Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Theranostics ; 11(2): 491-505, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391488

RESUMEN

Patients with neuroendocrine tumors (NETs) can be treated with peptide receptor radionuclide therapy (PRRT). Here, the somatostatin analogue octreotate radiolabeled with lutetium-177 is targeted to NET cells by binding to the somatostatin receptor subtype 2 (SST2). During radioactive decay, DNA damage is induced, leading to NET cell death. Although the therapy proves to be effective, mortality rates remain high. To appropriately select more optimal treatment strategies, it is essential to first better understand the radiobiological responses of tumor cells to PRRT. Methods: We analyzed PRRT induced radiobiological responses in SST2 expressing cells and xenografted mice using SPECT/MRI scanning and histological and molecular analyses. We measured [177Lu]Lu-DOTA-TATE uptake and performed analyses to visualize induction of DNA damage, cell death and other cellular characteristics. Results: The highest accumulation of radioactivity was measured in the tumor and kidneys. PRRT induced DNA damage signaling and repair in a time-dependent manner. We observed intra-tumor heterogeneity of DNA damage and apoptosis, which was not attributed to proliferation or bioavailability. We found a strong correlation between high DNA damage levels and high SST2 expression. PRRT elicited a different therapeutic response between models with different SST2 expression levels. Heterogeneous SST2 expression levels were also confirmed in patient NETs. Conclusion: Heterogeneous SST2 expression levels within NETs cause differentially induced DNA damage levels, influence recurrent tumor phenotypes and impact the therapeutic response in different models and potentially in patients. Our results contribute to a better understanding of PRRT effects, which might impact future therapeutic outcome of NET patients.


Asunto(s)
Complejos de Coordinación/uso terapéutico , Octreótido/análogos & derivados , Neoplasias Pancreáticas/radioterapia , Radiofármacos/uso terapéutico , Receptores de Somatostatina/metabolismo , Animales , Apoptosis , Proliferación Celular , Complejos de Coordinación/farmacocinética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Octreótido/farmacocinética , Octreótido/uso terapéutico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Radiofármacos/farmacocinética , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Imaging Biol ; 20(6): 973-983, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29556947

RESUMEN

PURPOSE: The gastrin-releasing peptide receptor (GRPR), overexpressed on various tumor types, is an attractive target for receptor-mediated imaging and therapy. Another interesting approach would be the use of GRPR radioligands for pre-operative imaging and subsequent radio-guided surgery, with the goal to improve surgical outcome. GRPR radioligands were successfully implemented in clinical studies, especially Sarabesin 3 (SB3) is an appealing GRPR antagonist with high receptor affinity. Gallium-68 labeled SB3 has good in vivo stability, after labeling with Indium-111; however, the molecule shows poor in vivo stability, which negatively impacts tumor-targeting capacity. A novel approach to increase in vivo stability of radiopeptides is by co-administration of the neutral endopeptidase (NEP) inhibitor, phosphoramidon (PA). We studied in vivo stability and biodistribution of [111In]SB3 without/with (-/+) PA in mice. Furthermore, SPECT/MRI on a novel, state-of-the-art platform was performed. PROCEDURES: GRPR affinity of SB3 was determined on PC295 xenograft sections using [125I]Tyr4-bombesin with tracer only or with increasing concentrations of SB3. For in vivo stability, mice were injected with 200/2000 pmol [111In]SB3 -/+ 300 µg PA. Blood was collected and analyzed. Biodistribution and SPECT/MRI studies were performed at 1, 4, and 24 h postinjection (p.i.) of 2.5 MBq/200 pmol or 25 MBq/200 pmol [111In]SB3 -/+ 300 µg PA in PC-3-xenografted mice. RESULTS: SB3 showed high affinity for GRPR (IC50 3.5 nM). Co-administration of PA resulted in twice higher intact peptide in vivo vs [111In]SB3 alone. Biodistribution studies at 1, 4, and 24 h p.i. show higher tumor uptake values with PA co-administration (19.7 ± 3.5 vs 10.2 ± 1.5, 17.6 ± 5.1 vs 8.3 ± 1.1, 6.5 ± 3.3 vs 3.1 ± 1.9 % ID/g tissue (P < 0.0001)). Tumor imaging with SPECT/MRI clearly improved after co-injection of PA. CONCLUSIONS: Co-administration of PA increased in vivo tumor targeting capacity of [111In]SB3, making this an attractive combination for GRPR-targeted tumor imaging.


Asunto(s)
Diagnóstico por Imagen/métodos , Cuidados Intraoperatorios/métodos , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/cirugía , Receptores de Bombesina/antagonistas & inhibidores , Animales , Unión Competitiva , Endocitosis , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos BALB C , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único
3.
J Nucl Med ; 58(10): 1609-1614, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28450563

RESUMEN

Recent studies have shown enhanced tumor targeting by novel somatostatin receptor (SSTR) antagonists compared with clinically widely used agonists. However, these results have been obtained mostly in neuroendocrine tumors, and only limited data are available for cancer types with lower SSTR expression, including breast cancer (BC). To date, two studies have reported higher binding of the antagonist than the agonist in BC, but in both studies only a limited number of cases were evaluated. In this preclinical study, we further investigated whether the application of an SSTR antagonist can improve SSTR-mediated BC imaging in a large panel of BC specimens. We also generated an in vivo BC mouse model and performed SPECT/MRI and biodistribution studies. Methods: Binding of 111In-DOTA-Tyr3-octreotate (SSTR agonist) and 111In-DOTA-JR11 (SSTR antagonist) to 40 human BC specimens was compared using in vitro autoradiography. SSTR2 immunostaining was performed to confirm SSTR2 expression of the tumor cells. Furthermore, binding of the radiolabeled SSTR agonist and antagonist was analyzed in tissue material from 6 patient-derived xenografts. One patient-derived xenograft, the estrogen receptor-positive model T126, was chosen to generate in vivo mouse models containing orthotopic breast tumors for in vivo SPECT/MRI and biodistribution studies after injection with 177Lu-DOTA-Tyr3-octreotate or 177Lu-DOTA-JR11. Results:111In-DOTA-JR11 binding to human BC tissue was significantly higher than 111In-DOTA-Tyr3-octreotate binding (P < 0.001). The median ratio of antagonist binding versus agonist binding was 3.39 (interquartile range, 2-5). SSTR2 immunostaining confirmed SSTR2 expression on the tumor cells. SPECT/MRI of the mouse model found better tumor visualization with the antagonist. This result was in line with the significantly higher tumor uptake of the radiolabeled antagonist than of the agonist as measured in biodistribution studies 285 min after radiotracer injection (percentage injected dose per gram of tissue: 1.92 ± 0.43 vs. 0.90 ± 0.17; P = 0.002). Conclusion: SSTR antagonists are promising candidates for BC imaging.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Péptidos Cíclicos/farmacología , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Autorradiografía , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Complejos de Coordinación/química , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Marcaje Isotópico , Ratones , Ratones Endogámicos BALB C , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacocinética , Distribución Tisular
4.
J Nucl Med ; 58(2): 293-299, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27609789

RESUMEN

Because overexpression of the gastrin-releasing peptide receptor (GRPR) has been reported on various cancer types, for example, prostate cancer and breast cancer, targeting this receptor with radioligands might have a significant impact on staging and treatment of GRPR-expressing tumors. NeoBOMB1 is a novel DOTA-coupled GRPR antagonist with high affinity for GRPR and excellent in vivo stability. The purpose of this preclinical study was to further explore the use of NeoBOMB1 for theranostic application by determining the biodistribution of 68Ga-NeoBOMB1 and 177Lu-NeoBOMB1. METHODS: PC-3 tumor-xenografted BALB/c nu/nu mice were injected with either approximately 13 MBq/250 pmol 68Ga-NeoBOMB1 or a low (∼1 MBq/200 pmol) versus high (∼1 MBq/10 pmol) peptide amount of 177Lu-NeoBOMB1, after which biodistribution and imaging studies were performed. At 6 time points (15, 30, 60, 120, 240, and 360 min for 68Ga-NeoBOMB1 and 1, 4, 24, 48, 96, and 168 h for 177Lu-NeoBOMB1) postinjection tumor and organ uptake was determined. To assess receptor specificity, additional groups of animals were coinjected with an excess of unlabeled NeoBOMB1. Results of the biodistribution studies were used to determine pharmacokinetics and dosimetry. Furthermore, PET/CT and SPECT/MRI were performed. RESULTS: Injection of approximately 250 pmol 68Ga-NeoBOMB1 resulted in a tumor and pancreas uptake of 12.4 ± 2.3 and 22.7 ± 3.3 percentage injected dose per gram (%ID/g) of tissue, respectively, at 120 min after injection. 177Lu-NeoBOMB1 biodistribution studies revealed a higher tumor uptake (17.9 ± 3.3 vs. 11.6 ± 1.3 %ID/g of tissue at 240 min after injection) and a lower pancreatic uptake (19.8 ± 6.9 vs. 105 ± 13 %ID/g of tissue at 240 min after injection) with the higher peptide amount injected, leading to a significant increase in the absorbed dose to the tumor versus the pancreas (200 pmol, 570 vs. 265 mGy/MBq; 10 pmol, 435 vs. 1393 mGy/MBq). Using these data to predict patient dosimetry, we found a kidney, pancreas, and liver exposure of 0.10, 0.65, and 0.06 mGy/MBq, respectively. Imaging studies resulted in good visualization of the tumor with both 68Ga-NeoBOMB1 and 177Lu-NeoBOMB1. CONCLUSION: Our findings indicate that 68Ga- or 177Lu-labeled NeoBOMB1 is a promising radiotracer with excellent tumor uptake and favorable pharmacokinetics for imaging and therapy of GRPR-expressing tumors.


Asunto(s)
Bombesina/uso terapéutico , Radioisótopos de Galio/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/radioterapia , Receptores de Bombesina/antagonistas & inhibidores , Nanomedicina Teranóstica/métodos , Animales , Línea Celular Tumoral , Radioisótopos de Galio/química , Humanos , Marcaje Isotópico/métodos , Masculino , Ratones , Ratones Desnudos , Terapia Molecular Dirigida/métodos , Neoplasias Experimentales/metabolismo , Radiofármacos/síntesis química , Radiofármacos/uso terapéutico , Resultado del Tratamiento
5.
J Nucl Med ; 57(2): 260-5, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26514177

RESUMEN

UNLABELLED: Peptide receptor scintigraphy and peptide receptor radionuclide therapy using radiolabeled somatostatin receptor (SSTR) agonists are successfully used in the clinic for imaging and treatment of neuroendocrine tumors. Contrary to the paradigm that internalization and the resulting accumulation of radiotracers in cells is necessary for efficient tumor targeting, recent studies have demonstrated the superiority of radiolabeled SSTR antagonists for imaging purposes, despite little to no internalization in cells. However, studies comparing the therapeutic antitumor effects of radiolabeled SSTR agonists versus antagonists are lacking. The aim of this study was to directly compare the therapeutic effect of (177)Lu-DOTA-octreotate, an SSTR agonist, and (177)Lu-DOTA-JR11, an SSTR antagonist. METHODS: We analyzed radiotracer uptake (both membrane-bound and internalized fractions) and the produced DNA double-strand breaks, by determining the number of p53 binding protein 1 foci, after incubating SSTR2-positive cells with (177)Lu-diethylene triamine pentaacetic acid, (177)Lu-DOTA-octreotate, or (177)Lu-DOTA-JR11. Also, biodistribution studies were performed in tumor-xenografted mice to determine the optimal dose for therapy experiments. Afterward, in vivo therapy experiments comparing the effect of (177)Lu-DOTA-octreotate and (177)Lu-DOTA-JR11 were performed in this same animal model. RESULTS: We found a 5-times-higher uptake of (177)Lu-DOTA-JR11 than of (177)Lu-DOTA-octreotate. The major part (88% ± 1%) of the antagonist uptake was membrane-bound, whereas 74% ± 3% of the total receptor agonist uptake was internalized. Cells treated with (177)Lu-DOTA-JR11 showed 2 times more p53-binding protein 1 foci than cells treated with (177)Lu-DOTA-octreotate. Biodistribution studies with (177)Lu-DOTA-JR11 (0.5 µg/30 MBq) resulted in the highest tumor radiation dose of 1.8 ± 0.7 Gy/MBq, 4.4 times higher than the highest tumor radiation dose found for (177)Lu-DOTA-octreotate. In vivo therapy studies with (177)Lu-DOTA-octreotate and (177)Lu-DOTA-JR11 resulted in a tumor growth delay time of 18 ± 5 and 26 ± 7 d, respectively. Median survival rates were 43.5, 61, and 71 d for the control group, (177)Lu-DOTA-octreotate group, and the (177)Lu-DOTA-JR11-treated group, respectively. CONCLUSION: On the basis of these results, we concluded that the use of radiolabeled SSTR antagonists such as JR11 might enhance peptide receptor scintigraphy and peptide receptor radionuclide therapy of neuroendocrine tumors and provide successful imaging and therapeutic strategies for cancer types with relatively low SSTR expression.


Asunto(s)
Complejos de Coordinación/uso terapéutico , Tumores Neuroendocrinos/radioterapia , Octreótido/análogos & derivados , Péptidos Cíclicos/uso terapéutico , Radiofármacos/uso terapéutico , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Complejos de Coordinación/farmacocinética , Daño del ADN , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Octreótido/farmacocinética , Octreótido/uso terapéutico , Péptidos Cíclicos/farmacocinética , Radiometría , Radiofármacos/farmacocinética , Análisis de Supervivencia , Distribución Tisular , Proteína 1 de Unión al Supresor Tumoral P53 , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Contrast Media Mol Imaging ; 11(2): 106-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463264

RESUMEN

Cationic liposomes carrying high [Gd] can be used as efficient cell-labeling agents. In a compartmentalized state, Gd can cause signal loss (relaxivity quenching). The contributions of liposomal [Gd], size and compartmentalization state to relaxivity quenching were assessed. The dependency of signal intensity (SI) on intraliposomal [Gd] was assessed comparing three different [Gd] (0.3, 0.6 and 1.0 M Gd) in both small (80 nm) and large (120 nm) cationic liposomes. In addition, five compartmentalization states were compared: free Gd, intact Gd liposomes, ruptured Gd liposomes, Gd liposomes in intact cells and Gd liposomes in ruptured cells (simulating cell death). Gd also causes R2 effects, which is often overlooked. Therefore, both R1 and R2 relaxation rates of a dilution range were measured by T1 and T2 mapping on a 7 T clinical scanner. Less is more. As the unidirectional water efflux rate (outbound across the liposome membrane, κle) is proportional to the surface:volume ratio, smaller liposomes yielded a consistently higher R1 than larger liposomes. For equal voxel [Gd] less concentrated liposomes (0.3 M Gd) yielded higher R1/R2 ratio because of the higher extraliposomal water fraction (vl ). Gd exhibits a dualistic behavior: from hypointensity to hyperintensity to hypointensity, with decreasing [Gd]. Regarding compartmentalization, fewer membrane barriers means a higher R1 /R2 ratio. Gd liposomes exhibit a versatile contrast behavior, dependent on the compartmentalization state, liposomal size, intraliposomal [Gd] and liposome number. Both R1 and R2 effects contribute to this. The versatility allows one to tailor the optimal liposomal formulation to desired goals in cell labeling and tracking.


Asunto(s)
Medios de Contraste/administración & dosificación , Gadolinio/administración & dosificación , Liposomas/administración & dosificación , Imagen por Resonancia Magnética , Línea Celular , Rastreo Celular/métodos , Medios de Contraste/química , Gadolinio/química , Humanos , Liposomas/química
7.
PLoS One ; 10(9): e0138572, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26394043

RESUMEN

Impaired homing and delayed recovery upon hematopoietic stem cell transplantation (HSCT) with hematopoietic stem cells (HSC) derived from umbilical cord blood (UCB) is a major problem. Tracking transplanted cells in vivo will be helpful to detect impaired homing at an early stage and allows early interventions to improve engraftment and outcome after transplantation. In this study, we show sufficient intracellular labeling of UCB-derived CD34+ cells, with 19F-containing PLGA nanoparticles which were detectable with both flow cytometry and magnetic resonance spectroscopy (MRS). In addition, labeled CD34+ cells maintain their capacity to proliferate and differentiate, which is pivotal for successful engraftment after transplantation in vivo. These results set the stage for in vivo tracking experiments, through which the homing efficiency of transplanted cells can be studied.


Asunto(s)
Antígenos CD34/metabolismo , Rastreo Celular/métodos , Sangre Fetal/citología , Células Madre Hematopoyéticas/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Radioisótopos de Flúor , Células Madre Hematopoyéticas/química , Humanos , Ácido Láctico/química , Microscopía Confocal , Nanopartículas/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Reproducibilidad de los Resultados , Factores de Tiempo
8.
J Magn Reson Imaging ; 39(4): 901-10, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24123470

RESUMEN

PURPOSE: To evaluate the reproducibility and sensitivity of the modified CINE inversion recovery (mCINE-IR) acquisition on rats for measuring the myocardial T1 at 7 Tesla. MATERIALS AND METHODS: The recently published mCINE-IR acquisition on humans was applied on rats for the first time, enabling the possibility of translational studies with an identical sequence. Simulations were used to study signal evolution and heart rate dependency. Gadolinium phantoms, a heart specimen and a healthy rat were used to study reproducibility. Two cryo-infarcted rats were scanned to measure late gadolinium enhancement (LGE). RESULTS: In the phantom reproducibility studies the T1 measurements had a maximum coefficient of variation (COV) of 1.3%. For the in vivo reproducibility the COV was below 5% in the anterior cardiac segments. In simulations with phantoms and specimens, a heart rate dependency of approximately 0.5 ms/bpm was present. The T1 maps of the cryo-infarcted rats showed a clear lowering of T1 in de LGE region. CONCLUSION: The results show that mCINE-IR is highly reproducible and that the sensitivity allows detecting T1 changes in the rat myocardium.


Asunto(s)
Algoritmos , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Imagen por Resonancia Cinemagnética/métodos , Infarto del Miocardio/patología , Miocardio/patología , Animales , Ratas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA