Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 35(51): e2304654, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37753928

RESUMEN

Monoclonal antibodies (mAbs) hold promise in treating Parkinson's disease (PD), although poor delivery to the brain hinders their therapeutic application. In the current study, it is demonstrated that brain-targeted liposomes (BTL) enhance the delivery of mAbs across the blood-brain-barrier (BBB) and into neurons, thereby allowing the intracellular and extracellular treatment of the PD brain. BTL are decorated with transferrin to improve brain targeting through overexpressed transferrin-receptors on the BBB during PD. BTL are loaded with SynO4, a mAb that inhibits alpha-synuclein (AS) aggregation, a pathological hallmark of PD. It is shown that 100-nm BTL cross human BBB models intact and are taken up by primary neurons. Within neurons, SynO4 is released from the nanoparticles and bound to its target, thereby reducing AS aggregation, and enhancing neuronal viability. In vivo, intravenous BTL administration results in a sevenfold increase in mAbs in brain cells, decreasing AS aggregation and neuroinflammation. Treatment with BTL also improve behavioral motor function and learning ability in mice, with a favorable safety profile. Accordingly, targeted nanotechnologies offer a valuable platform for drug delivery to treat brain neurodegeneration.


Asunto(s)
Enfermedad de Parkinson , Animales , Humanos , Ratones , alfa-Sinucleína/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Síntomas Conductuales , Encéfalo/metabolismo , Liposomas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Transferrinas
2.
Proc Natl Acad Sci U S A ; 119(38): e2207525119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095208

RESUMEN

Progress in bottom-up synthetic biology has stimulated the development of synthetic cells (SCs), autonomous protein-manufacturing particles, as dynamic biomimetics for replacing diseased natural cells and addressing medical needs. Here, we report that SCs genetically encoded to produce proangiogenic factors triggered the physiological process of neovascularization in mice. The SCs were constructed of giant lipid vesicles and were optimized to facilitate enhanced protein production. When introduced with the appropriate genetic code, the SCs synthesized a recombinant human basic fibroblast growth factor (bFGF), reaching expression levels of up to 9⋅106 protein copies per SC. In culture, the SCs induced endothelial cell proliferation, migration, tube formation, and angiogenesis-related intracellular signaling, confirming their proangiogenic activity. Integrating the SCs with bioengineered constructs bearing endothelial cells promoted the remodeling of mature vascular networks, supported by a collagen-IV basement membrane-like matrix. In vivo, prolonged local administration of the SCs in mice triggered the infiltration of blood vessels into implanted Matrigel plugs without recorded systemic immunogenicity. These findings emphasize the potential of SCs as therapeutic platforms for activating physiological processes by autonomously producing biological drugs inside the body.


Asunto(s)
Células Artificiales , Factores de Crecimiento de Fibroblastos , Neovascularización Fisiológica , Animales , Células Artificiales/trasplante , Movimiento Celular , Proliferación Celular , Colágeno Tipo IV/metabolismo , Células Endoteliales/fisiología , Factores de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Biosíntesis de Proteínas
3.
Nat Commun ; 13(1): 2328, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35484097

RESUMEN

Development of regulated cellular processes and signaling methods in synthetic cells is essential for their integration with living materials. Light is an attractive tool to achieve this, but the limited penetration depth into tissue of visible light restricts its usability for in-vivo applications. Here, we describe the design and implementation of bioluminescent intercellular and intracellular signaling mechanisms in synthetic cells, dismissing the need for an external light source. First, we engineer light generating SCs with an optimized lipid membrane and internal composition, to maximize luciferase expression levels and enable high-intensity emission. Next, we show these cells' capacity to trigger bioprocesses in natural cells by initiating asexual sporulation of dark-grown mycelial cells of the fungus Trichoderma atroviride. Finally, we demonstrate regulated transcription and membrane recruitment in synthetic cells using bioluminescent intracellular signaling with self-activating fusion proteins. These functionalities pave the way for deploying synthetic cells as embeddable microscale light sources that are capable of controlling engineered processes inside tissues.


Asunto(s)
Células Artificiales , Optogenética , Luz , Luciferasas , Optogenética/métodos , Transducción de Señal
4.
ACS Nano ; 16(4): 5246-5257, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35293714

RESUMEN

Throughout the female menstrual cycle, physiological changes occur that affect the biodistribution of nanoparticles within the reproductive system. We demonstrate a 2-fold increase in nanoparticle accumulation in murine ovaries and uterus during ovulation, compared to the nonovulatory stage, following intravenous administration. This biodistribution pattern had positive or negative effects when drug-loaded nanoparticles, sized 100 nm or smaller, were used to treat different cancers. For example, treating ovarian cancer with nanomedicines during mouse ovulation resulted in higher drug accumulation in the ovaries, improving therapeutic efficacy. Conversely, treating breast cancer during ovulation, led to reduced therapeutic efficacy, due to enhanced nanoparticle accumulation in the reproductive system rather than at the tumor site. Moreover, chemotherapeutic nanoparticles administered during ovulation increased ovarian toxicity and decreased fertility compared to the free drug. The menstrual cycle should be accounted for when designing and implementing nanomedicines for females.


Asunto(s)
Nanopartículas , Neoplasias , Femenino , Ratones , Animales , Distribución Tisular , Fertilidad , Ovulación , Genitales Femeninos
5.
Sci Adv ; 7(41): eabj5435, 2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34613777

RESUMEN

Neurons within the tumor microenvironment promote cancer progression; thus, their local targeting has potential clinical benefits. We designed PEGylated lipid nanoparticles loaded with a non-opioid analgesic, bupivacaine, to target neurons within breast cancer tumors and suppress nerve-to-cancer cross-talk. In vitro, 100-nm nanoparticles were taken up readily by primary neurons, trafficking from the neuronal body and along the axons. We demonstrate that signaling between triple-negative breast cancer cells (4T1) and neurons involves secretion of cytokines stimulating neurite outgrowth. Reciprocally, neurons stimulated 4T1 proliferation, migration, and survival through secretion of neurotransmitters. Bupivacaine curbs neurite growth and signaling with cancer cells, inhibiting cancer cell viability. In vivo, bupivacaine-loaded nanoparticles intravenously administered suppressed neurons in orthotopic triple-negative breast cancer tumors, inhibiting tumor growth and metastatic dissemination. Overall, our findings suggest that reducing nerve involvement in tumors is important for treating cancer.

6.
J Vis Exp ; (158)2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32391815

RESUMEN

The bottom-up assembly approach for construction of synthetic cells is an effective tool for isolating and investigating cellular processes in a cell mimicking environment. Furthermore, the development of cell-free expression systems has demonstrated the ability to reconstitute the protein production, transcription and translation processes (DNA→RNA→protein) in a controlled manner, harnessing synthetic biology. Here we describe a protocol for preparing a cell-free expression system, including the production of a potent bacterial lysate and encapsulating this lysate inside cholesterol-rich lipid-based giant unilamellar vesicles (GUVs) (i.e., stable liposomes), to form synthetic cells. The protocol describes the methods for preparing the components of the synthetic cells including the production of active bacterial lysates, followed by a detailed step-by-step preparation of the synthetic cells based on a water-in-oil emulsion transfer method. These facilitate the production of millions of synthetic cells in a simple and affordable manner with a high versatility for producing different types of proteins. The obtained synthetic cells can be used to investigate protein/RNA production and activity in an isolated environment, in directed evolution, and also as a controlled drug delivery platform for on-demand production of therapeutic proteins inside the body.


Asunto(s)
Células Artificiales/metabolismo , Emulsiones/química , Escherichia coli/metabolismo , Biosíntesis de Proteínas , Biología Sintética/métodos , Sistema Libre de Células/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Liposomas/química , Luciferasas/metabolismo
7.
Adv Mater ; 32(13): e1901989, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31286573

RESUMEN

Artificial intelligence (AI) and nanotechnology are two fields that are instrumental in realizing the goal of precision medicine-tailoring the best treatment for each cancer patient. Recent conversion between these two fields is enabling better patient data acquisition and improved design of nanomaterials for precision cancer medicine. Diagnostic nanomaterials are used to assemble a patient-specific disease profile, which is then leveraged, through a set of therapeutic nanotechnologies, to improve the treatment outcome. However, high intratumor and interpatient heterogeneities make the rational design of diagnostic and therapeutic platforms, and analysis of their output, extremely difficult. Integration of AI approaches can bridge this gap, using pattern analysis and classification algorithms for improved diagnostic and therapeutic accuracy. Nanomedicine design also benefits from the application of AI, by optimizing material properties according to predicted interactions with the target drug, biological fluids, immune system, vasculature, and cell membranes, all affecting therapeutic efficacy. Here, fundamental concepts in AI are described and the contributions and promise of nanotechnology coupled with AI to the future of precision cancer medicine are reviewed.


Asunto(s)
Inteligencia Artificial , Nanomedicina/métodos , Nanotecnología/métodos , Medicina de Precisión/métodos , Animales , Biología Computacional/métodos , Sistemas de Liberación de Medicamentos/métodos , Humanos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Neoplasias/diagnóstico , Neoplasias/terapia
8.
ACS Nano ; 13(10): 11008-11021, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31503443

RESUMEN

Overexpressed extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) limits drug penetration into the tumor and is associated with poor prognosis. Here, we demonstrate that a pretreatment based on a proteolytic-enzyme nanoparticle system disassembles the dense PDAC collagen stroma and increases drug penetration into the pancreatic tumor. More specifically, the collagozome, a 100 nm liposome encapsulating collagenase, was rationally designed to protect the collagenase from premature deactivation and prolonged its release rate at the target site. Collagen is the main component of the PDAC stroma, reaching 12.8 ± 2.3% vol in diseased mice pancreases, compared to 1.4 ± 0.4% in healthy mice. Upon intravenous injection of the collagozome, ∼1% of the injected dose reached the pancreas over 8 h, reducing the level of fibrotic tissue to 5.6 ± 0.8%. The collagozome pretreatment allowed increased drug penetration into the pancreas and improved PDAC treatment. PDAC tumors, pretreated with the collagozome followed by paclitaxel micelles, were 87% smaller than tumors pretreated with empty liposomes followed by paclitaxel micelles. Interestingly, degrading the ECM did not increase the number of circulating tumor cells or metastasis. This strategy holds promise for degrading the extracellular stroma in other diseases as well, such as liver fibrosis, enhancing tissue permeability before drug administration.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Colagenasas/farmacología , Nanopartículas/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colágeno/química , Colágeno/genética , Colagenasas/química , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Fibrosis/prevención & control , Humanos , Liposomas/química , Liposomas/farmacología , Ratones , Nanopartículas/uso terapéutico , Paclitaxel/química , Paclitaxel/farmacología , Páncreas/efectos de los fármacos , Páncreas/patología , Microambiente Tumoral/efectos de los fármacos
9.
J Control Release ; 307: 331-341, 2019 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-31238049

RESUMEN

Lipid nanoparticles are used widely as anticancer drug and gene delivery systems. Internalizing into the target cell is a prerequisite for the proper activity of many nanoparticulate drugs. We show here, that the lipid composition of a nanoparticle affects its ability to internalize into triple-negative breast cancer cells. The lipid headgroup had the greatest effect on enhancing cellular uptake compared to other segments of the molecule. Having a receptor-targeted headgroup induced the greatest increase in cellular uptake, followed by cationic amine headgroups, both being superior to neutral (zwitterion) phosphatidylcholine or to negatively-charged headgroups. The lipid tails also affected the magnitude of cellular uptake. Longer acyl chains facilitated greater liposomal cellular uptake compared to shorter tails, 18:0 > 16:0 > 14:0. When having the same lipid tail length, unsaturated lipids were superior to saturated ones, 18:1 > 18:0. Interestingly, liposomes composed of phospholipids having 14:0 or 12:0-carbon-long-tails, such as DMPC and DLPC, decreased cell viability in a concertation dependent manner, due to a destabilizing effect these lipids had on the cancer cell membrane. Contrarily, liposomes composed of phospholipids having longer carbon tails (16:0 and 18:0), such as DPPC and HSPC, enhanced cancer cell proliferation. This effect is attributed to the integration of the exogenous liposomal lipids into the cancer-cell membrane, supporting the proliferation process. Cholesterol is a common lipid additive in nanoscale formulations, rigidifying the membrane and stabilizing its structure. Liposomes composed of DMPC (14:0) showed increased cellular uptake when enriched with cholesterol, both by endocytosis and by fusion. Contrarily, the effect of cholesterol on HSPC (18:0) liposomal uptake was minimal. Furthermore, the concentration of nanoparticles in solution affected their cellular uptake. The higher the concentration of nanoparticles the greater the absolute number of nanoparticles taken up per cell. However, the efficiency of nanoparticle uptake, i.e. the percent of nanoparticles taken up by cells, decreased as the concentration of nanoparticles increased. This study demonstrates that tuning the lipid composition and concentration of nanoscale drug delivery systems can be leveraged to modulate their cellular uptake.


Asunto(s)
Sistemas de Liberación de Medicamentos , Lípidos/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Endocitosis , Lípidos/química , Ratones , Nanopartículas/química
10.
J Control Release ; 296: 1-13, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30615983

RESUMEN

Acidic pH in the tumor microenvironment is associated with cancer metabolism and creates a physiological barrier that prevents from drugs to penetrate cells. Specifically, ionizable weak-base drugs, such as doxorubicin, freely permeate membranes in their uncharged form, however, in the acidic tumor microenvironment these drugs become charged and their cellular permeability is retarded. In this study, 100-nm liposomes loaded with sodium bicarbonate were used as adjuvants to elevate the tumor pH. Combined treatment of triple-negative breast cancer cells (4T1) with doxorubicin and sodium-bicarbonate enhanced drug uptake and increased its anti-cancer activity. In vivo, mice bearing orthotropic 4T1 breast cancer tumors were administered either liposomal or free bicarbonate intravenously. 3.7 ±â€¯0.3% of the injected liposomal dose was detected in the tumor after twenty-four hours, compared to 0.17% ±â€¯0.04% in the group injected free non-liposomal bicarbonate, a 21-fold increase. Analyzing nanoparticle biodistribution within the tumor tissue revealed that 93% of the PEGylated liposomes accumulated in the extracellular matrix, while 7% were detected intracellularly. Mice administered bicarbonate-loaded liposomes reached an intra-tumor pH value of 7.38 ±â€¯0.04. Treating tumors with liposomal bicarbonate combined with a sub-therapeutic dose of doxorubicin achieved an improved therapeutic outcome, compared to mice treated with doxorubicin or bicarbonate alone. Interestingly, analysis of the tumor microenvironment demonstrated an increase in immune cell' population (T-cell, B-cell and macrophages) in tumors treated with liposomal bicarbonate. This study demonstrates that targeting metabolic adjuvants with nanoparticles to the tumor microenvironment can enhance anticancer drug activity and improve treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias , Bicarbonato de Sodio/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Transporte Biológico/efectos de los fármacos , Recuento de Células , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacocinética , Femenino , Humanos , Concentración de Iones de Hidrógeno , Liposomas , Ratones Endogámicos BALB C , Neoplasias/química , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Bicarbonato de Sodio/farmacocinética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
11.
Sci Rep ; 8(1): 7589, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773873

RESUMEN

As the world population grows, there is a need for efficient agricultural technologies to provide global food requirements and reduce environmental toll. In medicine, nanoscale drug delivery systems grant improved therapeutic precision by overcoming biological barriers and enhancing drug targeting to diseased tissues. Here, we loaded nanoscale drug-delivery systems with agricultural nutrients, and applied them to the leaves of tomato plants. We show that the nanoparticles - liposomes composed of plant-derived lipids, penetrate the leaf and translocate in a bidirectional manner, distributing to other leaves and to the roots. The liposomes were then internalized by the plant cells, where they released their active ingredient. Up to 33% of the applied nanoparticles penetrated the leaf, compared to less than one percent of free-molecules applied in a similar manner. In our study, tomato plants treated with liposomes loaded with Fe and Mg overcame acute nutrient deficiency which was not treatable using ordinary agricultural nutrients. Furthermore, to address regulatory concerns regarding airborne nanoparticles, we rationally designed liposomes that were stable only over short spraying distances (less than 2 meters), while the liposomes disintegrated into safe molecular building blocks (phospholipids) over longer airborne distances. These findings support expanding the implementation of nanotechnology for delivering micronutrients to agricultural crops for increasing yield.


Asunto(s)
Productos Agrícolas/metabolismo , Sistemas de Liberación de Medicamentos , Liposomas/química , Nanopartículas/administración & dosificación , Nutrientes/administración & dosificación , Hojas de la Planta/metabolismo , Solanum lycopersicum/metabolismo , Productos Agrícolas/crecimiento & desarrollo , Solanum lycopersicum/crecimiento & desarrollo , Nanopartículas/química , Hojas de la Planta/crecimiento & desarrollo , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/metabolismo
12.
ACS Nano ; 12(2): 1482-1490, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29365250

RESUMEN

Surgical blades are common medical tools. However, blades cannot distinguish between healthy and diseased tissue, thereby creating unnecessary damage, lengthening recovery, and increasing pain. We propose that surgical procedures can rely on natural tissue remodeling tools-enzymes, which are the same tools our body uses to repair itself. Through a combination of nanotechnology and a controllably activated proteolytic enzyme, we performed a targeted surgical task in the oral cavity. More specifically, we engineered nanoparticles that contain collagenase in a deactivated form. Once placed at the surgical site, collagenase was released at a therapeutic concentration and activated by calcium, its biological cofactor that is naturally present in the tissue. Enhanced periodontal remodeling was recorded due to enzymatic cleavage of the supracrestal collagen fibers that connect the teeth to the underlying bone. When positioned in their new orientation, natural tissue repair mechanisms supported soft and hard tissue recovery and reduced tooth relapse. Through the combination of nanotechnology and proteolytic enzymes, localized surgical procedures can now be less invasive.


Asunto(s)
Colágeno/metabolismo , Colagenasas/administración & dosificación , Colagenasas/farmacología , Tejido Conectivo/efectos de los fármacos , Liposomas/química , Nanopartículas/química , Animales , Colagenasas/farmacocinética , Tejido Conectivo/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Enzimas Inmovilizadas/administración & dosificación , Enzimas Inmovilizadas/farmacocinética , Enzimas Inmovilizadas/farmacología , Masculino , Boca/efectos de los fármacos , Boca/metabolismo , Boca/cirugía , Nanotecnología/métodos , Proteolisis/efectos de los fármacos , Ratas Wistar
13.
Chem Eng J ; 340: 9-14, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31384170

RESUMEN

Polylactic acid (PLA) is the most commonly used biodegradable polymer in clinical applications today. Examples range from drug delivery systems, tissue engineering, temporary and long-term implantable devices; constantly expanding to new fields. This is owed greatly to the polymer's favorable biocompatibility and to its safe degradation products. Once coming in contact with biological media, the polymer begins breaking down, usually by hydrolysis, into lactic acid (LA) or to carbon dioxide and water. These products are metabolized intracellularly or excreted in the urine and breath. Bacterial infection and foreign-body inflammation enhance the breakdown of PLA, through the secretion of enzymes that degrade the polymeric matrix. The biodegradation occurs both on the surface of the polymeric device and inside the polymer body, by diffusion of water between the polymer chains. The median half-life of the polymer is 30 weeks; however, this can be lengthened or shortened to address the clinical needs. Degradation kinetics can be tuned by determining the molecular composition and the physical architecture of the device. Using L- or D- chirality of the LA will greatly slow or lengthen the degradation rates, respectively. Despite the fact that this polymer is more than 150 years old, PLA remains a fertile platform for biomedical innovation and fundamental understanding of how artificial polymers can safely coexist with biological systems.

14.
Adv Healthc Mater ; 7(9): e1701163, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29283226

RESUMEN

Synthetic cells, artificial cell-like particles, capable of autonomously synthesizing RNA and proteins based on a DNA template, are emerging platforms for studying cellular functions and for revealing the origins-of-life. Here, it is shown for the first time that artificial lipid-based vesicles, containing the molecular machinery necessary for transcription and translation, can be used to synthesize anticancer proteins inside tumors. The synthetic cells are engineered as stand-alone systems, sourcing nutrients from their biological microenvironment to trigger protein synthesis. When pre-loaded with template DNA, amino acids and energy-supplying molecules, up to 2 × 107 copies of green fluorescent protein are synthesized in each synthetic cell. A variety of proteins, having molecular weights reaching 66 kDa and with diagnostic and therapeutic activities, are synthesized inside the particles. Incubating synthetic cells, encoded to secrete Pseudomonas exotoxin A (PE) with 4T1 breast cancer cells in culture, resulted in killing of most of the malignant cells. In mice bearing 4T1 tumors, histological evaluation of the tumor tissue after a local injection of PE-producing particles indicates robust apoptosis. Synthetic cells are new platforms for synthesizing therapeutic proteins on-demand in diseased tissues.


Asunto(s)
ADP Ribosa Transferasas/biosíntesis , Células Artificiales/metabolismo , Toxinas Bacterianas/biosíntesis , Exotoxinas/biosíntesis , Neoplasias Experimentales , Microambiente Tumoral , Factores de Virulencia/biosíntesis , Animales , Línea Celular Tumoral , Femenino , Proteínas Fluorescentes Verdes/biosíntesis , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Exotoxina A de Pseudomonas aeruginosa
15.
Nanotechnology ; 28(43): 43LT01, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-28872058

RESUMEN

Despite advances in cancer therapy, treating cancer after it has metastasized remains an unmet clinical challenge. In this study we demonstrate that 100 nm liposomes target triple-negative murine breast-cancer metastases post intravenous administration. Metastatic breast cancer was induced in BALB/c mice either experimentally, by a tail vein injection of 4T1 cells, or spontaneously, after implanting a primary tumor xenograft. To track their biodistribution in vivo the liposomes were labeled with multi-modal diagnostic agents, including indocyanine green and rhodamine for whole-animal fluorescent imaging, gadolinium for magnetic resonance imaging (MRI), and europium for a quantitative biodistribution analysis. The accumulation of liposomes in the metastases peaked at 24 h post the intravenous administration, similar to the time they peaked in the primary tumor. The efficiency of liposomal targeting to the metastatic tissue exceeded that of a non-liposomal agent by 4.5-fold. Liposomes were detected at very early stages in the metastatic progression, including metastatic lesions smaller than 2 mm in diameter. Surprisingly, while nanoparticles target breast cancer metastasis, they may also be found in elevated levels in the pre-metastatic niche, several days before metastases are visualized by MRI or histologically in the tissue. This study highlights the promise of diagnostic and therapeutic nanoparticles for treating metastatic cancer, possibly even for preventing the onset of the metastatic dissemination by targeting the pre-metastatic niche.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Sistemas de Liberación de Medicamentos/métodos , Liposomas/farmacocinética , Neoplasias Pulmonares/diagnóstico por imagen , Metástasis de la Neoplasia/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/secundario , Línea Celular Tumoral , Europio/química , Europio/farmacocinética , Femenino , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacocinética , Liposomas/síntesis química , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias , Imagen Óptica , Rodaminas/química , Rodaminas/farmacocinética , Distribución Tisular , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/secundario
16.
J Control Release ; 257: 68-75, 2017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-27744036

RESUMEN

Injectable drug delivery systems that autonomously detect, propel towards, and ultimately treat the cancerous tissue, are the future of targeted medicine. Here, we developed a drug delivery system that swims autonomously towards cancer cells, where it releases a therapeutic cargo. This platform is based on viable bacteria, loaded with nanoparticles that contain the chemotherapeutic-antibiotic drug doxorubicin. The bacteria ferry across media and invade the cancer cells, increasing their velocity in the presence of nutrients that are present within the tumor microenvironment. Inside the cancer cells, doxorubicin is released from the nanoparticles, destroying the bacterial swimmer (antibiotic activity) and executing the therapeutic activity against the cancer cells (chemotherapeutic activity). This mode of delivery, where both the carrier and the cancer cell are destroyed, supports implementing nanoswimmers in drug delivery (Fig. 1).


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos , Escherichia coli/metabolismo , Neoplasias/tratamiento farmacológico , Salmonella typhimurium/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Doxorrubicina/farmacología , Escherichia coli/citología , Escherichia coli/efectos de los fármacos , Liposomas , Ratones , Salmonella typhimurium/citología , Salmonella typhimurium/efectos de los fármacos
17.
PLoS One ; 11(10): e0165137, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27768741

RESUMEN

Cell-free protein synthesis (CFPS) systems are important laboratory tools that are used for various synthetic biology applications. Here, we present a simple and inexpensive laboratory-scale method for preparing a CFPS system from E. coli. The procedure uses basic lab equipment, a minimal set of reagents, and requires less than one hour to process the bacterial cell mass into a functional S30-T7 extract. BL21(DE3) and MRE600 E. coli strains were used to prepare the S30-T7 extract. The CFPS system was used to produce a set of fluorescent and therapeutic proteins of different molecular weights (up to 66 kDa). This system was able to produce 40-150 µg-protein/ml, with variations depending on the plasmid type, expressed protein and E. coli strain. Interestingly, the BL21-based CFPS exhibited stability and increased activity at 40 and 45°C. To the best of our knowledge, this is the most rapid and affordable lab-scale protocol for preparing a cell-free protein synthesis system, with high thermal stability and efficacy in producing therapeutic proteins.


Asunto(s)
Proteínas de Escherichia coli/biosíntesis , Escherichia coli/metabolismo , Sistema Libre de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...