Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Commun ; 14(1): 771, 2023 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-36774352

RESUMEN

Glioblastoma, the most common and aggressive primary brain tumor type, is considered an immunologically "cold" tumor with sparse infiltration by adaptive immune cells. Immunosuppressive tumor-associated myeloid cells are drivers of tumor progression. Therefore, targeting and reprogramming intratumoral myeloid cells is an appealing therapeutic strategy. Here, we investigate a ß-cyclodextrin nanoparticle (CDNP) formulation encapsulating the Toll-like receptor 7 and 8 (TLR7/8) agonist R848 (CDNP-R848) to reprogram myeloid cells in the glioma microenvironment. We show that intravenous monotherapy with CDNP-R848 induces regression of established syngeneic experimental glioma, resulting in increased survival rates compared with unloaded CDNP controls. Mechanistically, CDNP-R848 treatment reshapes the immunosuppressive tumor microenvironment and orchestrates tumor clearing by pro-inflammatory tumor-associated myeloid cells, independently of T cells and NK cells. Using serial magnetic resonance imaging, we identify a radiomic signature in response to CDNP-R848 treatment and ultrasmall superparamagnetic iron oxide (USPIO) imaging reveals that immunosuppressive macrophage recruitment is reduced by CDNP-R848. In conclusion, CDNP-R848 induces tumor regression in experimental glioma by targeting blood-borne macrophages without requiring adaptive immunity.


Asunto(s)
Glioma , Nanopartículas , Receptor Toll-Like 7 , Receptor Toll-Like 8 , Humanos , Adyuvantes Inmunológicos , Glioma/tratamiento farmacológico , Macrófagos , Linfocitos T , Receptor Toll-Like 7/agonistas , Microambiente Tumoral , Receptor Toll-Like 8/agonistas
2.
Nat Nanotechnol ; 16(7): 830-839, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33958764

RESUMEN

Nanoparticulate albumin bound paclitaxel (nab-paclitaxel, nab-PTX) is among the most widely prescribed nanomedicines in clinical use, yet it remains unclear how nanoformulation affects nab-PTX behaviour in the tumour microenvironment. Here, we quantified the biodistribution of the albumin carrier and its chemotherapeutic payload in optically cleared tumours of genetically engineered mouse models, and compared the behaviour of nab-PTX with other clinically relevant nanoparticles. We found that nab-PTX uptake is profoundly and distinctly affected by cancer-cell autonomous RAS signalling, and RAS/RAF/MEK/ERK inhibition blocked its selective delivery and efficacy. In contrast, a targeted screen revealed that IGF1R kinase inhibitors enhance uptake and efficacy of nab-PTX by mimicking glucose deprivation and promoting macropinocytosis via AMPK, a nutrient sensor in cells. This study thus shows how nanoparticulate albumin bound drug efficacy can be therapeutically improved by reprogramming nutrient signalling and enhancing macropinocytosis in cancer cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutación , Nanopartículas , Neoplasias Experimentales/tratamiento farmacológico , Paclitaxel , Proteínas Proto-Oncogénicas p21(ras)/genética , Albúmina Sérica Humana , Animales , Línea Celular Tumoral , Glucosa/deficiencia , Glucosa/metabolismo , Humanos , Ratones , Ratones Transgénicos , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células RAW 264.7 , Albúmina Sérica Humana/química , Albúmina Sérica Humana/farmacología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
3.
Radiology ; 298(1): 123-132, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33107799

RESUMEN

Background Anaplastic thyroid cancer (ATC) is aggressive with a poor prognosis, partly because of the immunosuppressive microenvironment created by tumor-associated macrophages (TAMs). Purpose To understand the relationship between TAM infiltration, tumor vascularization, and corresponding drug delivery by using ferumoxytol-enhanced MRI and macrin in an ATC mouse model. Materials and Methods ATC tumors were generated in 6-8-week-old female B6129SF1/J mice through intrathyroid injection to model orthotopic tumors, or intravenously to model hematogenous metastasis, and prospectively enrolled randomly into treatment cohorts (n = 94 total; August 1, 2018, to January 15, 2020). Mice were treated with vehicle or combined serine/threonine-protein kinase B-Raf (BRAF) kinase inhibitor (BRAFi) and anti-PDL1 antibody (aPDL1). A subset was cotreated with therapies, including an approximately 70-nm model drug delivery nanoparticle (DDNP) to target TAM, and an antibody-neutralizing colony stimulating factor 1 receptor (CSF1R). Imaging was performed at the macroscopic level with ferumoxytol-MRI and microscopically with macrin. Genetically engineered BrafV600E/WT p53-null allografts were used and complemented by a GFP-transgenic derivative and human xenografts. Tumor-bearing organs were processed by using tissue clearing and imaged with confocal microscopy and MRI. Two-tailed Wilcoxon tests were used for comparison (≥five per group). Results TAM levels were higher in orthotopic thyroid tumors compared with pulmonary metastatic lesions by 79% ± 23 (standard deviation; P < .001). These findings were concordant with ferumoxytol MRI, which showed 136% ± 88 higher uptake in thyroid lesions (P = .02) compared with lung lesions. BRAFi and aPDL1 combination therapy resulted in higher tumor DDNP delivery by 39% ± 14 in pulmonary lesions (P = .004). Compared with the untreated group, tumors following BRAFi, aPDL1, and CSF1R-blocking antibody combination therapy did not show greater levels of TAM or DDNP (P = .82). Conclusion In a mouse model of anaplastic thyroid cancer, ferumoxytol MRI showed 136% ± 88 greater uptake in orthotopic thyroid tumors compared with pulmonary lesions, which reflected high vascularization and greater tumor-associated macrophage (TAM) levels. Serine/threonine-protein kinase B-Raf inhibitor and anti-programmed death ligand 1 antibody elicited higher local TAM levels and 43% ± 20 greater therapeutic nanoparticle delivery but not higher vascularization in pulmonary tumors. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Luker in this issue.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Imagen por Resonancia Magnética/métodos , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Carcinoma Anaplásico de Tiroides/diagnóstico por imagen , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Antineoplásicos/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Óxido Ferrosoférrico , Inmunidad/inmunología , Ratones , Nanopartículas , Proteínas Proto-Oncogénicas B-raf/inmunología , Carcinoma Anaplásico de Tiroides/inmunología , Macrófagos Asociados a Tumores/inmunología
4.
Cell Chem Biol ; 27(1): 94-104.e5, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31902676

RESUMEN

Immune-checkpoint blockers can promote sustained clinical responses in a subset of cancer patients. Recent research has shown that a subpopulation of tumor-infiltrating dendritic cells functions as gatekeepers, sensitizing tumors to anti-PD-1 treatment via production of interleukin-12 (IL-12). Hypothesizing that myeloid cell-targeted nanomaterials could be used to deliver small-molecule IL-12 inducers, we performed high-content image-based screening to identify the most efficacious small-molecule compounds. Using one lead candidate, LCL161, we created a myeloid-targeted nanoformulation that induced IL-12 production in intratumoral myeloid cells in vivo, slowed tumor growth as a monotherapy, and had no significant systemic toxicity. These results pave the way for developing combination immunotherapeutics by harnessing IL-12 production for immunostimulation.


Asunto(s)
Alquinos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/terapia , Inmunoterapia , Células Mieloides/efectos de los fármacos , Oligopéptidos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Tiazoles/farmacología , Alquinos/química , Animales , Antineoplásicos/química , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células Dendríticas , Portadores de Fármacos/química , Evaluación Preclínica de Medicamentos , Interleucina-12/biosíntesis , Ratones , Células Mieloides/metabolismo , Células Mieloides/patología , Nanopartículas/química , Oligopéptidos/química , Bibliotecas de Moléculas Pequeñas/química , Tiazoles/química
5.
ACS Nano ; 12(12): 12015-12029, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30508377

RESUMEN

Tumor-associated macrophages (TAMs) are widely implicated in cancer progression, and TAM levels can influence drug responses, particularly to immunotherapy and nanomedicines. However, it has been difficult to quantify total TAM numbers and their dynamic spatiotemporal distribution in a non-invasive and translationally relevant manner. Here, we address this need by developing a pharmacokinetically optimized, 64Cu-labeled polyglucose nanoparticle (Macrin) for quantitative positron emission tomography (PET) imaging of macrophages in tumors. By combining PET with high-resolution in vivo confocal microscopy and ex vivo imaging of optically cleared tissue, we found that Macrin was taken up by macrophages with >90% selectivity. Uptake correlated with the content of macrophages in both healthy tissue and tumors ( R2 > 0.9) and showed striking heterogeneity in the TAM content of an orthotopic and immunocompetent mouse model of lung carcinoma. In a proof-of-principle application, we imaged Macrin to monitor the macrophage response to neo-adjuvant therapy, using a panel of chemotherapeutic and γ-irradiation regimens. Multiple treatments elicited 180-650% increase in TAMs. Imaging identified especially TAM-rich tumors thought to exhibit enhanced permeability and retention of nanotherapeutics. Indeed, these TAM-rich tumors accumulated >700% higher amounts of a model poly(d,l-lactic- co-glycolic acid)- b-polyethylene glycol (PLGA-PEG) therapeutic nanoparticle compared to TAM-deficient tumors, suggesting that imaging may guide patient selection into nanomedicine trials. In an orthotopic breast cancer model, chemoradiation enhanced TAM and Macrin accumulation in tumors, which corresponded to the improved delivery and efficacy of two model nanotherapies, PEGylated liposomal doxorubicin and a TAM-targeted nanoformulation of the toll-like receptor 7/8 agonist resiquimod (R848). Thus, Macrin imaging offers a selective and translational means to quantify TAMs and inform therapeutic decisions.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Glucanos/química , Marcaje Isotópico , Neoplasias Pulmonares/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Nanopartículas/química , Animales , Radioisótopos de Cobre , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Neoplasias Pulmonares/diagnóstico por imagen , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Terapia Neoadyuvante , Tomografía de Emisión de Positrones
6.
Theranostics ; 8(21): 5842-5854, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30613266

RESUMEN

Tumor-associated macrophages (TAM) have attracted attention as they can modulate key cancer-related activities, yet TAM represent a heterogenous group of cells that remain incompletely characterized. In growing tumors, TAM are often referred to as M2-like macrophages, which are cells that display immunosuppressive and tumorigenic functions and express the enzyme arginase 1 (Arg1). Methods: Here we combined high resolution intravital imaging with single cell RNA seq to uncover the topography and molecular profiles of immunosuppressive macrophages in mice. We further assessed how immunotherapeutic interventions impact these cells directly in vivo. Results: We show that: i) Arg1+ macrophages are more abundant in tumors compared to other organs; ii) there exist two morphologically distinct subsets of Arg1 TAM defined by previously unknown markers (Gbp2b, Bst1, Sgk1, Pmepa1, Ms4a7); iii) anti-Programmed Cell Death-1 (aPD-1) therapy decreases the number of Arg1+ TAM while increasing Arg1- TAM; iv) accordingly, pharmacological inhibition of arginase 1 does not synergize with aPD-1 therapy. Conclusion: Overall, this research shows how powerful complementary single cell analytical approaches can be used to improve our understanding of drug action in vivo.


Asunto(s)
Arginasa/análisis , Expresión Génica , Tolerancia Inmunológica , Linfoma/patología , Macrófagos/química , Macrófagos/inmunología , Melanoma/patología , Animales , Modelos Animales de Enfermedad , Microscopía Intravital , Ratones , Análisis de Secuencia de ARN
7.
Nanotheranostics ; 1(3): 296-312, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29071194

RESUMEN

Anticancer nanotherapeutics have shown mixed results in clinical trials, raising the questions of whether imaging should be used to i) identify patients with a higher likelihood of nanoparticle accumulation, ii) assess nanotherapeutic efficacy before traditional measures show response, and iii) guide adjuvant treatments to enhance therapeutic nanoparticle (TNP) delivery. Here we review the use of a clinically approved MRI nanoparticle (ferumoxytol, FMX) to predict TNP delivery and efficacy. It is becoming increasingly apparent that nanoparticles used for imaging, despite clearly distinct physicochemical properties, often co-localize with TNP in tumors. This evidence offers the possibility of using FMX as a generic "companion diagnostic" nanoparticle for multiple TNP formulations, thus potentially allowing many of the complex regulatory and cost challenges of other approaches to be avoided.

8.
Proc Natl Acad Sci U S A ; 112(19): 6146-51, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25902531

RESUMEN

At their margins, tumors often contain neutrophils, dendritic cells, and activated macrophages, which express class II MHC and CD11b products. The interplay between stromal cells, tumor cells, and migratory cells such as lymphocytes creates opportunities for noninvasive imaging of immune responses. We developed alpaca-derived antibody fragments specific for mouse class II MHC and CD11b products, expressed on the surface of a variety of myeloid cells. We validated these reagents by flow cytometry and two-photon microscopy to obtain images at cellular resolution. To enable noninvasive imaging of the targeted cell populations, we developed a method to site-specifically label VHHs [the variable domain (VH) of a camelid heavy-chain only antibody] with (18)F or (64)Cu. Radiolabeled VHHs rapidly cleared the circulation (t1/2 ≈ 20 min) and clearly visualized lymphoid organs. We used VHHs to explore the possibility of imaging inflammation in both xenogeneic and syngeneic tumor models, which resulted in detection of tumors with remarkable specificity. We also imaged the infiltration of myeloid cells upon injection of complete Freund's adjuvant. Both anti-class II MHC and anti-CD11b VHHs detected inflammation with excellent specificity. Given the ease of manufacture and labeling of VHHs, we believe that this method could transform the manner in which antitumor responses and/or infectious events may be tracked.


Asunto(s)
Sistema Inmunológico/fisiología , Neoplasias/inmunología , Tomografía de Emisión de Positrones , Aminoaciltransferasas/fisiología , Animales , Anticuerpos/inmunología , Antineoplásicos/uso terapéutico , Proteínas Bacterianas/fisiología , Células de la Médula Ósea/metabolismo , Radioisótopos de Cobre/química , Cisteína Endopeptidasas/fisiología , Citometría de Flujo , Radioisótopos de Flúor/química , Adyuvante de Freund , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Inflamación , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Trasplante de Neoplasias , Neoplasias/terapia
9.
Arthritis Res Ther ; 14(1): R29, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22314006

RESUMEN

INTRODUCTION: Neuromedin U (NMU) is a neuropeptide with pro-inflammatory activity. The primary goal of this study was to determine if NMU promotes autoantibody-induced arthritis. Additional studies addressed the cellular source of NMU and sought to define the NMU receptor responsible for its pro-inflammatory effects. METHODS: Serum containing arthritogenic autoantibodies from K/BxN mice was used to induce arthritis in mice genetically lacking NMU. Parallel experiments examined whether NMU deficiency impacted the early mast-cell-dependent vascular leak response induced by these autoantibodies. Bone-marrow chimeric mice were generated to determine whether pro-inflammatory NMU is derived from hematopoietic cells or stromal cells. Mice lacking the known NMU receptors singly and in combination were used to determine susceptibility to serum-transferred arthritis and in vitro cellular responses to NMU. RESULTS: NMU-deficient mice developed less severe arthritis than control mice. Vascular leak was not affected by NMU deficiency. NMU expression by bone-marrow-derived cells mediated the pro-arthritogenic effect. Deficiency of all of the known NMU receptors, however, had no impact on arthritis severity and did not affect the ability of NMU to stimulate intracellular calcium flux. CONCLUSIONS: NMU-deficient mice are protected from developing autoantibody-induced inflammatory arthritis. NMU derived from hematopoietic cells, not neurons, promotes the development of autoantibody-induced inflammatory arthritis. This effect is mediated by a receptor other than the currently known NMU receptors.


Asunto(s)
Artritis/inmunología , Autoanticuerpos/inmunología , Neuropéptidos/inmunología , Receptores de Neurotransmisores/inmunología , Animales , Artritis/genética , Artritis/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Calcio/inmunología , Calcio/metabolismo , Femenino , Masculino , Mastocitos/inmunología , Mastocitos/metabolismo , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Neuropéptidos/deficiencia , Neuropéptidos/genética , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptores de Neurotensina/deficiencia , Receptores de Neurotensina/genética , Receptores de Neurotensina/inmunología , Receptores de Neurotransmisores/deficiencia , Receptores de Neurotransmisores/genética , Bazo/inmunología , Bazo/metabolismo , Bazo/patología
10.
Mol Imaging Biol ; 13(2): 314-20, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20559742

RESUMEN

PURPOSE: To study the effect of mammalian target of rapamycin (mTOR) inhibition on angiogenesis with magnetic resonance imaging (MRI) using magnetic iron oxide nanoparticles (MNP). PROCEDURES: One million CAK-1 renal cell carcinoma cells were subcutaneously implanted into each of 20 nude mice. When tumors reached ∼750 µl, four daily treatment arms began and continued for 4 weeks: rapamycin (mTOR inhibitor) 10 mg/kg/day; sorafenib (VEGF inhibitor) high dose (80 mg/kg/day) and low dose (30 mg/kg/day); and saline control. Weekly MRI (4.7 T Bruker Pharmascan) was performed before and after IV MION-48, a prototype MNP similar to MNP in clinical trials. Vascular volume fraction (VVF) was quantified as ΔR2 (from multi-contrast T2 sequences) and normalized to assumed muscle VVF of 3%. Linear regression compared VVF to microvascular density (MVD) as determined by histology. RESULTS: VVF correlated with MVD (R(2) = 0.95). VVF in all treatment arms differed from control (p < 0.05) and declined weekly with treatment. VVF changes with rapamycin were similar to high-dose sorafenib. CONCLUSION: This study demonstrates noninvasive, in vivo anti-angiogenic monitoring using MRI of mTOR inhibition.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Imagen por Resonancia Magnética/métodos , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Óxido Ferrosoférrico , Ratones , Ratones Desnudos , Microvasos/efectos de los fármacos , Microvasos/patología , Nanopartículas , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Trasplante Heterólogo
11.
Small ; 6(18): 2041-9, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20721949

RESUMEN

The synthesis and utility of a multimodal theranostic nanoagent based upon magnetofluorescent nanoparticles for the treatment of inflammatory atherosclerosis is described. These particles are modified with near-infrared fluorophores and light-activated therapeutic moieties, which allow for the optical determination of agent localization and phototoxic activation at spectrally distinct wavelengths. The resulting agent is readily taken up by murine macrophages in vitro and is highly phototoxic, with an LD(50) of 430 pM. Intravenous administration results in the localization of the nanoagent within macrophage-rich atherosclerotic lesions that can be imaged by intravital fluorescence microscopy. Irradiation of the atheroma with 650 nm light activates the therapeutic component and results in eradication of inflammatory macrophages, which may induce lesion stabilization. Importantly, these agents display limited skin photosensitivity, are highly efficacious, and provide an integrated imaging and therapeutic nanoplatform for atherosclerosis.


Asunto(s)
Aterosclerosis/terapia , Sistemas de Liberación de Medicamentos/métodos , Inflamación/terapia , Macrófagos/efectos de los fármacos , Nanoestructuras/uso terapéutico , Fototerapia/métodos , Animales , Apolipoproteínas E/genética , Aterosclerosis/complicaciones , Aterosclerosis/patología , Separación Celular/métodos , Células Cultivadas , Femenino , Inflamación/complicaciones , Inflamación/patología , Luz , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Nanoestructuras/administración & dosificación , Nanoestructuras/efectos de la radiación
12.
Nanomedicine ; 6(2): 362-70, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19800988

RESUMEN

A one-step process for the production of nanoparticles presenting advanced magnetic properties can be achieved using vapor condensation. In this article, we report on the fabrication of Fe particles covered by a uniform MgO epitaxial shell. MgO has a lower surface energy than Fe, which results in a core-shell crystal formation. The particles satisfy a few of technical requirements for the practical use in real clinics, such as a high biocompatibility in living cells in-vitro, an injection through blood vessels without any clothing problems in murine model, a high absorption rate for magnetic hyperthermia at small particle concentration, and the potential to be used as contrast agent in the field of diagnostic magnetic imaging. They are also able to be used in drug delivery and magnetic-activated cell sorting. FROM THE CLINICAL EDITOR: In this paper, the authors report on the synthesis of Fe particles covered by a uniform MgO epitaxial shell resulting in a core-shell crystal formation. The particles are proven to be useful as contrast agents for magnetic resonance imaging and have the potential to be useful as heating mediators for cancer therapy through hyperthermia. They also might be used in drug delivery and magnetic-activated cell sorting.


Asunto(s)
Medios de Contraste/química , Hipertermia Inducida/métodos , Hierro/química , Óxido de Magnesio/química , Imagen por Resonancia Magnética/métodos , Nanosferas/química , Cristalización/métodos , Composición de Medicamentos/métodos
13.
J Clin Invest ; 118(12): 4058-66, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19033674

RESUMEN

Eosinophils are multifunctional leukocytes that degrade and remodel tissue extracellular matrix through production of proteolytic enzymes, release of proinflammatory factors to initiate and propagate inflammatory responses, and direct activation of mucus secretion and smooth muscle cell constriction. Thus, eosinophils are central effector cells during allergic airway inflammation and an important clinical therapeutic target. Here we describe the use of an injectable MMP-targeted optical sensor that specifically and quantitatively resolves eosinophil activity in the lungs of mice with experimental allergic airway inflammation. Through the use of real-time molecular imaging methods, we report the visualization of eosinophil responses in vivo and at different scales. Eosinophil responses were seen at single-cell resolution in conducting airways using near-infrared fluorescence fiberoptic bronchoscopy, in lung parenchyma using intravital microscopy, and in the whole body using fluorescence-mediated molecular tomography. Using these real-time imaging methods, we confirmed the immunosuppressive effects of the glucocorticoid drug dexamethasone in the mouse model of allergic airway inflammation and identified a viridin-derived prodrug that potently inhibited the accumulation and enzyme activity of eosinophils in the lungs. The combination of sensitive enzyme-targeted sensors with noninvasive molecular imaging approaches permitted evaluation of airway inflammation severity and was used as a model to rapidly screen for new drug effects. Both fluorescence-mediated tomography and fiberoptic bronchoscopy techniques have the potential to be translated into the clinic.


Asunto(s)
Androstenos/farmacología , Antiinflamatorios/farmacología , Bacteriocinas/farmacología , Broncoscopía/métodos , Dexametasona/farmacología , Profármacos/farmacología , Hipersensibilidad Respiratoria/tratamiento farmacológico , Hipersensibilidad Respiratoria/patología , Tomografía Óptica/métodos , Androstenos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Bacteriocinas/uso terapéutico , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Eosinófilos/enzimología , Eosinófilos/patología , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/patología , Inflamación/fisiopatología , Pulmón/enzimología , Pulmón/patología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Contracción Muscular/efectos de los fármacos , Músculo Liso/enzimología , Músculo Liso/patología , Profármacos/uso terapéutico , Hipersensibilidad Respiratoria/enzimología , Hipersensibilidad Respiratoria/fisiopatología
14.
Nanomedicine (Lond) ; 2(2): 153-67, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17716118

RESUMEN

Magnetic nanoparticles and their magnetofluorescent analogues have become important tools for in vivo imaging using magnetic resonance imaging and fluorescent optical methods. A number of monodisperse magnetic nanoparticle preparations have been developed over the last decade for angiogenesis imaging, cancer staging, tracking of immune cells (monocyte/macrophage, T cells) and for molecular and cellular targeting. Phage display and data mining have enabled the procurement of novel tissue- or receptor-specific peptides, while high-throughput screening of diversity-oriented synthesis libraries has identified small molecules that permit or prevent uptake by specific cell types. Next-generation magnetic nanoparticles are expected to be truly multifunctional, incorporating therapeutic functionalities and further enhancing an already diverse repertoire of capabilities.


Asunto(s)
Medios de Contraste/química , Sistemas de Liberación de Medicamentos/métodos , Aumento de la Imagen/métodos , Magnetismo/uso terapéutico , Nanomedicina/tendencias , Nanopartículas/química , Nanopartículas/uso terapéutico
15.
Arthritis Rheum ; 56(1): 117-28, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17195214

RESUMEN

OBJECTIVE: The NF-kappaB signaling pathway promotes the immune response in rheumatoid arthritis (RA) and in rodent models of RA. NF-kappaB activity is regulated by the IKK-2 kinase during inflammatory responses. To elucidate how IKK-2 inhibition suppresses disease development, we used a combination of in vivo imaging, transcription profiling, and histopathology technologies to study mice with antibody-induced arthritis. METHODS: ML120B, a potent, small molecule inhibitor of IKK-2, was administered to arthritic animals, and disease activity was monitored. NF-kappaB activity in diseased joints was quantified by in vivo imaging. Quantitative reverse transcriptase-polymerase chain reaction was used to evaluate gene expression in joints. Protease-activated near-infrared fluorescence (NIRF) in vivo imaging was applied to assess the amounts of active proteases in the joints. RESULTS: Oral administration of ML120B suppressed both clinical and histopathologic manifestations of disease. In vivo imaging demonstrated that NF-kappaB activity in inflamed arthritic paws was inhibited by ML120B, resulting in significant suppression of multiple genes in the NF-kappaB pathway, i.e., KC, epithelial neutrophil-activating peptide 78, JE, intercellular adhesion molecule 1, CD3, CD68, tumor necrosis factor alpha, interleukin-1beta, interleukin-6, inducible nitric oxide synthase, cyclooxygenase 2, matrix metalloproteinase 3, cathepsin B, and cathepsin K. NIRF in vivo imaging demonstrated that ML120B treatment dramatically reduced the amount of active proteases in the joints. CONCLUSION: Our data demonstrate that IKK-2 inhibition in the murine model of antibody-induced arthritis suppresses both inflammation and joint destruction. In addition, this study highlights how gene expression profiling can facilitate the identification of surrogate biomarkers of disease activity and treatment response in an experimental model of arthritis.


Asunto(s)
Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Carbolinas/farmacología , Inhibidores Enzimáticos/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Niacinamida/análogos & derivados , Espectroscopía Infrarroja Corta/métodos , Administración Oral , Animales , Artritis Experimental/enzimología , Artritis Experimental/patología , Artritis Reumatoide/enzimología , Artritis Reumatoide/patología , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Articulaciones/efectos de los fármacos , Articulaciones/metabolismo , Articulaciones/patología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/metabolismo , Niacinamida/farmacología , ARN Mensajero/metabolismo , Espectrometría de Fluorescencia/métodos , Regulación hacia Arriba/efectos de los fármacos
16.
Science ; 312(5777): 1168-71, 2006 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-16728630

RESUMEN

Medical imaging technologies have undergone explosive growth over the past few decades and now play a central role in clinical oncology. But the truly transformative power of imaging in the clinical management of cancer patients lies ahead. Today, imaging is at a crossroads, with molecularly targeted imaging agents expected to broadly expand the capabilities of conventional anatomical imaging methods. Molecular imaging will allow clinicians to not only see where a tumor is located in the body, but also to visualize the expression and activity of specific molecules (e.g., proteases and protein kinases) and biological processes (e.g., apoptosis, angiogenesis, and metastasis) that influence tumor behavior and/or response to therapy. This information is expected to have a major impact on cancer detection, individualized treatment, and drug development, as well as our understanding of how cancer arises.


Asunto(s)
Diagnóstico por Imagen , Técnicas de Diagnóstico Molecular , Neoplasias/diagnóstico , Neoplasias/terapia , Radiofármacos , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Evaluación Preclínica de Medicamentos , Humanos , Indicadores y Reactivos , Neoplasias/química , Tomografía de Emisión de Positrones
17.
RNA ; 12(5): 797-806, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16556935

RESUMEN

We have recently described an RNA-only gene regulation system for mammalian cells in which inhibition of self-cleavage of an mRNA carrying ribozyme sequences provides the basis for control of gene expression. An important proof of principle for that system was provided by demonstrating the ability of one specific small molecule inhibitor of RNA self-cleavage, toyocamycin, to control gene expression in vitro and vivo. Here, we describe the development of the high-throughput screening (HTS) assay that led to the identification of toyocamycin and other molecules capable of inhibiting RNA self-cleavage in mammalian cells. To identify small molecules that can serve as inhibitors of ribozyme self-cleavage, we established a cell-based assay in which expression of a luciferase (luc) reporter is controlled by ribozyme sequences, and screened 58,076 compounds for their ability to induce luciferase expression. Fifteen compounds able to inhibit ribozyme self-cleavage in cells were identified through this screen. The most potent of the inhibitors identified were toyocamycin and 5-fluorouridine (FUR), nucleoside analogs carrying modifications of the 7-position and 5-position of the purine or pyrimidine bases. Individually, these two compounds were able to induce gene expression of the ribozyme-controlled reporter approximately 365-fold and 110-fold, respectively. Studies of the mechanism of action of the ribozyme inhibitors indicate that the compounds must be incorporated into RNA in order to inhibit RNA self-cleavage.


Asunto(s)
Inhibidores Enzimáticos/química , Regulación Enzimológica de la Expresión Génica , ARN Catalítico/antagonistas & inhibidores , Naranja de Acridina/química , Adenosina/análogos & derivados , Adenosina/química , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular , Citidina/análogos & derivados , Citidina/química , Evaluación Preclínica de Medicamentos , Etidio/química , Fluorouracilo/farmacología , Guanina/análogos & derivados , Guanina/química , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Toyocamicina/farmacología , Tubercidina/farmacología , Uridina/análogos & derivados , Uridina/química , beta-Galactosidasa/metabolismo
18.
Neoplasia ; 7(3): 234-40, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15799823

RESUMEN

The development of tumor vasculature is thought to occur through two complementary processes: sprouting angiogenesis from preexisting blood vessels of the host, and vasculogenesis, which involves the spontaneous development of vessels through specific recruitment, differentiation, and vascular incorporation of circulating endothelial cells (EC), endothelial progenitor cells (EPC), or potentially bone marrow-derived cells. Recent reports, however, have challenged the belief that bone marrow-derived cells contribute to tumor neovascularization, claiming an exclusive role for sprouting angiogenesis in tumor blood vessel development. In the present study, we explored the recruitment behavior of bone marrow-derived lin(-)c-kit(+)Sca-1+ stem cells to subcutaneously implanted Lewis lung carcinoma in a syngeneic bone marrow transplantation model. We observed that although lin(-)c-kit(+)Sca-1+ and their derived cells demonstrate significant recruitment to carcinomas in vivo, they do not appear to functionally contribute to tumor neovascularization. Furthermore, our results support the hypothesis that new vessel formation in carcinomas occurs primarily through endothelialization from adjacent and preexisting vasculature.


Asunto(s)
Antígenos Ly/genética , Antígenos Ly/fisiología , Células de la Médula Ósea/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Neovascularización Patológica , Oxiquinolina/análogos & derivados , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/fisiología , Animales , Médula Ósea/patología , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Carcinoma , Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular , Línea Celular Tumoral , Linaje de la Célula , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Trasplante de Neoplasias , Compuestos Organometálicos/química , Oxiquinolina/química , Células Madre/citología , Distribución Tisular
19.
Arthritis Res Ther ; 7(2): R310-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15743478

RESUMEN

A recently developed near-infrared fluorescence-labeled folate probe (NIR2-folate) was tested for in vivo imaging of arthritis using a lipopolysaccharide intra-articular injection model and a KRN transgenic mice serum induction mouse model. In the lipopolysaccharide injection model, the fluorescence signal intensity of NIR2-folate (n = 12) and of free NIR2 (n = 5) was compared between lipopolysaccharide-treated and control joints. The fluorescence signal intensity of the NIR2-folate probe at the inflammatory joints was found to be significantly higher than the control normal joints (up to 2.3-fold, P < 0.001). The NIR2-free dye injection group showed a persistent lower enhancement ratio than the NIR2-folate probe injection group. Excessive folic acid was also given to demonstrate a competitive effect with the NIR2-folate. In the KRN serum transfer model (n = 4), NIR2-folate was applied at different time points after serum transfer, and the inflamed joints could be detected as early as 30 hours after arthritogenic antibody transfer (1.8-fold increase in signal intensity). Fluorescence microscopy, histology, and immunohistochemistry validated the optical imaging results. We conclude that in vivo arthritis detection was feasible using a folate-targeted near-infrared fluorescence probe. This receptor-targeted imaging method may facilitate improved arthritis diagnosis and early assessment of the disease progress by providing an in vivo characterization of active macrophage status in inflammatory joint diseases.


Asunto(s)
Artritis Experimental/diagnóstico , Proteínas Portadoras/análisis , Colorantes Fluorescentes/análisis , Ácido Fólico/análogos & derivados , Activación de Macrófagos , Receptores de Superficie Celular/análisis , Espectroscopía Infrarroja Corta , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/patología , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Diagnóstico Precoz , Estudios de Factibilidad , Receptores de Folato Anclados a GPI , Ácido Fólico/análisis , Ácido Fólico/metabolismo , Procesamiento de Imagen Asistido por Computador , Técnicas para Inmunoenzimas , Lipopolisacáridos/toxicidad , Macrófagos/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Isoformas de Proteínas/análisis , Isoformas de Proteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Líquido Sinovial/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA