Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
2.
Circ Cardiovasc Imaging ; 17(4): e016876, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38626100
3.
ACS Chem Neurosci ; 15(9): 1882-1892, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38634759

RESUMEN

The sphingosine-1-phosphate receptor 1 (S1PR1) radiotracer [11C]CS1P1 has shown promise in proof-of-concept PET imaging of neuroinflammation in multiple sclerosis (MS). Our HPLC radiometabolite analysis of human plasma samples collected during PET scans with [11C]CS1P1 detected a radiometabolite peak that is more lipophilic than [11C]CS1P1. Radiolabeled metabolites that cross the blood-brain barrier complicate quantitative modeling of neuroimaging tracers; thus, characterizing such radiometabolites is important. Here, we report our detailed investigation of the metabolite profile of [11C]CS1P1 in rats, nonhuman primates, and humans. CS1P1 is a fluorine-containing ligand that we labeled with C-11 or F-18 for preclinical studies; the brain uptake was similar for both radiotracers. The same lipophilic radiometabolite found in human studies also was observed in plasma samples of rats and NHPs for CS1P1 labeled with either C-11 or F-18. We characterized the metabolite in detail using rats after injection of the nonradioactive CS1P1. To authenticate the molecular structure of this radiometabolite, we injected rats with 8 mg/kg of CS1P1 to collect plasma for solvent extraction and HPLC injection, followed by LC/MS analysis of the same metabolite. The LC/MS data indicated in vivo mono-oxidation of CS1P1 produces the metabolite. Subsequently, we synthesized three different mono-oxidized derivatives of CS1P1 for further investigation. Comparing the retention times of the mono-oxidized derivatives with the metabolite observed in rats injected with CS1P1 identified the metabolite as N-oxide 1, also named TZ82121. The MS fragmentation pattern of N-oxide 1 also matched that of the major metabolite in rat plasma. To confirm that metabolite TZ82121 does not enter the brain, we radiosynthesized [18F]TZ82121 by the oxidation of [18F]FS1P1. Radio-HPLC analysis confirmed that [18F]TZ82121 matched the radiometabolite observed in rat plasma post injection of [18F]FS1P1. Furthermore, the acute biodistribution study in SD rats and PET brain imaging in a nonhuman primate showed that [18F]TZ82121 does not enter the rat or nonhuman primate brain. Consequently, we concluded that the major lipophilic radiometabolite N-oxide [11C]TZ82121, detected in human plasma post injection of [11C]CS1P1, does not enter the brain to confound quantitative PET data analysis. [11C]CS1P1 is a promising S1PR1 radiotracer for detecting S1PR1 expression in the CNS.


Asunto(s)
Encéfalo , Tomografía de Emisión de Positrones , Radiofármacos , Animales , Humanos , Tomografía de Emisión de Positrones/métodos , Ratas , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagen , Radiofármacos/farmacocinética , Masculino , Receptores de Esfingosina-1-Fosfato/metabolismo , Ratas Sprague-Dawley , Radioisótopos de Flúor , Radioisótopos de Carbono
4.
J Nucl Med ; 65(5): 775-780, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38548349

RESUMEN

Tissue-resident macrophages are complementary to proinflammatory macrophages to promote the progression of atherosclerosis. The noninvasive detection of their presence and dynamic variation will be important to the understanding of their role in the pathogenesis of atherosclerosis. The goal of this study was to develop a targeted PET radiotracer for imaging CD163-positive (CD163+) macrophages in multiple mouse atherosclerosis models and assess the potential of CD163 as a biomarker for atherosclerosis in humans. Methods: CD163-binding peptide was identified using phage display and conjugated with a NODAGA chelator for 64Cu radiolabeling ([64Cu]Cu-ICT-01). CD163-overexpressing U87 cells were used to measure the binding affinity of [64Cu]Cu-ICT-01. Biodistribution studies were performed on wild-type C57BL/6 mice at multiple time points after tail vein injection. The sensitivity and specificity of [64Cu]Cu-ICT-01 in imaging CD163+ macrophages upregulated on the surface of atherosclerotic plaques were assessed in multiple mouse atherosclerosis models. Immunostaining, flow cytometry, and single-cell RNA sequencing were performed to characterize the expression of CD163 on tissue-resident macrophages. Human carotid atherosclerotic plaques were used to measure the expression of CD163+ resident macrophages and test the binding specificity of [64Cu]Cu-ICT-01. Results: [64Cu]Cu-ICT-01 showed high binding affinity to U87 cells. The biodistribution study showed rapid blood and renal clearance with low retention in all major organs at 1, 2, and 4 h after injection. In an ApoE-/- mouse model, [64Cu]Cu-ICT-01 demonstrated sensitive and specific detection of CD163+ macrophages and capability for tracking the progression of atherosclerotic lesions; these findings were further confirmed in Ldlr-/- and PCSK9 mouse models. Immunostaining showed elevated expression of CD163+ macrophages across the plaques. Flow cytometry and single-cell RNA sequencing confirmed the specific expression of CD163 on tissue-resident macrophages. Human tissue characterization demonstrated high expression of CD163+ macrophages on atherosclerotic lesions, and ex vivo autoradiography revealed specific binding of [64Cu]Cu-ICT-01 to human CD163. Conclusion: This work reported the development of a PET radiotracer binding CD163+ macrophages. The elevated expression of CD163+ resident macrophages on human plaques indicated the potential of CD163 as a biomarker for vulnerable plaques. The sensitivity and specificity of [64Cu]Cu-ICT-01 in imaging CD163+ macrophages warrant further investigation in translational settings.


Asunto(s)
Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Aterosclerosis , Macrófagos , Tomografía de Emisión de Positrones , Receptores de Superficie Celular , Animales , Ratones , Tomografía de Emisión de Positrones/métodos , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos CD/metabolismo , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/metabolismo , Macrófagos/metabolismo , Receptores de Superficie Celular/metabolismo , Humanos , Ratones Endogámicos C57BL , Radioisótopos de Cobre , Distribución Tisular , Radiofármacos/farmacocinética
5.
J Nucl Med ; 65(2): 287-293, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38176717

RESUMEN

The immune-fibrosis axis plays a critical role in cardiac remodeling after acute myocardial infarction. Imaging approaches to monitor temporal inflammation and fibroblast activation in mice have seen wide application in recent years. However, the repeatability of quantitative measurements remains challenging, particularly across multiple imaging centers. We aimed to determine reproducibility of quantitative inflammation and fibroblast activation images acquired at 2 facilities after myocardial infarction in mice. Methods: Mice underwent coronary artery ligation and sequential imaging with 68Ga-DOTA-ECL1i to assess chemokine receptor type 2 expression at 3 d after myocardial infarction and 68Ga-FAPI-46 to assess fibroblast activation protein expression at 7 d after myocardial infarction. Images were acquired at 1 center using either a local or a consensus protocol developed with the second center; the protocols differed in the duration of isoflurane anesthesia and the injected tracer dose. A second group of animals were scanned at the second site using the consensus protocol. Image analyses performed by each site and just by 1 site were also compared. Results: The uptake of 68Ga-DOTA-ECL1i in the infarct territory tended to be higher when the consensus protocol was used (P = 0.03). No difference was observed between protocol acquisitions for 68Ga-FAPI-46. Compared with the local protocol, the consensus protocol decreased variability between individual animals. When a matched consensus protocol was used, the 68Ga-DOTA-ECL1i infarct territory percentage injected dose per gram of tissue was higher on images acquired at site B than on those acquired at site A (P = 0.006). When normalized to body weight as SUV, this difference was mitigated. Both the percentage injected dose per gram of tissue and the SUV were comparable between sites for 68Ga-FAPI-46. Image analyses at the sites differed significantly, but this difference was mitigated when all images were analyzed at site A. Conclusion: The application of a standardized acquisition protocol may lower variability within datasets and facilitate comparison of molecular radiotracer distribution between preclinical imaging centers. Like clinical studies, multicenter preclinical studies should use centralized core-based image analysis to maximize reproducibility across sites.


Asunto(s)
Radioisótopos de Galio , Infarto del Miocardio , Ratones , Animales , Reproducibilidad de los Resultados , Infarto del Miocardio/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Inflamación , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos
6.
medRxiv ; 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37986880

RESUMEN

Abdominal aortic aneurysm (AAA) is a degenerative vascular disease impacting aging populations with a high mortality upon rupture. There are no effective medical therapies to prevent AAA expansion and rupture. We previously demonstrated the role of the monocyte chemoattractant protein-1 (MCP-1) / C-C chemokine receptor type 2 (CCR2) axis in rodent AAA pathogenesis via positron emission tomography/computed tomography (PET/CT) using CCR2 targeted radiotracer 64 Cu-DOTA-ECL1i. We have since translated this radiotracer into patients with AAA. CCR2 PET showed intense radiotracer uptake along the AAA wall in patients while little signal was observed in healthy volunteers. AAA tissues collected from individuals scanned with 64 Cu-DOTA-ECL1i and underwent open-repair later demonstrated more abundant CCR2+ cells compared to non-diseased aortas. We then used a CCR2 inhibitor (CCR2i) as targeted therapy in our established male and female rat AAA rupture models. We observed that CCR2i completely prevented AAA rupture in male rats and significantly decreased rupture rate in female AAA rats. PET/CT revealed substantial reduction of 64 Cu-DOTA-ECL1i uptake following CCR2i treatment in both rat models. Characterization of AAA tissues demonstrated decreased expression of CCR2+ cells and improved histopathological features. Taken together, our results indicate the potential of CCR2 as a theranostic biomarker for AAA management.

7.
Circ Cardiovasc Imaging ; 16(11): e016269, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37988444
8.
Circ Cardiovasc Imaging ; 16(11): e000081, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37916407

RESUMEN

Infiltrative cardiomyopathies comprise a broad spectrum of inherited or acquired conditions caused by deposition of abnormal substances within the myocardium. Increased wall thickness, inflammation, microvascular dysfunction, and fibrosis are the common pathological processes that lead to abnormal myocardial filling, chamber dilation, and disruption of conduction system. Advanced disease presents as heart failure and cardiac arrhythmias conferring poor prognosis. Infiltrative cardiomyopathies are often diagnosed late or misclassified as other more common conditions, such as hypertrophic cardiomyopathy, hypertensive heart disease, ischemic or other forms of nonischemic cardiomyopathies. Accurate diagnosis is also critical because clinical features, testing methodologies, and approach to treatment vary significantly even within the different types of infiltrative cardiomyopathies on the basis of the type of substance deposited. Substantial advances in noninvasive cardiac imaging have enabled accurate and early diagnosis. thereby eliminating the need for endomyocardial biopsy in most cases. This scientific statement discusses the role of contemporary multimodality imaging of infiltrative cardiomyopathies, including echocardiography, nuclear and cardiac magnetic resonance imaging in the diagnosis, prognostication, and assessment of response to treatment.


Asunto(s)
Cardiomiopatías , Insuficiencia Cardíaca , Humanos , American Heart Association , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/patología , Corazón , Miocardio/patología , Imagen por Resonancia Magnética
9.
Circ Cardiovasc Imaging ; 16(10): e016165, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37847763
10.
ACS Chem Neurosci ; 14(22): 4039-4050, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37882753

RESUMEN

Multiple sclerosis (MS) is an immune-mediated disease that is characterized by demyelination and inflammation in the central nervous system (CNS). Previous studies demonstrated that sphingosine-1-phosphate receptor (S1PR) modulators effectively inhibit S1PR1 in immune cell trafficking and reduce entry of pathogenic cells into the CNS. Studies have also implicated a nonimmune, inflammatory role of S1PR1 within the CNS in MS. In this study, we explored the expression of S1PR1 in the development and progression of demyelinating pathology of MS by quantitative assessment of S1PR1 expression using our S1PR1-specific radioligand, [3H]CS1P1, in the postmortem human CNS tissues including cortex, cerebellum, and spinal cord of MS cases and age- and sex-matched healthy cases. Immunohistochemistry with whole slide scanning for S1PR1 and various myelin proteins was also performed. Autoradiographic analysis using [3H]CS1P1 showed that the expression of S1PR1 was statistically significantly elevated in lesions compared to nonlesion regions in the MS cases, as well as normal healthy controls. The uptake of [3H]CS1P1 in the gray matter and nonlesion white matter did not significantly differ between healthy and MS CNS tissues. Saturation autoradiography analysis showed an increased binding affinity (Kd) of [3H]CS1P1 to S1PR1 in both gray matter and white matter of MS brains compared to healthy brains. Our blocking study using NIBR-0213, a S1PR1 antagonist, indicated [3H]CS1P1 is highly specific to S1PR1. Our findings demonstrated the activation of S1PR1 and an increased uptake of [3H]CS1P1 in the lesions of MS CNS. In summary, our quantitative autoradiography analysis using [3H]CS1P1 on human postmortem tissues shows the feasibility of novel imaging strategies for MS by targeting S1PR1.


Asunto(s)
Esclerosis Múltiple , Sustancia Blanca , Humanos , Esclerosis Múltiple/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Médula Espinal/metabolismo , Encéfalo/metabolismo , Sustancia Blanca/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
11.
Circ Cardiovasc Imaging ; 16(9): e016051, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37725671
13.
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.

14.
Circ Cardiovasc Imaging ; 16(8): e015913, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37582157
15.
Nucl Med Biol ; 122-123: 108370, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37556928

RESUMEN

BACKGROUND: Diabetes mellitus is a chronic progressive metabolic disorder that affects millions of people worldwide. Emerging evidence suggests the important roles of sphingolipid metabolism in diabetes. In particular, sphingosine-1-phosphate (S1P) and S1P receptor 2 (S1PR2) have important metabolic functions and are involved in several metabolic diseases. In diabetes, S1PR2 can effectively preserve ß cells and improve glucose/insulin tolerance in high-fat diet induced and streptozotocin (STZ)-induced diabetic mouse models. We previously developed a group of potent and selective S1PR2 ligands and radioligands. METHODS: In this study, we continued our efforts and characterized our leading S1PR2 radioligand, [11C]TZ34125, in a STZ-induced diabetic mouse model. [11C]TZ34125 was radiosynthesized in an automated synthesis module and in vitro saturation binding assay was performed using recombinant human S1PR2 membrane. In vitro saturation autoradiography analysis was also performed to determine the binding affinity of [11C]TZ34125 against mouse tissues. Type-1 diabetic mouse model was developed following a single high dose of STZ in C57BL/6 mice. Ex vivo biodistribution was performed to evaluate the distribution and amount of [11C]TZ34125 in tissues. In vitro autoradiography analysis was performed to compare the uptake of [11C]TZ34125 between diabetic and control animals in mouse spleen and pancreas. RESULTS: Our in vitro saturation binding assay using [11C]TZ34125 confirmed [11C]TZ34125 is a potent radioligand to recombinant human S1PR2 membrane with a Kd value of 0.9 nM. Saturation autoradiographic analysis showed [11C]TZ34125 has a Kd of 67.5, 45.9, and 25.0 nM to mouse kidney, spleen, and liver tissues respectively. Biodistribution study in STZ-induced diabetic mice showed the uptake of [11C]TZ34125 was significantly elevated in the spleen (~2 fold higher) and pancreas (~1.4 fold higher) compared to normal controls. The increased uptake of [11C]TZ34125 was further confirmed using autoradiographic analysis in the spleen and pancreases of STZ-induced diabetic mice, indicating S1PR2 can potentially act as a biomarker of diabetes in pancreases and inflammation in spleen. Future mechanistic analysis and in vivo quantitative assessment using non-invasive PET imaging in large animal model of diabetes is worthwhile. CONCLUSIONS: Overall, our data showed an increased uptake of our lead S1PR2-specific radioligand, [11C]TZ34125, in the spleen and pancreases of STZ-induced diabetic mice, and demonstrated [11C]TZ34125 has a great potential for preclinical and clinical usage for assessment of S1PR2 in diabetes and inflammation.


Asunto(s)
Diabetes Mellitus Experimental , Ratones , Humanos , Animales , Modelos Animales de Enfermedad , Estreptozocina/efectos adversos , Diabetes Mellitus Experimental/diagnóstico por imagen , Distribución Tisular , Ratones Endogámicos C57BL , Inflamación , Receptores de Esfingosina-1-Fosfato
16.
Circ Cardiovasc Imaging ; 16(7): 521-522, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37463239
17.
Circ Cardiovasc Imaging ; 16(6): e015724, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37339174
18.
Circ Cardiovasc Imaging ; 16(5): e015628, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37192297
19.
Med Phys ; 50(7): 4122-4137, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37010001

RESUMEN

BACKGROUND: Artificial intelligence-based methods have generated substantial interest in nuclear medicine. An area of significant interest has been the use of deep-learning (DL)-based approaches for denoising images acquired with lower doses, shorter acquisition times, or both. Objective evaluation of these approaches is essential for clinical application. PURPOSE: DL-based approaches for denoising nuclear-medicine images have typically been evaluated using fidelity-based figures of merit (FoMs) such as root mean squared error (RMSE) and structural similarity index measure (SSIM). However, these images are acquired for clinical tasks and thus should be evaluated based on their performance in these tasks. Our objectives were to: (1) investigate whether evaluation with these FoMs is consistent with objective clinical-task-based evaluation; (2) provide a theoretical analysis for determining the impact of denoising on signal-detection tasks; and (3) demonstrate the utility of virtual imaging trials (VITs) to evaluate DL-based methods. METHODS: A VIT to evaluate a DL-based method for denoising myocardial perfusion SPECT (MPS) images was conducted. To conduct this evaluation study, we followed the recently published best practices for the evaluation of AI algorithms for nuclear medicine (the RELAINCE guidelines). An anthropomorphic patient population modeling clinically relevant variability was simulated. Projection data for this patient population at normal and low-dose count levels (20%, 15%, 10%, 5%) were generated using well-validated Monte Carlo-based simulations. The images were reconstructed using a 3-D ordered-subsets expectation maximization-based approach. Next, the low-dose images were denoised using a commonly used convolutional neural network-based approach. The impact of DL-based denoising was evaluated using both fidelity-based FoMs and area under the receiver operating characteristic curve (AUC), which quantified performance on the clinical task of detecting perfusion defects in MPS images as obtained using a model observer with anthropomorphic channels. We then provide a mathematical treatment to probe the impact of post-processing operations on signal-detection tasks and use this treatment to analyze the findings of this study. RESULTS: Based on fidelity-based FoMs, denoising using the considered DL-based method led to significantly superior performance. However, based on ROC analysis, denoising did not improve, and in fact, often degraded detection-task performance. This discordance between fidelity-based FoMs and task-based evaluation was observed at all the low-dose levels and for different cardiac-defect types. Our theoretical analysis revealed that the major reason for this degraded performance was that the denoising method reduced the difference in the means of the reconstructed images and of the channel operator-extracted feature vectors between the defect-absent and defect-present cases. CONCLUSIONS: The results show the discrepancy between the evaluation of DL-based methods with fidelity-based metrics versus the evaluation on clinical tasks. This motivates the need for objective task-based evaluation of DL-based denoising approaches. Further, this study shows how VITs provide a mechanism to conduct such evaluations computationally, in a time and resource-efficient setting, and avoid risks such as radiation dose to the patient. Finally, our theoretical treatment reveals insights into the reasons for the limited performance of the denoising approach and may be used to probe the effect of other post-processing operations on signal-detection tasks.


Asunto(s)
Aprendizaje Profundo , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Inteligencia Artificial , Tomografía Computarizada de Emisión de Fotón Único/métodos , Redes Neurales de la Computación
20.
Circ Cardiovasc Imaging ; 16(4): e015520, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37071714
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...