Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
iScience ; 27(6): 109813, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38799578

RESUMO

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.

2.
Microb Biotechnol ; 17(5): e14461, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38758181

RESUMO

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.


Assuntos
Lipopolissacarídeos , Neoplasias , Animais , Lipopolissacarídeos/imunologia , Neoplasias/terapia , Neoplasias/imunologia , Camundongos , Salmonella/imunologia , Salmonella/genética , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Células Dendríticas/imunologia , Imunoterapia/métodos , Humanos
3.
Cell Host Microbe ; 31(10): 1574-1592, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37827116

RESUMO

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.


Assuntos
Interações entre Hospedeiro e Microrganismos , Neoplasias , Humanos , Biologia Sintética/métodos , Neoplasias/terapia
4.
Front Immunol ; 14: 1228532, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37868996

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , Criança , Ovalbumina , Camundongos Endogâmicos C57BL , Antígenos de Neoplasias/metabolismo , Vacinação , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Salmonella/genética
5.
Biotechnol Bioeng ; 120(5): 1437-1448, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36710503

RESUMO

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.


Assuntos
Núcleo Celular , Neoplasias , Núcleo Celular/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Citoplasma/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo
6.
ACS Infect Dis ; 8(5): 969-981, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35404574

RESUMO

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.


Assuntos
Mucosa Intestinal , Probióticos , Bactérias , Citocinas , Humanos , Inflamação , Salmonella , Fator de Necrose Tumoral alfa
7.
Semin Cancer Biol ; 86(Pt 2): 1163-1178, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34547442

RESUMO

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.


Assuntos
Listeria monocytogenes , Neoplasias , Humanos , Imunoterapia , Antígenos de Neoplasias , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Listeria monocytogenes/genética , Citocinas , Microambiente Tumoral/genética
8.
Nat Commun ; 12(1): 6116, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34675204

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Caspase 3/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Hepáticas/prevenção & controle , Neoplasias Pulmonares/tratamento farmacológico , Salmonella/genética , Animais , Bacteriólise , Carcinoma Hepatocelular/fisiopatologia , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sistemas de Liberação de Medicamentos/instrumentação , Feminino , Humanos , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Salmonella/fisiologia , Salmonella typhimurium
9.
ACS Appl Mater Interfaces ; 13(15): 17196-17206, 2021 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-33821607

RESUMO

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.


Assuntos
Escherichia coli/fisiologia , Movimento/efeitos dos fármacos , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Hidrogéis/química , Natação
10.
Biotechnol Bioeng ; 117(8): 2540-2555, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32396232

RESUMO

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.


Assuntos
Interleucina-8/metabolismo , Modelos Biológicos , Muco/fisiologia , Probióticos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Bactérias/metabolismo , Bactérias/patogenicidade , Células HT29 , Humanos , Inflamação , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia
11.
Int J Cancer ; 146(1): 137-149, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31093970

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Corantes Fluorescentes/metabolismo , Neoplasias/diagnóstico , Salmonella enterica/metabolismo , Animais , Contagem de Colônia Microbiana , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Salmonella enterica/isolamento & purificação , Sensibilidade e Especificidade
12.
ACS Appl Bio Mater ; 3(5): 2897-2909, 2020 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-34322659

RESUMO

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.

13.
J Immunother Cancer ; 7(1): 44, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755273

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Neoplasias/microbiologia , Salmonella enterica/genética , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Dispositivos Lab-On-A-Chip , Salmonella enterica/fisiologia
14.
J Immunother Cancer ; 6(1): 78, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-30081947

RESUMO

In this White Paper, we discuss the current state of microbial cancer therapy. This paper resulted from a meeting ('Microbial Based Cancer Therapy') at the US National Cancer Institute in the summer of 2017. Here, we define 'Microbial Therapy' to include both oncolytic viral therapy and bacterial anticancer therapy. Both of these fields exploit tumor-specific infectious microbes to treat cancer, have similar mechanisms of action, and are facing similar challenges to commercialization. We designed this paper to nucleate this growing field of microbial therapeutics and increase interactions between researchers in it and related fields. The authors of this paper include many primary researchers in this field. In this paper, we discuss the potential, status and opportunities for microbial therapy as well as strategies attempted to date and important questions that need to be addressed. The main areas that we think will have the greatest impact are immune stimulation, control of efficacy, control of delivery, and safety. There is much excitement about the potential of this field to treat currently intractable cancer. Much of the potential exists because these therapies utilize unique mechanisms of action, difficult to achieve with other biological or small molecule drugs. By better understanding and controlling these mechanisms, we will create new therapies that will become integral components of cancer care.


Assuntos
Bactérias , Terapia Biológica/métodos , Vetores Genéticos , Neoplasias/prevenção & controle , Neoplasias/terapia , Vírus , Animais , Bactérias/genética , Terapia Biológica/normas , Terapia Biológica/tendências , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Estudos Clínicos como Assunto , Terapia Combinada , Avaliação Pré-Clínica de Medicamentos , Engenharia Genética , Vetores Genéticos/genética , Humanos , Neoplasias/etiologia , Terapia Viral Oncolítica , Resultado do Tratamento , Vírus/genética
15.
J Theor Biol ; 438: 21-33, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29066114

RESUMO

A method to predict the effect of tissue transport on the scheduling of chemotherapeutic treatment could increase efficacy. Many drugs with desirable pharmacokinetic properties fail in vivo due to poor transport through tissue. To predict the effect of treatment schedule on drug efficacy we developed an in silico method that integrates diffusion through tissue and cell binding into a pharmacokinetic model. The model was evaluated with an array of theoretical drugs that had different rates of diffusivity, binding, and clearance. The efficacy of each drug, quantified as the fraction of cells killed, was calculated for twenty dosage schedules. Simulations showed that efficacy strongly depended on tissue transport, with a range of 0.00 to 99.99%, despite each drug having equal plasma areas under the curve (AUC). For most drugs, schedules that increased exposure also increased efficacy. Drugs with fast clearance benefited the most from increasing the number of doses and this was most effective for those with intermediary binding. All drugs with slow diffusivity were ineffective. For a subset of drugs, increasing the number of doses decreased efficacy. This phenomenon was unexpected because, when considering uptake into tissue, sustained plasma levels from multiple doses are generally assumed to be more effective. This counterintuitive decrease in efficacy was caused by drug retention within tumor tissue. These results established a set of rules that suggests how transport parameters affect the efficacy of drugs at different schedules. The two most predominant rules are (1) multiple doses improve efficacy for drugs with fast clearance, fast diffusivity and low to intermediate cell binding; and (2) one dose is most effective for drugs with slow clearance, slow diffusivity or strong cell binding. Understanding the role of tissue transport when determining drug treatment schedules would improve the outcome of preclinical animal experiments and early clinical trials.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Antineoplásicos/classificação , Transporte Biológico/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Esquema de Medicação , Meia-Vida , Modelos Biológicos , Resultado do Tratamento
16.
Biotechnol Bioeng ; 113(11): 2474-84, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27159665

RESUMO

Tumor heterogeneity makes cancer difficult to treat. Many small molecule cancer drugs target rapidly dividing cells on the periphery of tumors but have difficulty in penetrating deep into tumors and are ineffective at treating entire tumors. Targeting both rapidly dividing and slower growing regions of tumors is essential to effectively treat cancer. A cancer drug carrier that penetrates deep into tumors and identifies metabolically activity could supply treatment to those areas based on the local microenvironment. We hypothesized that glucose sensing bacteria could identify sugar gradients in solid tumors. To test this hypothesis, a genetic circuit was designed to trigger expression of a green fluorescent protein (GFP) reporter through the chemotaxis-osmoporin fusion protein, Trz1, a receptor for sensing glucose and ribose sugars. E. coli equipped with the Trz1-GFP expression system, were administered to an in vitro model of a continuously perfused tumor tissue that mimics systemic delivery and clearance of bacteria through a blood vessel adjacent to a solid tumor. The level of GFP expressed, per bacterium, was time independent and indicated the glucose concentration as a function of penetration depth within the microfluidic tumors. The measured glucose concentration, correlated (P-value = 2.6 × 10(-5) ) with tumor cell viability as a function of depth. Mathematical analysis predicted drug delivery by glucose-sensing bacteria would eliminate a higher percentage of the viable tumor cell population than a systemically administered drug. Glucose-sensing bacteria could deliver cancer therapies with increased drug penetration and nutrient-dependent dosing to continuously treat viable regions of cancer tissue that have a higher prevalence for metastatic dissemination. Biotechnol. Bioeng. 2016;113: 2474-2484. © 2016 Wiley Periodicals, Inc.


Assuntos
Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/microbiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Engenharia Genética/métodos , Glucose/metabolismo , Vacinas Bacterianas , Humanos , Análise Espaço-Temporal
17.
Methods Mol Biol ; 1409: 35-48, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26846800

RESUMO

Microfluidic devices enable precise quantification of the interactions between anti-cancer bacteria and tumor tissue. Direct observation of bacterial movement and gene expression in tissue is difficult with either monolayers of cells or tumor-bearing mice. Quantification of these interactions is necessary to understand the inherent mechanisms of bacterial targeting and to develop modified organisms with enhanced therapeutic properties. Here we describe the procedures for designing, printing, and assembling microfluidic tumor-on-a-chip devices. We also describe the procedures for inserting three-dimensional tumor-cell masses, exposure to bacteria, and analyzing the resultant images.


Assuntos
Bactérias/metabolismo , Microfluídica/instrumentação , Microfluídica/métodos , Neoplasias/metabolismo , Técnicas de Cultura de Tecidos , Animais , Dispositivos Lab-On-A-Chip , Camundongos , Neoplasias/patologia , Neoplasias/terapia , Esferoides Celulares , Imagem com Lapso de Tempo , Células Tumorais Cultivadas
18.
Biotechnol Bioeng ; 112(11): 2397-405, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25976712

RESUMO

Motile bacteria can overcome the transport limitations that hinder many cancer therapies. Active bacteria can penetrate through tissue to deliver treatment to resistant tumor regions. Bacterial therapy has had limited success, however, because this motility is heterogeneous, and within a population many individuals are non-motile. In human trials, heterogeneity led to poor dispersion and incomplete tumor colonization. To address these problems, a swarm-plate selection method was developed to increase swimming velocity. Video microscopy was used to measure the velocity distribution of selected bacteria and a microfluidic tumor-on-a-chip device was used to measure penetration through tumor cell masses. Selection on swarm plates increased average velocity fourfold, from 4.9 to 18.7 µm/s (P < 0.05) and decreased the number of non-motile individuals from 51% to 3% (P < 0.05). The selected phenotype was both robust and stable. Repeating the selection process consistently increased velocity and eliminated non-motile individuals. When selected strains were cryopreserved and subcultured for 30.1 doublings, the high-motility phenotype was preserved. In the microfluidic device, selected Salmonella penetrated deeper into cell masses than unselected controls. By 10 h after inoculation, control bacteria accumulated in the front 30% of cell masses, closest to the flow channel. In contrast, selected Salmonella accumulated in the back 30% of cell masses, farthest from the channel. Selection increased the average penetration distance from 150 to 400 µm (P < 0.05). This technique provides a simple and rapid method to generate high-motility Salmonella that has increased penetration and potential for greater tumor dispersion and clinical efficacy.


Assuntos
Locomoção , Neoplasias/microbiologia , Salmonella/fisiologia , Técnicas Bacteriológicas , Terapia Biológica/métodos , Humanos , Dispositivos Lab-On-A-Chip , Microscopia de Vídeo , Modelos Biológicos , Neoplasias/terapia , Salmonella/isolamento & purificação , Seleção Genética
19.
Ther Deliv ; 6(3): 385-99, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25853312

RESUMO

Bacteria are perfect vessels for targeted cancer therapy. Conventional chemotherapy is limited by passive diffusion, and systemic administration causes severe side effects. Bacteria can overcome these obstacles by delivering therapeutic proteins specifically to tumors. Bacteria have been modified to produce proteins that directly kill cells, induce apoptosis via signaling pathways, and stimulate the immune system. These three modes of bacterial treatment have all been shown to reduce tumor growth in animal models. Bacteria have also been designed to convert nontoxic prodrugs to active therapeutic compounds. The ease of genetic manipulation enables creation of arrays of bacteria that release many new protein drugs. This versatility will allow targeting of multiple cancer pathways and will establish a platform for individualized cancer medicine.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/uso terapêutico , Terapia Biológica/métodos , Neoplasias/terapia , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/uso terapêutico , Humanos , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico
20.
Integr Biol (Camb) ; 7(4): 423-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25737274

RESUMO

Current tomographic methods of cancer detection have limited sensitivity and are unable to detect malignant masses smaller than half a centimeter in diameter. Mortality from tumor recurrence and metastatic disease would be reduced if small lesions could be detected earlier. To overcome this limitation, we created a detection system that combines the specificity of tumor-targeting bacteria with the sensitivity of a synthetic biomarker. Bacteria, specifically Salmonella, preferentially accumulate in tumors and microscopic metastases as small as five cell layers thick. To create tumor detecting bacteria, an attenuated strain of Salmonella was engineered to express and release the fluorescent protein ZsGreen. A single-layer antibody method was developed to measure low concentrations of ZsGreen. Engineered bacteria were administered to a microfluidic tumor-on-a-chip device to measure protein production. In culture, half of produced ZsGreen was released by viable bacteria at a rate of 87.6 fg bacterium(-1) h(-1). Single-layer antibody dots were able to detect bacterially produced ZsGreen at concentrations down to 4.5 ng ml(-1). Bacteria colonized in 0.12 mm(3) of tumor tissue in the microfluidic device released ZsGreen at a rate of 23.9 µg h(-1). This release demonstrates that ZsGreen readily diffuses through tissue and accumulates at detectable concentrations. Based on a mathematical pharmacokinetic model, the measured rate of release would enable detection of 0.043 mm(3) tumor masses, which is 2600 times smaller than the current limit of tomographic techniques. Tumor-detecting bacteria would provide a sensitive, minimally invasive method to detect tumor recurrence, monitor treatment efficacy, and identify the onset of metastatic disease.


Assuntos
Proteínas de Fluorescência Verde/química , Microscopia de Fluorescência/métodos , Neoplasias Experimentais/química , Neoplasias Experimentais/patologia , Salmonella/fisiologia , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Engenharia Genética/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Neoplasias Experimentais/microbiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , Salmonella/genética , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...