Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
MRS Bull ; 49(4): 299-309, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38645611

RESUMEN

Abstract: The current work presents a novel flexible multifunctional platform for biological interface applications. The use of titania nanotube arrays (TNAs) as a multifunctional material is explored for soft-tissue interface applications. In vitro biocompatibility of TNAs to brain-derived cells was first examined by culturing microglia cells-the resident immune cells of the central nervous system on the surface of TNAs. The release profile of an anti-inflammatory drug, dexamethasone from TNAs-on-polyimide substrates, was then evaluated under different bending modes. Flexible TNAs-on-polyimide sustained a linear release of anti-inflammatory dexamethasone up to ~11 days under different bending conditions. Finally, microfabrication processes for patterning and transferring TNA microsegments were developed to facilitate structural stability during device flexing and to expand the set of compatible polymer substrates. The techniques developed in this study can be applied to integrate TNAs or other similar nanoporous inorganic films onto various polymer substrates. Impact statement: Titania nanotube arrays (TNAs) are highly tunable and biocompatible structures that lend themselves to multifunctional implementation in implanted devices. A particularly important aspect of titania nanotubes is their ability to serve as nano-reservoirs for drugs or other therapeutic agents that slowly release after implantation. To date, TNAs have been used to promote integration with rigid, dense tissues for dental and orthopedic applications. This work aims to expand the implant applications that can benefit from TNAs by integrating them onto soft polymer substrates, thereby promoting compatibility with soft tissues. The successful direct growth and integration of TNAs on polymer substrates mark a critical step toward developing mechanically compliant implantable systems with drug delivery from nanostructured inorganic functional materials. Diffusion-driven release kinetics and the high drug-loading efficiency of TNAs offer tremendous potential for sustained drug delivery for scientific investigations, to treat injury and disease, and to promote device integration with biological tissues. This work opens new opportunities for developing novel and more effective implanted devices that can significantly improve patient outcomes and quality of life. Supplementary information: The online version contains supplementary material available at 10.1557/s43577-023-00628-y.

2.
Nanomedicine ; 28: 102216, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32413511

RESUMEN

Poor prognosis for glioblastoma (GBM) is a consequence of the aggressive and infiltrative nature of gliomas where individual cells migrate away from the main tumor to distant sites, making complete surgical resection and treatment difficult. In this manuscript, we characterize an invasive pediatric glioma model and determine if nanoparticles linked to a peptide recognizing the GBM tumor biomarker PTPmu can specifically target both the main tumor and invasive cancer cells in adult and pediatric glioma models. Using both iron and lipid-based nanoparticles, we demonstrate by magnetic resonance imaging, optical imaging, histology, and iron quantification that PTPmu-targeted nanoparticles effectively label adult gliomas. Using PTPmu-targeted nanoparticles in a newly characterized orthotopic pediatric SJ-GBM2 model, we demonstrate individual tumor cell labeling both within the solid tumor margins and at invasive and dispersive sites.


Asunto(s)
Glioblastoma/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Femenino , Compuestos Férricos/química , Glioblastoma/metabolismo , Glioma/diagnóstico por imagen , Glioma/metabolismo , Humanos , Ratones , Ratones Desnudos
3.
Mol Pharm ; 16(10): 4352-4360, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31442061

RESUMEN

Deposition of nanoparticles to tumors often can be enhanced by targeting receptors overexpressed in a tumor. However, a tumor may exhibit a finite number of a biomarker that is accessible and targetable by nanoparticles, limiting the available landing spots. To explore this, we selected two different biomarkers that effectively home nanoparticles in brain tumors. Specifically, we used either an αvß3 integrin-targeting peptide or a fibronectin-targeting peptide as a ligand on nanoparticles termed RGD-NP and CREKA-NP, respectively. In mouse models of glioblastoma multiforme, we systemically injected the nanoparticles loaded with a cytotoxic drug at different doses ranging from 2 to 8 mg/kg drug. The upper dose threshold of RGD-NP is ∼2 mg/kg. CREKA-NP reached its upper dose threshold at 5 mg/kg. For both targeted nanoparticle variants, higher dose did not ensure higher intratumoral drug levels, but it contributed to elevated off-target deposition and potentially greater toxicity. A cocktail combining RGD-NP and CREKA-NP was then administered at a dose corresponding to the upper dose threshold for each formulation resulting in a 3-fold higher intratumoral deposition than the individual formulations. The combination of the two different targeting schemes at the appropriate dose for each nanoparticle variant facilitated remarkable increase in intratumoral drug levels that was not achievable by a sole targeting nanoparticle alone.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina/farmacología , Fibronectinas/metabolismo , Integrina alfaVbeta3/metabolismo , Nanopartículas/administración & dosificación , Fragmentos de Péptidos/metabolismo , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/química , Apoptosis , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Proliferación Celular , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ligandos , Ratones , Ratones Desnudos , Nanopartículas/química , Fragmentos de Péptidos/química , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Pharm Res ; 31(6): 1460-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23934254

RESUMEN

PURPOSE: To evaluate the ability of radiofrequency (RF)-triggered drug release from a multicomponent chain-shaped nanoparticle to inhibit the growth of an aggressive breast tumor. METHODS: A two-step solid phase chemistry was employed to synthesize doxorubicin-loaded nanochains, which were composed of three iron oxide nanospheres and one doxorubicin-loaded liposome assembled in a 100-nm-long linear nanochain. The nanochains were tested in the 4T1-LUC-GFP orthotopic mouse model, which is a highly aggressive breast cancer model. The 4T1-LUC-GFP cell line stably expresses firefly luciferase, which allowed the non-invasive in vivo imaging of tumor response to the treatment using bioluminescence imaging (BLI). RESULTS: Longitudinal BLI imaging showed that a single nanochain treatment followed by application of RF resulted in an at least 100-fold lower BLI signal compared to the groups treated with nanochains (without RF) or free doxorubicin followed by RF. A statistically significant increase in survival time of the nanochain-treated animals followed by RF (64.3 days) was observed when compared to the nanochain-treated group without RF (35.7 days), free doxorubicin-treated group followed by RF (38.5 days), and the untreated group (30.5 days; n=5 animals per group). CONCLUSIONS: These studies showed that the combination of RF and nanochains has the potential to effectively treat highly aggressive cancers and prolong survival.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanopartículas/administración & dosificación , Adyuvantes Farmacéuticos , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Doxorrubicina/administración & dosificación , Doxorrubicina/análogos & derivados , Doxorrubicina/uso terapéutico , Femenino , Humanos , Luminiscencia , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/uso terapéutico , Ondas de Radio , Análisis de Supervivencia , Carga Tumoral
5.
J Control Release ; 362: 812-819, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37011838

RESUMEN

Targeting ligands have been widely used to increase the intratumoral accumulation of nanoparticles and their uptake by cancer cells. However, these ligands aim at targets that are often also upregulated in inflamed tissues. Here, we assessed the ability of targeted nanoparticles to distinguish metastatic cancer from sites of inflammation. Using common targeting ligands and a 60-nm liposome as a representative nanoparticle, we generated three targeted nanoparticle (NP) variants that targeted either fibronectin, folate, or αvß3 integrin, whose deposition was compared against that of standard untargeted NP. Using fluorescently labeled NPs and ex vivo fluorescence imaging of organs, we assessed the deposition of the NPs into the lungs of mice modeling 4 different biological landscapes, including healthy lungs, aggressive metastasis in lungs, dormant/latent metastasis in lungs, and lungs with general pulmonary inflammation. Among the four NP variants, fibronectin-targeting NP and untargeted NP exhibited the highest deposition in lungs harboring aggressive metastases. However, the deposition of all targeted NP variants in lungs with metastasis was similar to the deposition in lungs with inflammation. Only the untargeted NP was able to exhibit higher deposition in metastasis than inflammation. Moreover, flow-cytometry analysis showed all NP variants accumulated predominantly in immune cells rather than cancer cells. For example, the number of NP+ macrophages and dendritic cells was 16-fold greater than NP+ cancer cells in the case of fibronectin-targeting NP. Overall, targeted NPs were unable to distinguish cancer metastasis from general inflammation, which may have clinical implications to the nanoparticle-mediated delivery of cancer drugs.

6.
Nanoscale ; 14(4): 1144-1159, 2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35023530

RESUMEN

Lethal cancer is characterized by drug-resistant relapse and metastasis. Here, we evaluate the efficacy of a neoadjuvant therapeutic strategy prior to surgery that combines the immune checkpoint inhibitor anti-PD1 with a powerful immunostimulatory nanoparticle (immuno-NP). Lipid-based immuno-NPs are uniquely designed to co-encapsulate a STING and TLR4 agonist that are functionally synergistic. Efficacy of neoadjuvant combination immunotherapy was assessed in three aggressive murine tumor models, including B16F10 melanoma and 4T1 and D2.A1 breast cancer. Primary splenocytes treated with dual-agonist immuno-NPs produced a 75-fold increased production of interferon ß compared to single-agonist treatments. Systemic delivery facilitated the widespread deposition of immuno-NPs in the perivascular space throughout the tumor mass and their preferential uptake by tumor-resident antigen-presenting cells. Our findings strongly suggested that immuno-NPs, when administered in combination with anti-PD1, harnessed and activated the otherwise "exhausted" CD8+ T cells as key mediators of tumor clearance. Neoadjuvant combination immunotherapy resulted in significant efficacy, curative responses, and protective immunological memory in 71% of good-responding mice bearing B16F10 melanoma tumors and showed similar trends in the two breast cancer models. Finally, this neoadjuvant combination immunotherapy drove the generation of B and T cell de novo epitopes for a comprehensive memory response.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Linfocitos T CD8-positivos , Inmunización , Inmunoterapia , Ratones , Terapia Neoadyuvante
7.
J Mater Chem B ; 10(2): 224-235, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34846443

RESUMEN

To alter the immunosuppressive tumor microenvironment (TME), we developed an immunostimulatory nanoparticle (NP) to reprogram a tumor's dysfunctional and inhibitory antigen-presenting cells (APCs) into properly activated APCs that stimulate tumor-reactive cytotoxic T cells. Importantly, systemic delivery allowed NPs to efficiently utilize the entire microvasculature and gain access into the majority of the perivascular TME, which coincided with the APC-rich tumor areas leading to uptake of the NPs predominantly by APCs. In this work, a 60 nm NP was loaded with a STING agonist, which triggered robust production of interferon ß, resulting in activation of APCs. In addition to untargeted NPs, we employed 'mainstream' ligands targeting fibronectin, αvß3 integrin and P-selectin that are commonly used to direct nanoparticles to tumors. Using the 4T1 mouse model, we assessed the microdistribution of the four NP variants in the tumor immune microenvironment in three different breast cancer landscapes, including primary tumor, early metastasis, and late metastasis. The different NP variants resulted in variable uptake by immune cell subsets depending on the organ and tumor stage. Among the NP variants, therapeutic studies indicated that the untargeted NPs and the integrin-targeting NPs exhibited a remarkable short- and long-term immune response and long-lasting antitumor effect.


Asunto(s)
Neoplasias de la Mama/terapia , GMP Cíclico/análogos & derivados , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Nanopartículas/química , 1,2-Dipalmitoilfosfatidilcolina/química , Animales , Línea Celular Tumoral , GMP Cíclico/uso terapéutico , Células Dendríticas/efectos de los fármacos , Ligandos , Macrófagos/efectos de los fármacos , Ratones Endogámicos BALB C , Péptidos/química , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Linfocitos T/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
8.
Nanotechnology ; 22(11): 115101, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21387846

RESUMEN

In the recent past, remarkable advances in nanotechnology have generated nanoparticles of different shapes and sizes, which have been shown to exhibit unique properties suitable for biomedical applications such as cancer therapy and imaging. Obviously, all nanoparticles are not made equal. This becomes evident when we consider their transport behavior under blood flow in microcirculation. In this work, we evaluated the effect of critical physical characteristics such as the particle shape, size and density on a nanoparticle's tendency to marginate towards the vessel walls in microcirculation using an in vitro model. The wall deposition of nanoparticles was tested in a fibronectin-coated microfluidic channel at a physiologically relevant flow rate. Different classes of nanoparticles (liposome, metal particles) of different sizes (60-130 nm), densities (1-19 g ml(-1)) and shapes (sphere, rod) displayed significantly different deposition as a result of different margination rates. The smaller-sized and the oblate-shaped particles displayed a favorable behavior as indicated by their higher margination rates. Notably, the particle density showed an even more essential role, as it was observed that the lighter particles marginated significantly more. Since nanoparticles must escape the flow in order to approach the vascular bed and subsequently extravascular components for meaningful interactions, the design of nanoparticles strongly affects their margination, a key factor for their ultimate in vivo effectiveness.


Asunto(s)
Liposomas/química , Técnicas Analíticas Microfluídicas/instrumentación , Nanopartículas/química , Diseño de Equipo , Microcirculación , Tamaño de la Partícula
9.
Adv Healthc Mater ; 10(5): e2001044, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33225633

RESUMEN

Recent advancements in unravelling elements of cancer biology involved in disease progression and treatment resistance have highlighted the need for a holistic approach to effectively tackle cancer. Stimuli-responsive nanotheranostics based on iron oxide nanoparticles are an emerging class of versatile nanomedicines with powerful capabilities to "seek, sense, and attack" multiple components of solid tumors. In this work, the rationale for using iron oxide nanoparticles and the basic physical principles that impact their function in biomedical applications are reviewed. Subsequently, recent advances in the integration of iron oxide nanoparticles with various stimulus mechanisms to facilitate the development of stimuli-responsive nanotheranostics for application in cancer therapy are summarized. The integration of an iron oxide core with various surface coating mechanisms results in the generation of hybrid nanoconstructs with capabilities to codeliver a wide variety of highly potent anticancer therapeutics and immune modulators. Finally, emerging future directions and considerations for their clinical translation are touched upon.


Asunto(s)
Neoplasias , Nanomedicina Teranóstica , Compuestos Férricos , Humanos , Nanomedicina , Neoplasias/tratamiento farmacológico
10.
Nanoscale Adv ; 3(17): 4961-4972, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34485818

RESUMEN

The efficacy of immunotherapies is often limited by the immunosuppressive tumor microenvironment, which is populated with dysfunctional innate immune cells. To reprogram the tumor-resident innate immune cells, we developed immunostimulatory silica mesoporous nanoparticles (immuno-MSN). The cargo of immuno-MSN is a Stimulator of Interferon Gene (STING) agonist, which activates innate immune cells leading to production of interferon (IFN) ß. By proficiently trafficking its cargo into immune cells, the immuno-MSN induced a 9-fold increase of IFN-ß secretion compared to free agonist. While an external PEG shield has historically been used to protect nanoparticles from immune recognition, a PEGylated immunostimulatory nanoparticle needs to strike a balance between immune evasion to avoid off-site accumulation and uptake by target immune cells in tumors. Using the 4T1 mouse model of metastatic breast cancer and flow cytometry, it was determined that the degree of PEGylation significantly influenced the uptake of 'empty' MSNs by tumor-resident innate immune cells. This was not the case for the agonist-loaded immuno-MSN variants. It should be noted the surface charge of the 'empty' MSNs was positive rather than neutral for the agonist-loaded immuno-MSNs. However, even though the cellular uptake was similar at 24 h after injection for the three immuno-MSN variants, we observed a significant beneficial effect on the activation and expansion of APCs especially in lung metastasis using the lightly PEGylated immuno-MSN variant.

11.
Nanoscale Horiz ; 6(2): 156-167, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33400743

RESUMEN

The high mortality associated with glioblastoma multiforme (GBM) is attributed to its invasive nature, hypoxic core, resistant cell subpopulations and a highly immunosuppressive tumor microenvironment (TME). To support adaptive immune function and establish a more robust antitumor immune response, we boosted the local innate immune compartment of GBM using an immunostimulatory mesoporous silica nanoparticle, termed immuno-MSN. The immuno-MSN was specifically designed for systemic and proficient delivery of a potent innate immune agonist to dysfunctional antigen-presenting cells (APCs) in the brain TME. The cargo of the immuno-MSN was cyclic diguanylate monophosphate (cdGMP), a Stimulator of Interferon Gene (STING) agonist. Studies showed the immuno-MSN promoted the uptake of STING agonist by APCs in vitro and the subsequent release of the pro-inflammatory cytokine interferon ß, 6-fold greater than free agonist. In an orthotopic GBM mouse model, systemically administered immuno-MSN particles were taken up by APCs in the near-perivascular regions of the brain tumor with striking efficiency. The immuno-MSNs facilitated the recruitment of dendritic cells and macrophages to the TME while sparing healthy brain tissue and peripheral organs, resulting in elevated circulating CD8+ T cell activity (2.5-fold) and delayed GBM tumor growth. We show that an engineered immunostimulatory nanoparticle can support pro-inflammatory innate immune function in GBM and subsequently augment current immunotherapeutic interventions and improve their therapeutic outcome.


Asunto(s)
Neoplasias Encefálicas/terapia , GMP Cíclico/análogos & derivados , Glioblastoma/terapia , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Nanopartículas/uso terapéutico , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , GMP Cíclico/síntesis química , GMP Cíclico/uso terapéutico , Células Dendríticas/efectos de los fármacos , Femenino , Factores Inmunológicos/síntesis química , Inmunoterapia/métodos , Interferón Tipo I/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Porosidad , Células RAW 264.7 , Dióxido de Silicio/química , Microambiente Tumoral/efectos de los fármacos
12.
Nanoscale Adv ; 3(20): 5890-5899, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34746645

RESUMEN

Iron oxide nanoparticles (IONPs) have often been investigated for tumor hyperthermia. IONPs act as heating foci in the presence of an alternating magnetic field (AMF). It has been shown that hyperthermia can significantly alter the tumor immune microenvironment. Typically, mild hyperthermia invokes morphological changes within the tumor, which elicits a secretion of inflammatory cytokines and tumor neoantigens. Here, we focused on the direct effect of IONP-induced hyperthermia on the various tumor-resident immune cell subpopulations. We compared direct intratumoral injection to systemic administration of IONPs followed by application of an external AMF. We used the orthotopic 4T1 mouse model, which represents aggressive and metastatic breast cancer with a highly immunosuppressive microenvironment. A non-inflamed and 'cold' microenvironment inhibits peripheral effector lymphocytes from effectively trafficking into the tumor. Using intratumoral or systemic injection, IONP-induced hyperthermia achieved a significant reduction of all the immune cell subpopulations in the tumor. However, the systemic delivery approach achieved superior outcomes, resulting in substantial reductions in the populations of both innate and adaptive immune cells. Upon depletion of the existing dysfunctional tumor-resident immune cells, subsequent treatment with clinically approved immune checkpoint inhibitors encouraged the repopulation of the tumor with 'fresh' infiltrating innate and adaptive immune cells, resulting in a significant decrease of the tumor cell population.

13.
Sci Rep ; 10(1): 14824, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908180

RESUMEN

Nerve fibers are known to reside within malignant tumors and the greater the neuronal density the worse prognosis for the patient. Recent discoveries using tumor bearing animal models have eluded to the autonomic nervous system having a direct effect on tumor growth and metastasis. We report the first direct and chronic in vivo measurements of neural activity within tumors. Using a triple-negative mammary cancer mouse model and chronic neural interface techniques, we have recorded neural activity directly within the tumor mass while the tumor grows and metastasizes. The results indicate that there is a strong connection between the autonomic nervous system and the tumor and could help uncover the mechanisms of tumor growth and metastasis.


Asunto(s)
Glándulas Mamarias Animales/inervación , Neoplasias Mamarias Experimentales/patología , Neoplasias de la Mama Triple Negativas/patología , Animales , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia
14.
Radiology ; 250(2): 398-406, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19188313

RESUMEN

PURPOSE: To prospectively predict the effectiveness of a clinically used nanochemotherapeutic agent by detecting and measuring the intratumoral uptake of an x-ray contrast agent nanoprobe by using digital mammography. MATERIALS AND METHODS: All animal procedures were approved by the institutional animal care and use committee. A long-circulating 100-nm-scale injectable liposomal probe encapsulating 155 mg/mL iodine was developed. Preliminary studies were performed to identify the agent dose that would result in adequate tumor enhancement without enhancement of the normal vasculature in rats. This dose was used to image a rat breast tumor (n = 14) intermittently for 3 days by using a digital mammography system; subsequently, the animals were treated with liposomal doxorubicin. The predictive capability of the probe was characterized by creating good- and bad-prognosis subgroups, on the basis of tumor enhancement found during imaging, and analyzing the tumor growth after treatment of the animals in these two subgroups. RESULTS: A dose of 455 mg of iodine per kilogram of body weight was found to produce an undetectable signal from the blood while achieving enough intratumoral accumulation of the probe to produce adequate signal for detection. The good- and bad-prognosis subgroups demonstrated differential tumor growth rates (P < .003). An inverse linear relationship between the contrast enhancement rate constant during imaging and the tumor growth rate constant during treatment was found (slope = -0.576, R(2) = 0.838). CONCLUSION: In this animal model, quantitative measurement of vascular permeability enabled prediction of therapeutic responsiveness of tumors to liposomal doxorubicin.


Asunto(s)
Doxorrubicina/farmacología , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Análisis de Varianza , Animales , Medios de Contraste , Doxorrubicina/administración & dosificación , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Yodo , Neoplasias Mamarias Experimentales/patología , Mamografía , Método de Montecarlo , Nanopartículas , Ratas , Análisis de Regresión
15.
PLoS One ; 14(7): e0220474, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31356633

RESUMEN

Metastasis is responsible for the majority of deaths of breast cancer patients. While cytotoxic drugs are available with high potency to kill breast cancer cells, they are not designed to specifically seek and navigate in the dynamic and continuously changing microenvironment of metastatic disease. To effectively delivery chemotherapeutic agents to metastasis, we designed a dual-ligand nanoparticle loaded with doxorubicin by using two different types of ligands targeting EGFR and αvß3 integrin. Metastatic cancer cells continuously change resulting in heterogeneity even across adjacent micrometastatic regions with variable expression of these targetable receptors. Using a mouse model of breast cancer metastasis, in vivo and ex vivo imaging showed that both EGFR and αvß3 integrin-targeting were required to reliably direct the nanoparticle to metastasis and capture the spread and exact topology of the disease. Survival studies compared the anticancer efficacy of the standard drug, EGFR-targeting nanoparticle, αvß3 integrin-targeting nanoparticle and the dual-ligand nanoparticle. While all the other treatments produced moderate therapeutic outcomes, treatment with the dual-ligand nanoparticle yielded significant improvement and event-free survival in a mouse model of breast cancer metastasis.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Integrina alfaVbeta3/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/administración & dosificación , Animales , Antibióticos Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Doxorrubicina/química , Receptores ErbB/química , Receptores ErbB/metabolismo , Femenino , Humanos , Integrina alfaVbeta3/química , Ligandos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Resultado del Tratamiento , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Res ; 79(20): 5394-5406, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31431457

RESUMEN

Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I IFNß. Using a murine model of metastatic triple-negative breast cancer, systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune-potentiating cargoes for effective APC-driven local and systemic antitumor immunity. SIGNIFICANCE: Systemic administration of an immuno-nanoparticle in a murine breast tumor model drives a robust tumor site-specific APC response by delivering two synergistic immune-potentiating molecules, highlighting the potential of nanoparticles for immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , GMP Cíclico/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Interferón beta/fisiología , Lípido A/análogos & derivados , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Nanocápsulas/administración & dosificación , Receptor Toll-Like 4/agonistas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , GMP Cíclico/administración & dosificación , GMP Cíclico/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Células Asesinas Naturales/inmunología , Lípido A/administración & dosificación , Lípido A/uso terapéutico , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microcirculación , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología
17.
PLoS One ; 13(10): e0204296, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30335750

RESUMEN

Nanoparticles often only exploit the upregulation of a receptor on cancer cells to enhance intratumoral deposition of therapeutic and imaging agents. However, a single targeting moiety assumes that a tumor is homogenous and static. Tumoral microenvironments are both heterogenous and dynamic, often displaying variable spatial and temporal expression of targetable receptors throughout disease progression. Here, we evaluated the in vivo performance of an iron oxide nanoparticle in terms of targeting and imaging of orthotropic mouse models of aggressive breast tumors. The nanoparticle, a multi-component nanochain, was comprised of 3-5 iron oxide nanoparticles chemically linked in a linear chain. The nanoparticle's surface was decorated with two types of ligands each targeting two different upregulated biomarkers on the tumor endothelium, P-selectin and fibronectin. The nanochain exhibited improved tumor deposition not only through vascular targeting but also through its elongated structure. A single-ligand nanochain exhibited a ~2.5-fold higher intratumoral deposition than a spherical nanoparticle variant. Furthermore, the dual-ligand nanochain exhibited higher consistency in generating detectable MR signals compared to a single-ligand nanochain. Using a 7T MRI, the dual-ligand nanochains exhibited highly detectable MR signal within 3h after injection in two different animal models of breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Fibronectinas/metabolismo , Nanopartículas/química , Selectina-P/metabolismo , Péptidos/administración & dosificación , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Compuestos Ferrosos/química , Humanos , Imagen por Resonancia Magnética , Ratones , Imagen Molecular , Trasplante de Neoplasias , Péptidos/química , Sensibilidad y Especificidad
18.
Adv Drug Deliv Rev ; 113: 141-156, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27639317

RESUMEN

This review seeks to highlight the enormous potential of targeted nanoparticles for molecular imaging applications. Being the closest point-of-contact, circulating nanoparticles can gain direct access to targetable molecular markers of disease that appear on the endothelium. Further, nanoparticles are ideally suitable to vascular targeting due to geometrically enhanced multivalent attachment on the vascular target. This natural synergy between nanoparticles, vascular targeting and molecular imaging can provide new avenues for diagnosis and prognosis of disease with quantitative precision. In addition to the obvious applications of targeting molecular signatures of vascular diseases (e.g., atherosclerosis), deep-tissue diseases often manifest themselves by continuously altering and remodeling their neighboring blood vessels (e.g., cancer). Thus, the remodeled endothelium provides a wide range of targets for nanoparticles and molecular imaging. To demonstrate the potential of molecular imaging, we present a variety of nanoparticles designed for molecular imaging of cancer or atherosclerosis using different imaging modalities.


Asunto(s)
Vasos Sanguíneos/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Imagen Molecular , Nanopartículas/metabolismo , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/metabolismo , Aterosclerosis/patología , Vasos Sanguíneos/patología , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Neoplasias/patología
19.
J Control Release ; 113(2): 117-27, 2006 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-16765471

RESUMEN

An aerosol insulin carrier based on the agglomerated vesicle technology that the authors have previously advanced [E. Karathanasis et al. J. Control. Release 103 (2005) 159-175] was evaluated in vivo. The carrier consisted of insulin-loaded liposomes cross-linked via chemical bridges cleavable by cysteine. It was speculated that the cleavage of the cross links released internal surface area and possibly resulted in the disruption of the liposomal walls. The result was a rapid release of encapsulated insulin upon contact of the insulin carrier with cysteine. The particles exhibited a small aerodynamic diameter within the respirable range suggesting deposition into the deep lung of humans along with a large geometrical diameter, consistent with long residence time. Indeed the endotracheal instillation of the particles into hyperglycemic rats decreased the glucose levels rapidly while delivery of cysteine triggered a further drop of glucose implying acceleration of insulin release from the carrier. Euglycemic clamp studies verified the accelerated insulin release upon application of cysteine.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Insulina/administración & dosificación , Insulina/farmacocinética , Administración por Inhalación , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Humanos , Liposomas , Masculino , Ratas , Ratas Sprague-Dawley
20.
Artículo en Inglés | MEDLINE | ID: mdl-26749497

RESUMEN

Despite advancements in surgery and radiotherapy, the aggressive forms of brain tumors, such as gliomas, are still uniformly lethal with current therapies offering only palliation complicated by significant toxicities. Gliomas are characteristically diffuse with infiltrating edges, resistant to drugs and nearly inaccessible to systemic therapies due to the brain-tumor barrier. Currently, aggressive efforts are underway to further understand brain-tumor's microenvironment and identify brain tumor cell-specific regulators amenable to pharmacologic interventions. While new potent agents are continuously becoming available, efficient drug delivery to brain tumors remains a limiting factor. To tackle the drug delivery issues, a multicomponent chain-like nanoparticle has been developed. These nanochains are comprised of iron oxide nanospheres and a drug-loaded liposome chemically linked into a 100-nm linear, chain-like assembly with high precision. The nanochain possesses a unique ability to scavenge the tumor endothelium. By utilizing effective vascular targeting, the nanochains achieve rapid deposition on the vascular bed of glioma sites establishing well-distributed drug reservoirs on the endothelium of brain tumors. After reaching the target sites, an on-command, external low-power radiofrequency field can remotely trigger rapid drug release, due to mechanical disruption of the liposome, facilitating widespread and effective drug delivery into regions harboring brain tumor cells. Integration of the nanochain delivery system with the appropriate combination of complementary drugs has the potential to unfold the field and allow significant expansion of therapies for the disease where success is currently very limited. WIREs Nanomed Nanobiotechnol 2016, 8:678-695. doi: 10.1002/wnan.1387 For further resources related to this article, please visit the WIREs website.


Asunto(s)
Antineoplásicos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas , Sistemas de Liberación de Medicamentos , Liposomas , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Modelos Animales de Enfermedad , Humanos , Liposomas/farmacocinética , Liposomas/uso terapéutico , Nanopartículas de Magnetita , Ratones , Nanomedicina Teranóstica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA