms have been developed that improve safety and prolong treatment. Bacteria are a unique class of therapy that selectively colonizes most solid tumors. As delivery vehicles, bacteria have been genetically modified to express a range of therapies that match multiple cancer indications. In this review, we describe a modular "build-a-bug" method that focuses on five design characteristics: bacterial strain (chassis), therapeutic compound, delivery method, immune-modulating features, and genetic control circuits. We emphasize how fundamental research into gut microbe pathogenesis has created safe bacterial therapies, some of which have entered clinical trials. The genomes of gut microbes are fertile grounds for discovery of components to improve delivery and modulate host immune responses. Future work coupling these delivery vehicles with insights from gut microbes could lead to the next generation of microbial cancer therapy.


Subject(s)
Host Microbial Interactions , Neoplasms , Humans , Synthetic Biology/methods , Neoplasms/therapy
7.
Biotechnol Bioeng ; 120(5): 1437-1448, 2023 05.
Article in English | MEDLINE | ID: mdl-36710503

ABSTRACT

Targeting nucleic targets with therapeutic proteins would enhance the treatment of hard-to-treat cancers. However, exogenous proteins are excluded from the nucleus by both the cellular and nuclear membranes. We have recently developed Salmonella that deliver active proteins into the cytoplasm of cancer cells. Here, we hypothesized that bacterially delivered proteins accumulate within nuclei, nuclear localization sequences (NLSs) increase delivery, and bacterially delivered proteins kill cancer cells. To test this hypothesis, we developed intranuclear delivering (IND) Salmonella and quantified the delivery of three model proteins. IND Salmonella delivered both ovalbumin and green fluorescent protein to nuclei of MCF7 cancer cells. The amount of protein in nuclei was linearly dependent on the amount delivered to the cytoplasm. The addition of a NLSs increased both the amount of protein in each nucleus and the number of nuclei that received protein. Delivery of Omomyc, a protein inhibitor of the nuclear transcript factor, Myc, altered cell physiology, and significantly induced cell death. These results show that IND Salmonella deliver functional proteins to the nucleus of cancerous cells. Extending this method to other transcription factors will increase the number of accessible targets for cancer therapy.


Subject(s)
Cell Nucleus , Neoplasms , Cell Nucleus/metabolism , Green Fluorescent Proteins/metabolism , Cytoplasm/metabolism , Transcription Factors/metabolism , Neoplasms/therapy , Neoplasms/metabolism
8.
ACS Infect Dis ; 8(5): 969-981, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35404574

ABSTRACT

The balance of microbial species in the intestine must be maintained to prevent inflammation and disease. Healthy bacteria suppress infection by pathogens and prevent disorders such as inflammatory bowel diseases (IBDs). The role of mucus in the relation between pathogens and the intestinal microbiota is poorly understood. Here, we hypothesized that healthy bacteria inhibit infection by preventing pathogens from penetrating the mucus layer and that microbial imbalance leads to inflammation by promoting the penetration of the mucosal barrier. We tested this hypothesis with an in vitro model that contains mucus, an epithelial cell layer, and resident immune cells. We found that, unlike probiotic VSL#3 bacteria, Salmonella penetrated the mucosal layers and induced the production of interleukin-8 (IL-8) and tumor necrosis factor (TNF)-α. At ratios greater than 104:1, probiotic bacteria suppressed the growth and penetration of Salmonella and reduced the production of inflammatory cytokines. Counterintuitively, low densities of healthy bacteria increased both pathogen penetration and cytokine production. In all cases, mucus increased Salmonella penetration and the production of cytokines. These results suggest that mucus lessens the protective effect of probiotic bacteria by promoting barrier penetration. In this model, a more imbalanced microbial population caused infection and inflammation by selecting pathogens that are more invasive and immunogenic. Combined, the results suggest that the depletion of commensal bacteria or an insufficient dosage of probiotics could worsen an infection and cause increased inflammation. A better understanding of the interactions between pathogens, healthy microbes, and the mucosal barrier will improve the treatment of infections and inflammatory diseases.


Subject(s)
Intestinal Mucosa , Probiotics , Bacteria , Cytokines , Humans , Inflammation , Salmonella , Tumor Necrosis Factor-alpha
9.
Semin Cancer Biol ; 86(Pt 2): 1163-1178, 2022 11.
Article in English | MEDLINE | ID: mdl-34547442

ABSTRACT

Engineered bacterial therapies that target the tumor immune landscape offer a new class of cancer immunotherapy. Salmonella enterica and Listeria monocytogenes are two species of bacteria that have been engineered to specifically target tumors and serve as delivery vessels for immunotherapies. Therapeutic bacteria have been engineered to deliver cytokines, gene silencing shRNA, and tumor associated antigens that increase immune activation. Bacterial therapies stimulate both the innate and adaptive immune system, change the immune dynamics of the tumor microenvironment, and offer unique strategies for targeting tumors. Bacteria have innate adjuvant properties, which enable both the delivered molecules and the bacteria themselves to stimulate immune responses. Bacterial immunotherapies that deliver cytokines and tumor-associated antigens have demonstrated clinical efficacy. Harnessing the diverse set of mechanisms that Salmonella and Listeria use to alter the tumor-immune landscape has the potential to generate many new and effective immunotherapies.


Subject(s)
Listeria monocytogenes , Neoplasms , Humans , Immunotherapy , Antigens, Neoplasm , Neoplasms/therapy , Neoplasms/drug therapy , Listeria monocytogenes/genetics , Cytokines , Tumor Microenvironment/genetics
10.
Nanomedicine ; 40: 102506, 2022 02.
Article in English | MEDLINE | ID: mdl-34875352

ABSTRACT

Oncolytic viruses (OVs) selectively replicate in and destroy cancer cells resulting in anti-tumor immunity. However, clinical use remains a challenge because of virus clearance upon intravenous delivery. OV packaging using a nanomedicine approach could overcome this. Here we encapsulate an oncolytic adenovirus (Ad[I/PPT-E1A]) into CCL2-coated liposomes in order to exploit recruitment of CCR2-expressing circulating monocytes into tumors. We demonstrate successful encapsulation of Ad[I/PPT-E1A] into CCL2-coated liposomes that were preferentially taken up by CCR2-expressing monocytes. No complex-related toxicities were observed following incubation with prostate tumor cells and the encapsulation did not affect virus oncolytic activity in vitro. Furthermore, intravenous administration of our nanomedicine resulted in a significant reduction in tumor size and pulmonary metastasis in prostate cancer-bearing mice whereby a 1000-fold less virus was needed compared to Ad[I/PPT-E1A] alone. Taken together our data provide an opportunity to target OVs via circulation to inaccessible tumors using liposome-assisted drug delivery.


Subject(s)
Adenoviridae , Oncolytic Virotherapy , Adenoviridae/genetics , Animals , Cell Line, Tumor , Chemokine CCL2/genetics , Genetic Vectors , Humans , Liposomes , Male , Mice , Monocytes , Oncolytic Virotherapy/methods
11.
Nat Commun ; 12(1): 6116, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675204

ABSTRACT

Critical cancer pathways often cannot be targeted because of limited efficiency crossing cell membranes. Here we report the development of a Salmonella-based intracellular delivery system to address this challenge. We engineer genetic circuits that (1) activate the regulator flhDC to drive invasion and (2) induce lysis to release proteins into tumor cells. Released protein drugs diffuse from Salmonella containing vacuoles into the cellular cytoplasm where they interact with their therapeutic targets. Control of invasion with flhDC increases delivery over 500 times. The autonomous triggering of lysis after invasion makes the platform self-limiting and prevents drug release in healthy organs. Bacterial delivery of constitutively active caspase-3 blocks the growth of hepatocellular carcinoma and lung metastases, and increases survival in mice. This success in targeted killing of cancer cells provides critical evidence that this approach will be applicable to a wide range of protein drugs for the treatment of solid tumors.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Caspase 3/administration & dosage , Drug Delivery Systems/methods , Liver Neoplasms/prevention & control , Lung Neoplasms/drug therapy , Salmonella/genetics , Animals , Bacteriolysis , Carcinoma, Hepatocellular/physiopathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Delivery Systems/instrumentation , Female , Humans , Liver Neoplasms/secondary , Male , Mice , Salmonella/physiology , Salmonella typhimurium
12.
ACS Appl Mater Interfaces ; 13(15): 17196-17206, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33821607

ABSTRACT

Bacterial swimming in flow near surfaces is critical to the spread of infection and device colonization. Understanding how material properties affect flagella- and motility-dependent bacteria-surface interactions is a first step in designing new medical devices that mitigate the risk of infection. We report that, on biomaterial coatings such as polyethylene glycol (PEG) hydrogels and end-tethered layers that prevent adhesive bacteria accumulation, the coating mechanics and hydration control the near-surface travel and dynamic surface contact of E. coli cells in gentle shear flow (order 10 s-1). Along relatively stiff (order 1 MPa) PEG hydrogels or end-tethered layers of PEG chains of similar polymer correlation length, run-and-tumble E. coli travel nanometrically close to the coating's surface in the flow direction in distinguishable runs or "engagements" that persist for several seconds, after which cells leave the interface. The duration of these engagements was greater along stiff hydrogels and end-tethered layers compared with softer, more-hydrated hydrogels. Swimming cells that left stiff hydrogels or end-tethered layers proceeded out to distances of a few microns and then returned to engage the surface again and again, while cells engaging the soft hydrogel tended not to return after leaving. As a result of differences in the duration of engagements and tendency to return to stiff hydrogel and end-tethered layers, swimming E. coli experienced 3 times the integrated dynamic surface contact with stiff coatings compared with softer hydrogels. The striking similarity of swimming behaviors near 16-nm-thick end-tethered layers and 100-µm-thick stiff hydrogels argues that only the outermost several nanometers of a highly hydrated coating influence cell travel. The range of material stiffnesses, cell-surface distance during travel, and time scales of travel compared with run-and-tumble time scales suggests the influence of the coating derives from its interactions with flagella and its potential to alter flagellar bundling. Given that restriction of flagellar rotation is known to trigger increased virulence, bacteria influenced by surfaces in one region may become predisposed to form a biofilm downstream.


Subject(s)
Escherichia coli/physiology , Movement/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Bacterial Adhesion/drug effects , Hydrogels/chemistry , Swimming
13.
Biotechnol Bioeng ; 117(8): 2540-2555, 2020 08.
Article in English | MEDLINE | ID: mdl-32396232

ABSTRACT

The mucosal barrier in combination with innate immune system are the first line of defense against luminal bacteria at the intestinal mucosa. Dysfunction of the mucus layer and bacterial infiltration are linked to tissue inflammation and disease. To study host-bacterial interactions at the mucosal interface, we created an experimental model that contains luminal space, a mucus layer, an epithelial layer, and suspended immune cells. Reconstituted porcine small intestinal mucus formed an 880 ± 230 µm thick gel layer and had a porous structure. In the presence of mucus, sevenfold less probiotic and nonmotile VSL#3 bacteria transmigrated across the epithelial barrier compared to no mucus. The higher bacterial transmigration caused immune cell differentiation and increased the concentration of interleukin-8 (IL-8) and tumor necrosis factor-alpha (TNF-α; p < .01). Surprisingly, the mucus layer increased transmigration of pathogenic Salmonella and increased secretion of TNF-α and IL-8 (p < .05). Nonmotile, flagella knockout Salmonella had lower transmigration and caused lower IL-8 and TNF-α secretion (p < .05). These results demonstrate that motility enables pathogenic bacteria to cross the mucus and epithelial layers, which could lead to infection. Using an in vitro coculture platform to understand the interactions of bacteria with the intestinal mucosa has the potential to improve the treatment of intestinal diseases.


Subject(s)
Interleukin-8/metabolism , Models, Biological , Mucus/physiology , Probiotics/metabolism , Tumor Necrosis Factor-alpha/metabolism , Bacteria/metabolism , Bacteria/pathogenicity , HT29 Cells , Humans , Inflammation , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology
14.
Int J Pharm ; 582: 119266, 2020 May 30.
Article in English | MEDLINE | ID: mdl-32251694

ABSTRACT

Nanomedicines are well recognised for their ability to improve therapeutic outcomes. Yet, due to their complexity, nanomedicines are challenging and costly to produce using traditional manufacturing methods. For nanomedicines to be widely exploited, new manufacturing technologies must be adopted to reduce development costs and provide a consistent product. Within this study, we investigate microfluidic manufacture of nanomedicines. Using protein-loaded liposomes as a case study, we manufacture liposomes with tightly defined physico-chemical attributes (size, PDI, protein loading and release) from small-scale (1 mL) through to GMP volume production (200 mL/min). To achieve this, we investigate two different laminar flow microfluidic cartridge designs (based on a staggered herringbone design and a novel toroidal mixer design); for the first time we demonstrate the use of a new microfluidic cartridge design which delivers seamless scale-up production from bench-scale (12 mL/min) through GMP production requirements of over 20 L/h using the same standardised normal operating parameters. We also outline the application of tangential flow filtration for down-stream processing and high product yield. This work confirms that defined liposome products can be manufactured rapidly and reproducibly using a scale-independent production process, thereby de-risking the journey from bench to approved product.


Subject(s)
Doxorubicin/chemistry , Lipids/chemistry , Microfluidics , Nanomedicine , Nanoparticles , Ovalbumin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/standards , Drug Compounding , Drug Liberation , Lipids/standards , Liposomes , Microfluidics/instrumentation , Microfluidics/standards , Nanomedicine/instrumentation , Nanomedicine/standards , Ovalbumin/administration & dosage , Ovalbumin/standards , Particle Size , Quality Control , Solubility
15.
ACS Appl Bio Mater ; 3(5): 2897-2909, 2020 May 18.
Article in English | MEDLINE | ID: mdl-34322659

ABSTRACT

Dysfunction of the intestinal mucus barrier causes disorders such as ulcerative colitis and Crohn's disease. The function of this essential barrier may be affected by the periodically changing luminal environment. We hypothesized that the pH and ion concentration in mucus control its porosity, molecular permeability, and the penetration of microbes. To test this hypothesis, we developed a scalable method to extract porcine small intestinal mucus (PSIM). The aggregation and porosity of PSIM were determined using rheometry, spectrophotometry, and microscopy. Aggregation of PSIM at low pH increased both the elastic (G') and viscous (G″) moduli, and it slowed the transmigration of pathogenic Salmonella. Molecular transport was dependent on ion concentration. At moderate concentrations, many microscopic aggregates (2-5 µm in diameter) impeded diffusion. At higher concentrations, PSIM formed aggregate islands, increasing both porosity and diffusion. This in vitro model could lead to a better understanding of mucus barrier functions and improve the treatment of intestinal diseases.

16.
Int J Cancer ; 146(1): 137-149, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31093970

ABSTRACT

Combining the specificity of tumor-targeting bacteria with the sensitivity of biomarker detection would create a screening method able to detect small tumors and metastases. To create this system, we genetically modified an attenuated strain of Salmonella enterica to release a recombinant fluorescent biomarker (or fluoromarker). Salmonella expressing ZsGreen were intravenously administered to tumor-bearing mice and fluoromarker production was induced after 48 hr. The quantities and locations of bacteria and ZsGreen were measured in tumors, livers and spleens by immunofluorescence, and the plasma concentration of ZsGreen was measured using single-layer ELISA. In the plasma, the ZsGreen concentration was in the range of 0.5-1.5 ng/ml and was dependent on tumor mass (with a proportion of 0.81 ± 0.32 ng·ml-1 ·g-1 ). No adverse reaction to ZsGreen or bacteria was observed in any mice. ZsGreen was released at an average rate of 4.3 fg·CFU-1 ·hr-1 and cleared from the plasma with a rate constant of 0.259 hr-1 . ZsGreen production was highest in viable tissue (7.6 fg·CFU-1 ·hr-1 ) and lowest in necrotic tissue (0.47 fg·CFU-1 ·hr-1 ). The mass transfer rate constant from tumor to blood was 0.0125 hr-1 . Based on these measurements, this system has the capability to detect tumors as small as 0.12 g. These results demonstrate four essential mechanisms of this method: (i) preferential tumor colonization by bacteria, (ii) fluoromarker release in vivo, (iii) fluoromarker transport through tumor tissue and (iv) slow enough systemic clearance to enable measurement. This bacteria-based blood test would be minimally invasive and has the potential to identify previously undetectable microscopic tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Fluorescent Dyes/metabolism , Neoplasms/diagnosis , Salmonella enterica/metabolism , Animals , Colony Count, Microbial , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Salmonella enterica/isolation & purification , Sensitivity and Specificity
17.
Vet Surg ; 49(1): 19-21, 2020 01.
Article in English | MEDLINE | ID: mdl-31876013
18.
Int J Pharm ; 573: 118722, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31705976

ABSTRACT

Technology such as the use of microfluidics to generate liposomes has been well researched, yet the stabilisation of liposomal formulations is a major challenge to their greater implementation. To the best of our knowledge, this is the first study investigating the use of 96 well plates to freeze-dry ovalbumin (OVA) loaded neutral (DMPC:Chol and DSPC:Chol), anionic (DSPC:Chol:PS) and cationic (DSPC:Chol:DOTAP) liposomes. Through the use of high throughput screening, a freeze drying cycle was optimised; ramp freezing from from 4 °C to -45 °C, followed by primary drying at -30 °C and secondary drying at 30 °C under a vacuum of 0.1 mBar. These parameters maintained liposome physicochemical properties, with the liposomes remaining below 100 nm and were homogenous (polydispersity index of less than 0.2 post rehydration). Minimal leakage of the OVA protein was observed, with almost 100% OVA remaining encapsulated post rehydration of the formulations. Here we have identified a simple method that allows for the rapid screening and freeze-drying of a range of liposomal formulations.


Subject(s)
Drug Delivery Systems , Microfluidics , Ovalbumin/administration & dosage , Proteins/administration & dosage , Cholesterol/chemistry , Dimyristoylphosphatidylcholine/chemistry , Fatty Acids, Monounsaturated/chemistry , Freeze Drying , High-Throughput Screening Assays , Liposomes , Ovalbumin/chemistry , Phosphatidylcholines/chemistry , Proteins/chemistry , Quaternary Ammonium Compounds/chemistry , Technology, Pharmaceutical
19.
Pharmaceutics ; 11(12)2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31817217

ABSTRACT

The aim of this work was to assess the impact of solvent selection on the microfluidic production of liposomes. To achieve this, liposomes were manufactured using small-scale and bench-scale microfluidics systems using three aqueous miscible solvents (methanol, ethanol or isopropanol, alone or in combination). Liposomes composed of different lipid compositions were manufactured using these different solvents and characterised to investigate the influence of solvents on liposome attributes. Our studies demonstrate that solvent selection is a key consideration during the microfluidics manufacturing process, not only when considering lipid solubility but also with regard to the resultant liposome critical quality attributes. In general, reducing the polarity of the solvent (from methanol to isopropanol) increased the liposome particle size without impacting liposome short-term stability or release characteristics. Furthermore, solvent combinations such as methanol/isopropanol mixtures can be used to modify solvent polarity and the resultant liposome particle size. However, the impact of solvent choice on the liposome product is also influenced by the liposome formulation; liposomes containing charged lipids tended to show more sensitivity to solvent selection and formulations containing increased concentrations of cholesterol or pegylated-lipids were less influenced by the choice of solvent. Indeed, incorporation of 14 wt% or more of pegylated-lipid was shown to negate the impact of solvent selection.

20.
J Immunother Cancer ; 7(1): 44, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755273

ABSTRACT

BACKGROUND: Salmonella have potential as anticancer therapeutic because of their innate tumor specificity. In clinical studies, this specificity has been hampered by heterogeneous responses. Understanding the mechanisms that control tumor colonization would enable the design of more robust therapeutic strains. Two mechanisms that could affect tumor colonization are intracellular accumulation and intratumoral motility. Both of these mechanisms have elements that are controlled by the master motility regulator flhDC. We hypothesized that 1) overexpressing flhDC in Salmonella increases intracellular bacterial accumulation in tumor cell masses, and 2) intracellular accumulation of Salmonella drives tumor colonization in vitro. METHODS: To test these hypotheses, we transformed Salmonella with genetic circuits that induce flhDC and express green fluorescent protein after intracellular invasion. The genetically modified Salmonella was perfused into an in vitro tumor-on-a-chip device. Time-lapse fluorescence microscopy was used to quantify intracellular and colonization dynamics within tumor masses. A mathematical model was used to determine how these mechanisms are related to each other. RESULTS: Overexpression of flhDC increased intracellular accumulation and tumor colonization 2.5 and 5 times more than control Salmonella, respectively (P < 0.05). Non-motile Salmonella accumulated in cancer cells 26 times less than controls (P < 0.001). Minimally invasive, ΔsipB, Salmonella colonized tumor masses 2.5 times less than controls (P < 0.05). When flhDC was selectively induced after penetration into tumor masses, Salmonella both accumulated intracellularly and colonized tumor masses 2 times more than controls (P < 0.05). Mathematical modeling of tumor colonization dynamics demonstrated that intracellular accumulation increased retention of Salmonella in tumors by effectively causing the bacteria to bind to cancer cells and preventing leakage out of the tumors. These results demonstrated that increasing intracellular bacterial density increased overall tumor colonization and that flhDC could be used to control both. CONCLUSIONS: This study demonstrates a mechanistic link between motility, intracellular accumulation and tumor colonization. Based on our results, we envision that therapeutic strains of Salmonella could use inducible flhDC to drive tumor colonization. More intratumoral bacteria would enable delivery of higher therapeutic payloads into tumors and would improve treatment efficacy.


Subject(s)
Bacterial Proteins/genetics , Neoplasms/microbiology , Salmonella enterica/genetics , Cell Line, Tumor , Drug Delivery Systems , Humans , Lab-On-A-Chip Devices , Salmonella enterica/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...