Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
JACC Basic Transl Sci ; 8(7): 801-816, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37547068

RESUMEN

In the past 2 decades, research on atherosclerotic cardiovascular disease has uncovered inflammation to be a key driver of the pathophysiological process. A pressing need therefore exists to quantitatively and longitudinally probe inflammation, in preclinical models and in cardiovascular disease patients, ideally using non-invasive methods and at multiple levels. Here, we developed and employed in vivo multiparametric imaging approaches to investigate the immune response following myocardial infarction. The myocardial infarction models encompassed either transient or permanent left anterior descending coronary artery occlusion in C57BL/6 and Apoe-/-mice. We performed nanotracer-based fluorine magnetic resonance imaging and positron emission tomography (PET) imaging using a CD11b-specific nanobody and a C-C motif chemokine receptor 2-binding probe. We found that immune cell influx in the infarct was more pronounced in the permanent occlusion model. Further, using 18F-fluorothymidine and 18F-fluorodeoxyglucose PET, we detected increased hematopoietic activity after myocardial infarction, with no difference between the models. Finally, we observed persistent systemic inflammation and exacerbated atherosclerosis in Apoe-/- mice, regardless of which infarction model was used. Taken together, we showed the strengths and capabilities of multiparametric imaging in detecting inflammatory activity in cardiovascular disease, which augments the development of clinical readouts.

2.
Nat Biomed Eng ; 7(9): 1097-1112, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37291433

RESUMEN

Immunoparalysis is a compensatory and persistent anti-inflammatory response to trauma, sepsis or another serious insult, which increases the risk of opportunistic infections, morbidity and mortality. Here, we show that in cultured primary human monocytes, interleukin-4 (IL4) inhibits acute inflammation, while simultaneously inducing a long-lasting innate immune memory named trained immunity. To take advantage of this paradoxical IL4 feature in vivo, we developed a fusion protein of apolipoprotein A1 (apoA1) and IL4, which integrates into a lipid nanoparticle. In mice and non-human primates, an intravenously injected apoA1-IL4-embedding nanoparticle targets myeloid-cell-rich haematopoietic organs, in particular, the spleen and bone marrow. We subsequently demonstrate that IL4 nanotherapy resolved immunoparalysis in mice with lipopolysaccharide-induced hyperinflammation, as well as in ex vivo human sepsis models and in experimental endotoxemia. Our findings support the translational development of nanoparticle formulations of apoA1-IL4 for the treatment of patients with sepsis at risk of immunoparalysis-induced complications.


Asunto(s)
Interleucina-4 , Sepsis , Humanos , Animales , Ratones , Interleucina-4/metabolismo , Inmunidad Entrenada , Monocitos
3.
Sci Rep ; 12(1): 6185, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35418569

RESUMEN

In recent years, cardiovascular immuno-imaging by positron emission tomography (PET) has undergone tremendous progress in preclinical settings. Clinically, two approved PET tracers hold great potential for inflammation imaging in cardiovascular patients, namely FDG and DOTATATE. While the former is a widely applied metabolic tracer, DOTATATE is a relatively new PET tracer targeting the somatostatin receptor 2 (SST2). In the current study, we performed a detailed, head-to-head comparison of DOTATATE-based radiotracers and [18F]F-FDG in mouse and rabbit models of cardiovascular inflammation. For mouse experiments, we labeled DOTATATE with the long-lived isotope [64Cu]Cu to enable studying the tracer's mode of action by complementing in vivo PET/CT experiments with thorough ex vivo immunological analyses. For translational PET/MRI rabbit studies, we employed the more widely clinically used [68Ga]Ga-labeled DOTATATE, which was approved by the FDA in 2016. DOTATATE's pharmacokinetics and timed biodistribution were determined in control and atherosclerotic mice and rabbits by ex vivo gamma counting of blood and organs. Additionally, we performed in vivo PET/CT experiments in mice with atherosclerosis, mice subjected to myocardial infarction and control animals, using both [64Cu]Cu-DOTATATE and [18F]F-FDG. To evaluate differences in the tracers' cellular specificity, we performed ensuing ex vivo flow cytometry and gamma counting. In mice subjected to myocardial infarction, in vivo [64Cu]Cu-DOTATATE PET showed higher differential uptake between infarcted (SUVmax 1.3, IQR, 1.2-1.4, N = 4) and remote myocardium (SUVmax 0.7, IQR, 0.5-0.8, N = 4, p = 0.0286), and with respect to controls (SUVmax 0.6, IQR, 0.5-0.7, N = 4, p = 0.0286), than [18F]F-FDG PET. In atherosclerotic mice, [64Cu]Cu-DOTATATE PET aortic signal, but not [18F]F-FDG PET, was higher compared to controls (SUVmax 1.1, IQR, 0.9-1.3 and 0.5, IQR, 0.5-0.6, respectively, N = 4, p = 0.0286). In both models, [64Cu]Cu-DOTATATE demonstrated preferential accumulation in macrophages with respect to other myeloid cells, while [18F]F-FDG was taken up by macrophages and other leukocytes. In a translational PET/MRI study in atherosclerotic rabbits, we then compared [68Ga]Ga-DOTATATE and [18F]F-FDG for the assessment of aortic inflammation, combined with ex vivo radiometric assays and near-infrared imaging of macrophage burden. Rabbit experiments showed significantly higher aortic accumulation of both [68Ga]Ga-DOTATATE and [18F]F-FDG in atherosclerotic (SUVmax 0.415, IQR, 0.338-0.499, N = 32 and 0.446, IQR, 0.387-0.536, N = 27, respectively) compared to control animals (SUVmax 0.253, IQR, 0.197-0.285, p = 0.0002, N = 10 and 0.349, IQR, 0.299-0.423, p = 0.0159, N = 11, respectively). In conclusion, we present a detailed, head-to-head comparison of the novel SST2-specific tracer DOTATATE and the validated metabolic tracer [18F]F-FDG for the evaluation of inflammation in small animal models of cardiovascular disease. Our results support further investigations on the use of DOTATATE to assess cardiovascular inflammation as a complementary readout to the widely used [18F]F-FDG.


Asunto(s)
Aterosclerosis , Infarto del Miocardio , Compuestos Organometálicos , Animales , Aterosclerosis/diagnóstico por imagen , Fluorodesoxiglucosa F18/metabolismo , Radioisótopos de Galio , Humanos , Inflamación/diagnóstico por imagen , Ratones , Infarto del Miocardio/diagnóstico por imagen , Compuestos Organometálicos/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones/métodos , Conejos , Cintigrafía , Radiofármacos , Distribución Tisular
4.
STAR Protoc ; 2(2): 100434, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33899016

RESUMEN

Noninvasive immunoimaging holds great potential for studying and stratifying disease as well as therapeutic efficacy. Radiolabeled single-domain antibody fragments (i.e., nanobodies) are appealing probes for immune landscape profiling, as they display high stability, rapid targeting, and excellent specificity, while allowing extremely sensitive nuclear readouts. Here, we present a protocol for radiolabeling an anti-CD11b nanobody and studying its uptake in mice by a combination of positron emission tomography imaging, ex vivo gamma counting, and autoradiography. Our protocol is applicable to nanobodies against other antigens. For complete details on the use and execution of this protocol, please see Priem et al. (2020), Senders et al. (2019), or Rashidian et al. (2017).


Asunto(s)
Técnicas Inmunológicas/métodos , Tomografía de Emisión de Positrones/métodos , Anticuerpos de Dominio Único , Animales , Técnicas Histológicas , Ratones , Imagen Molecular/métodos , Especificidad de Órganos , Anticuerpos de Dominio Único/análisis , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo
6.
Sci Adv ; 7(10)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33674313

RESUMEN

Immunotherapies controlling the adaptive immune system are firmly established, but regulating the innate immune system remains much less explored. The intrinsic interactions between nanoparticles and phagocytic myeloid cells make these materials especially suited for engaging the innate immune system. However, developing nanotherapeutics is an elaborate process. Here, we demonstrate a modular approach that facilitates efficiently incorporating a broad variety of drugs in a nanobiologic platform. Using a microfluidic formulation strategy, we produced apolipoprotein A1-based nanobiologics with favorable innate immune system-engaging properties as evaluated by in vivo screening. Subsequently, rapamycin and three small-molecule inhibitors were derivatized with lipophilic promoieties, ensuring their seamless incorporation and efficient retention in nanobiologics. A short regimen of intravenously administered rapamycin-loaded nanobiologics (mTORi-NBs) significantly prolonged allograft survival in a heart transplantation mouse model. Last, we studied mTORi-NB biodistribution in nonhuman primates by PET/MR imaging and evaluated its safety, paving the way for clinical translation.


Asunto(s)
Sistema Inmunológico , Nanopartículas , Animales , Inmunoterapia , Ratones , Sirolimus/farmacología , Distribución Tisular
7.
Curr Opin Chem Biol ; 63: 78-85, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33735814

RESUMEN

Nanomedicine research is an active field that produces thousands of studies every year. However, translation of nanotherapeutics to the clinic has yet to catch up with such a vast output. In recent years, the need to better understand nanomedicines' in vivo behavior has been identified as one of the major challenges for efficient clinical translation. In this context, noninvasive imaging offers attractive solutions to provide valuable information about nanomedicine biodistribution, pharmacokinetics, stability, or therapeutic efficacy. Here, we review the latest imaging approaches used in the development of therapeutic nanomedicines, discuss why these strategies bring added value along the translational pipeline, and give a perspective on future advances in the field.


Asunto(s)
Portadores de Fármacos/química , Indicadores y Reactivos/química , Nanopartículas/química , Animales , Portadores de Fármacos/farmacocinética , Humanos , Imagen por Resonancia Magnética , Terapia Molecular Dirigida , Nanomedicina , Imagen Óptica , Distribución Tisular , Tomografía Computarizada de Emisión
8.
Sci Transl Med ; 13(584)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692130

RESUMEN

Macrophages play a central role in the pathogenesis of atherosclerosis. The inflammatory properties of these cells are dictated by their metabolism, of which the mechanistic target of rapamycin (mTOR) signaling pathway is a key regulator. Using myeloid cell-specific nanobiologics in apolipoprotein E-deficient (Apoe -/-) mice, we found that targeting the mTOR and ribosomal protein S6 kinase-1 (S6K1) signaling pathways rapidly diminished plaque macrophages' inflammatory activity. By investigating transcriptome modifications, we identified Psap, a gene encoding the lysosomal protein prosaposin, as closely related with mTOR signaling. Subsequent in vitro experiments revealed that Psap inhibition suppressed both glycolysis and oxidative phosphorylation. Transplantation of Psap -/- bone marrow to low-density lipoprotein receptor knockout (Ldlr -/-) mice led to a reduction in atherosclerosis development and plaque inflammation. Last, we confirmed the relationship between PSAP expression and inflammation in human carotid atherosclerotic plaques. Our findings provide mechanistic insights into the development of atherosclerosis and identify prosaposin as a potential therapeutic target.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Saposinas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE
9.
Sci Transl Med ; 12(568)2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33148623

RESUMEN

Acute bacterial endocarditis is a rapid, difficult to manage, and frequently lethal disease. Potent antibiotics often cannot efficiently kill Staphylococcus aureus that colonizes the heart's valves. S. aureus relies on virulence factors to evade therapeutics and the host's immune response, usurping the host's clotting system by activating circulating prothrombin with staphylocoagulase and von Willebrand factor-binding protein. An insoluble fibrin barrier then forms around the bacterial colony, shielding the pathogen from immune cell clearance. Targeting virulence factors may provide previously unidentified avenues to better diagnose and treat endocarditis. To tap into this unused therapeutic opportunity, we codeveloped therapeutics and multimodal molecular imaging to probe the host-pathogen interface. We introduced and validated a family of small-molecule optical and positron emission tomography (PET) reporters targeting active thrombin in the fibrin-rich environment of bacterial colonies. The imaging agents, based on the clinical thrombin inhibitor dabigatran, are bound to heart valve vegetations in mice. Using optical imaging, we monitored therapy with antibodies neutralizing staphylocoagulase and von Willebrand factor-binding protein in mice with S. aureus endocarditis. This treatment deactivated bacterial defenses against innate immune cells, decreased in vivo imaging signal, and improved survival. Aortic or tricuspid S. aureus endocarditis in piglets was also successfully imaged with clinical PET/magnetic resonance imaging. Our data map a route toward adjuvant immunotherapy for endocarditis and provide efficient tools to monitor this drug class for infectious diseases.


Asunto(s)
Endocarditis Bacteriana , Infecciones Estafilocócicas , Animales , Coagulasa , Endocarditis Bacteriana/diagnóstico por imagen , Endocarditis Bacteriana/tratamiento farmacológico , Ratones , Imagen Multimodal , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus , Porcinos
10.
Cell ; 183(3): 786-801.e19, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125893

RESUMEN

Trained immunity, a functional state of myeloid cells, has been proposed as a compelling immune-oncological target. Its efficient induction requires direct engagement of myeloid progenitors in the bone marrow. For this purpose, we developed a bone marrow-avid nanobiologic platform designed specifically to induce trained immunity. We established the potent anti-tumor capabilities of our lead candidate MTP10-HDL in a B16F10 mouse melanoma model. These anti-tumor effects result from trained immunity-induced myelopoiesis caused by epigenetic rewiring of multipotent progenitors in the bone marrow, which overcomes the immunosuppressive tumor microenvironment. Furthermore, MTP10-HDL nanotherapy potentiates checkpoint inhibition in this melanoma model refractory to anti-PD-1 and anti-CTLA-4 therapy. Finally, we determined MTP10-HDL's favorable biodistribution and safety profile in non-human primates. In conclusion, we show that rationally designed nanobiologics can promote trained immunity and elicit a durable anti-tumor response either as a monotherapy or in combination with checkpoint inhibitor drugs.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Nanotecnología , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animales , Conducta Animal , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Colesterol/metabolismo , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunidad/efectos de los fármacos , Inmunoterapia , Lipoproteínas HDL/metabolismo , Ratones Endogámicos C57BL , Primates , Distribución Tisular/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
11.
Circ Cardiovasc Imaging ; 13(10): e010586, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33076700

RESUMEN

BACKGROUND: Macrophages, innate immune cells that reside in all organs, defend the host against infection and injury. In the heart and vasculature, inflammatory macrophages also enhance tissue damage and propel cardiovascular diseases. METHODS: We here use in vivo positron emission tomography (PET) imaging, flow cytometry, and confocal microscopy to evaluate quantitative noninvasive assessment of cardiac, arterial, and pulmonary macrophages using the nanotracer 64Cu-Macrin-a 20-nm spherical dextran nanoparticle assembled from nontoxic polyglucose. RESULTS: PET imaging using 64Cu-Macrin faithfully reported accumulation of macrophages in the heart and lung of mice with myocardial infarction, sepsis, or pneumonia. Flow cytometry and confocal microscopy detected the near-infrared fluorescent version of the nanoparticle (VT680Macrin) primarily in tissue macrophages. In 5-day-old mice, 64Cu-Macrin PET imaging quantified physiologically more numerous cardiac macrophages. Upon intravenous administration of 64Cu-Macrin in rabbits and pigs, we detected heightened macrophage numbers in the infarcted myocardium, inflamed lung regions, and atherosclerotic plaques using a clinical PET/magnetic resonance imaging scanner. Toxicity studies in rats and human dosimetry estimates suggest that 64Cu-Macrin is safe for use in humans. CONCLUSIONS: Taken together, these results indicate 64Cu-Macrin could serve as a facile PET nanotracer to survey spatiotemporal macrophage dynamics during various physiological and pathological conditions. 64Cu-Macrin PET imaging could stage inflammatory cardiovascular disease activity, assist disease management, and serve as an imaging biomarker for emerging macrophage-targeted therapeutics.


Asunto(s)
Radioisótopos de Cobre , Dextranos , Corazón/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Macrófagos/patología , Imagen Molecular , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radiofármacos , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/patología , Radioisótopos de Cobre/administración & dosificación , Radioisótopos de Cobre/farmacocinética , Dextranos/administración & dosificación , Dextranos/farmacocinética , Modelos Animales de Enfermedad , Inyecciones Intravenosas , Pulmón/patología , Macrófagos Alveolares/patología , Ratones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Nanopartículas , Neumonía/diagnóstico por imagen , Neumonía/patología , Valor Predictivo de las Pruebas , Conejos , Radiofármacos/administración & dosificación , Radiofármacos/farmacocinética , Porcinos , Porcinos Enanos , Factores de Tiempo
12.
Adv Drug Deliv Rev ; 154-155: 123-141, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32721459

RESUMEN

Nanomedicine approaches can effectively modulate the biodistribution and bioavailability of therapeutic agents, improving their therapeutic index. However, despite the ever-increasing amount of literature reporting on preclinical nanomedicine, the number of nanotherapeutics receiving FDA approval remains relatively low. Several barriers exist that hamper the effective preclinical evaluation and clinical translation of nanotherapeutics. Key barriers include insufficient understanding of nanomedicines' in vivo behavior, inadequate translation from murine models to larger animals, and a lack of patient stratification strategies. Integrating quantitative non-invasive imaging techniques in nanomedicine development offers attractive possibilities to address these issues. Among the available imaging techniques, nuclear imaging by positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are highly attractive in this context owing to their quantitative nature and uncontested sensitivity. In basic and translational research, nuclear imaging techniques can provide critical quantitative information about pharmacokinetic parameters, biodistribution profiles or target site accumulation of nanocarriers and their associated payload. During clinical evaluation, nuclear imaging can be used to select patients amenable to nanomedicine treatment. Here, we review how nuclear imaging-based approaches are increasingly being integrated into nanomedicine development and discuss future developments that will accelerate their clinical translation.


Asunto(s)
Nanomedicina , Tomografía de Emisión de Positrones , Tomografía Computarizada de Emisión de Fotón Único , Animales , Humanos , Investigación Biomédica Traslacional
13.
ACS Nano ; 14(7): 7832-7846, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32413260

RESUMEN

Although the first nanomedicine was clinically approved more than two decades ago, nanoparticles' (NP) in vivo behavior is complex and the immune system's role in their application remains elusive. At present, only passive-targeting nanoformulations have been clinically approved, while more complicated active-targeting strategies typically fail to advance from the early clinical phase stage. This absence of clinical translation is, among others, due to the very limited understanding for in vivo targeting mechanisms. Dynamic in vivo phenomena such as NPs' real-time targeting kinetics and phagocytes' contribution to active NP targeting remain largely unexplored. To better understand in vivo targeting, monitoring NP accumulation and distribution at complementary levels of spatial and temporal resolution is imperative. Here, we integrate in vivo positron emission tomography/computed tomography imaging with intravital microscopy and flow cytometric analyses to study αvß3-integrin-targeted cyclic arginine-glycine-aspartate decorated liposomes and oil-in-water nanoemulsions in tumor mouse models. We observed that ligand-mediated accumulation in cancerous lesions is multifaceted and identified "NP hitchhiking" with phagocytes to contribute considerably to this intricate process. We anticipate that this understanding can facilitate rational improvement of nanomedicine applications and that immune cell-NP interactions can be harnessed to develop clinically viable nanomedicine-based immunotherapies.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Integrina alfaV , Integrina alfaVbeta3 , Lípidos , Ratones , Neoplasias/tratamiento farmacológico , Fagocitos
14.
Nat Nanotechnol ; 15(5): 398-405, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32313216

RESUMEN

Ischaemic heart disease evokes a complex immune response. However, tools to track the systemic behaviour and dynamics of leukocytes non-invasively in vivo are lacking. Here, we present a multimodal hot-spot imaging approach using an innovative high-density lipoprotein-derived nanotracer with a perfluoro-crown ether payload (19F-HDL) to allow myeloid cell tracking by 19F magnetic resonance imaging. The 19F-HDL nanotracer can additionally be labelled with zirconium-89 and fluorophores to detect myeloid cells by in vivo positron emission tomography imaging and optical modalities, respectively. Using our nanotracer in atherosclerotic mice with myocardial infarction, we observed rapid myeloid cell egress from the spleen and bone marrow by in vivo 19F-HDL magnetic resonance imaging. Concurrently, using ex vivo techniques, we showed that circulating pro-inflammatory myeloid cells accumulated in atherosclerotic plaques and at the myocardial infarct site. Our multimodality imaging approach is a valuable addition to the immunology toolbox, enabling the study of complex myeloid cell behaviour dynamically.


Asunto(s)
Células Mieloides/patología , Isquemia Miocárdica/diagnóstico por imagen , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/patología , Rastreo Celular/métodos , Éteres Corona/análisis , Femenino , Colorantes Fluorescentes/análisis , Flúor/análisis , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Imagen Multimodal/métodos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Isquemia Miocárdica/patología , Imagen Óptica/métodos , Tomografía de Emisión de Positrones/métodos , Radioisótopos/análisis , Circonio/análisis
15.
Arterioscler Thromb Vasc Biol ; 40(5): 1123-1134, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32237905

RESUMEN

Cardiovascular disease due to atherosclerosis is still the main cause of morbidity and mortality worldwide. This disease is a complex systemic disorder arising from a network of pathological processes within the arterial vessel wall, and, outside of the vasculature, in the hematopoietic system and organs involved in metabolism. Recent years have seen tremendous efforts in the development and validation of quantitative imaging technologies for the noninvasive evaluation of patients with atherosclerotic cardiovascular disease. Specifically, the advent of combined positron emission tomography and magnetic resonance imaging scanners has opened new exciting opportunities in cardiovascular imaging. In this review, we will describe how combined positron emission tomography/magnetic resonance imaging scanners can be leveraged to evaluate atherosclerotic cardiovascular disease at the whole-body level, with specific focus on preclinical animal models of disease, from mouse to nonhuman primates. We will broadly describe 3 major areas of application: (1) vascular imaging, for advanced atherosclerotic plaque phenotyping and evaluation of novel imaging tracers or therapeutic interventions; (2) assessment of the ischemic heart and brain; and (3) whole-body imaging of the hematopoietic system. Finally, we will provide insights on potential novel technical developments which may further increase the relevance of integrated positron emission tomography/magnetic resonance imaging in preclinical atherosclerosis studies.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Imagen por Resonancia Magnética , Tomografía de Emisión de Positrones , Imagen de Cuerpo Entero/métodos , Animales , Aterosclerosis/patología , Aterosclerosis/terapia , Modelos Animales de Enfermedad , Diseño de Equipo , Imagen por Resonancia Magnética/instrumentación , Ratones , Imagen Multimodal , Tomografía de Emisión de Positrones/instrumentación , Valor Predictivo de las Pruebas , Primates , Reproducibilidad de los Resultados , Imagen de Cuerpo Entero/instrumentación
16.
Arterioscler Thromb Vasc Biol ; 40(4): 865-873, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32078338

RESUMEN

The immune system's role in atherosclerosis has long been an important research topic and is increasingly investigated for therapeutic and diagnostic purposes. Therefore, noninvasive imaging of hematopoietic organs and immune cells will undoubtedly improve atherosclerosis phenotyping and serve as a monitoring method for immunotherapeutic treatments. Among the available imaging techniques, positron emission tomography's unique features make it an ideal tool to quantitatively image the immune response in the context of atherosclerosis and afford reliable readouts to guide medical interventions in cardiovascular disease. Here, we summarize the state of the art in the field of atherosclerosis positron emission tomography immunoimaging and provide an outlook on current and future applications.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Aterosclerosis/inmunología , Tomografía de Emisión de Positrones/métodos , Aterosclerosis/metabolismo , Sistema Hematopoyético/diagnóstico por imagen , Humanos , Nanopartículas , Fagocitos , Placa Aterosclerótica/diagnóstico por imagen , Radioinmunodetección , Radiofármacos
17.
J Nucl Med ; 61(3): 433-436, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31420495

RESUMEN

The immune function within the tumor microenvironment has become a prominent therapeutic target, with tumor-associated macrophages (TAMs) playing a critical role in immune suppression. We propose an 89Zr-labeled high-density lipoprotein (89Zr-HDL) nanotracer as a means of monitoring response to immunotherapy. Methods: Female MMTV-PyMT mice were treated with pexidartinib, a colony-stimulating factor 1 receptor (CSF1R) inhibitor, to reduce TAM density. The accumulation of 89Zr-HDL within the tumor was assessed using PET/CT imaging and autoradiography, whereas TAM burden was determined using immunofluorescence. Results: A significant reduction in 89Zr-HDL accumulation was observed in PET/CT images, with 2.9% ± 0.3% and 3.7% ± 0.2% injected dose/g for the pexidartinib- and vehicle-treated mice, respectively. This reduction was corroborated ex vivo and correlated with decreased TAM density. Conclusion: These results support the potential use of 89Zr-HDL nanoparticles as a PET tracer to quickly monitor the response to CSF1R inhibitors and other therapeutic strategies targeting TAMs.


Asunto(s)
Lipoproteínas HDL/química , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radioisótopos/química , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Circonio/química , Aminopiridinas/farmacología , Animales , Femenino , Lipoproteínas HDL/farmacocinética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Pirroles/farmacología , Trazadores Radiactivos , Distribución Tisular
18.
Bioconjug Chem ; 31(2): 360-368, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31095372

RESUMEN

Nanotherapy has recently emerged as an experimental treatment option for atherosclerosis. To fulfill its promise, robust noninvasive imaging approaches for subject selection and treatment evaluation are warranted. To that end, we present here a positron emission tomography (PET)-based method for quantification of liposomal nanoparticle uptake in the atherosclerotic vessel wall. We evaluated a modular procedure to label liposomal nanoparticles with the radioisotope zirconium-89 (89Zr). Their biodistribution and vessel wall targeting in a rabbit atherosclerosis model was evaluated up to 15 days after intravenous injection by PET/computed tomography (CT) and PET/magnetic resonance imaging (PET/MRI). Vascular permeability was assessed in vivo using three-dimensional dynamic contrast-enhanced MRI (3D DCE-MRI) and ex vivo using near-infrared fluorescence (NIRF) imaging. The 89Zr-radiolabeled liposomes displayed a biodistribution pattern typical of long-circulating nanoparticles. Importantly, they markedly accumulated in atherosclerotic lesions in the abdominal aorta, as evident on PET/MRI and confirmed by autoradiography, and this uptake moderately correlated with vascular permeability. The method presented herein facilitates the development of nanotherapy for atherosclerotic disease as it provides a tool to screen for nanoparticle targeting in individual subjects' plaques.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Liposomas/análisis , Placa Aterosclerótica/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radioisótopos/análisis , Circonio/análisis , Animales , Aorta Abdominal/diagnóstico por imagen , Masculino , Conejos , Distribución Tisular
19.
Sci Transl Med ; 11(506)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434756

RESUMEN

Nanomedicine research produces hundreds of studies every year, yet very few formulations have been approved for clinical use. This is due in part to a reliance on murine studies, which have limited value in accurately predicting translational efficacy in larger animal models and humans. Here, we report the scale-up of a nanoimmunotherapy from mouse to large rabbit and porcine atherosclerosis models, with an emphasis on the solutions we implemented to overcome production and evaluation challenges. Specifically, we integrated translational imaging readouts within our workflow to both analyze the nanoimmunotherapeutic's in vivo behavior and assess treatment response in larger animals. We observed our nanoimmunotherapeutic's anti-inflammatory efficacy in mice, as well as rabbits and pigs. Nanoimmunotherapy-mediated reduction of inflammation in the large animal models halted plaque progression, supporting the approach's translatability and potential to acutely treat atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Aterosclerosis/terapia , Imagenología Tridimensional , Inmunoterapia , Nanomedicina , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/toxicidad , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Tomografía de Emisión de Positrones , Conejos , Simvastatina/farmacología , Simvastatina/uso terapéutico , Especificidad de la Especie , Porcinos , Distribución Tisular
20.
Nat Rev Cardiol ; 16(1): 21-32, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30209355

RESUMEN

Atherosclerosis is a chronic disease of the large arteries and the underlying cause of myocardial infarction and stroke. Atherosclerosis is driven by cholesterol accumulation and subsequent inflammation in the vessel wall. Despite the clinical successes of lipid-lowering treatments, atherosclerosis remains one of the major threats to human health worldwide. Over the past 20 years, insights into cardiovascular immunopathology have provided a plethora of new potential therapeutic targets to reduce the risk of atherosclerosis and have shifted the therapeutic focus from lipids to inflammation. In 2017, the CANTOS trial demonstrated for the first time the beneficial effects of targeting inflammation to treat cardiovascular disease by showing that IL-1ß inhibition can reduce the recurrence rate of cardiovascular events in a large cohort of patients. At the same time, preclinical studies have highlighted nanotechnology approaches that facilitate the specific targeting of innate immune cells, which could potentially generate more effective immunomodulatory treatments to induce disease regression and prevent the recurrence of cardiovascular events. The clinical translation of such nanoimmunotherapies and their application to treat patients with ischaemic heart disease are challenges that lie ahead.


Asunto(s)
Antiinflamatorios/uso terapéutico , Aterosclerosis/tratamiento farmacológico , Inmunoterapia/métodos , Mediadores de Inflamación/antagonistas & inhibidores , Isquemia Miocárdica/tratamiento farmacológico , Nanomedicina/métodos , Animales , Aterosclerosis/diagnóstico , Aterosclerosis/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Mediadores de Inflamación/inmunología , Terapia Molecular Dirigida , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/inmunología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...