Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93.617
Filtrar
1.
Biomaterials ; 312: 122733, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106819

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrates unique characteristics in anticancer therapies as it selectively induces apoptosis in cancer cells. However, most cancer cells are TRAIL-resistant. Odanacatib (ODN), a cathepsin K inhibitor, is considered a novel sensitizer for cancer treatment. Combination therapy between TRAIL and sensitizers is considered a potent platform that improves TRAIL-based anticancer therapies beyond TRAIL monotherapy. Herein, we developed ODN loaded poly(lactic-co-glycolic) nanoparticles conjugated to GST-TRAIL (TRAIL-ODN-PLGA-NPs) to target and treat TRAIL-resistant cancer. TRAIL-ODN-PLGA-NPs demonstrated a significant increase in cellular uptake via death receptors (DR5 and DR4) on surface of cancer cells. TRAIL-ODN-PLGA-NPs exposure destroyed more TRAIL-resistant cells compared to a single treatment with free drugs. The released ODN decreased the Raptor protein, thereby increasing damage to mitochondria by elevating reactive oxygen species (ROS) generation. Additionally, Bim protein stabilization improved TRAIL-resistant cell sensitization to TRAIL-induced apoptosis. The in vivo biodistribution study revealed that TRAIL-ODN-PLGA-NPs demonstrated high location and retention in tumor sites via the intravenous route. Furthermore, TRAIL-ODN-PLGA-NPs significantly inhibited xenograft tumor models of TRAIL-resistant Caki-1 and TRAIL-sensitive MDA-MB-231 cells.The inhibition was associated with apoptosis activation, Raptor protein stabilizing Bim protein downregulation, Bax accumulation, and mitochondrial ROS generation elevation. Additionally, TRAIL-ODN-PLGA-NPs affected the tumor microenvironment by increasing tumor necrosis factor-α and reducing interleukin-6. In conclusion, we evealed that our formulation demonstrated synergistic effects against TRAIL compared with the combination of free drug in vitro and in vivo models. Therefore, TRAIL-ODN-PLGA-NPs may be a novel candidate for TRAIL-induced apoptosis in cancer treatment.


Asunto(s)
Antineoplásicos , Compuestos de Bifenilo , Resistencia a Antineoplásicos , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ligando Inductor de Apoptosis Relacionado con TNF , Animales , Femenino , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/uso terapéutico , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/química , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
2.
Biomaterials ; 313: 122748, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39180918

RESUMEN

Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/química , Animales , Humanos , Distribución Tisular , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Oxazoles/química , Ratones , Propiedades de Superficie , Línea Celular Tumoral , Ratones Endogámicos C57BL , Femenino
3.
Biomaterials ; 312: 122740, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39096839

RESUMEN

Metastasis stands as the primary contributor to mortality associated with tumors. Chemotherapy and immunotherapy are frequently utilized in the management of metastatic solid tumors. Nevertheless, these therapeutic modalities are linked to serious adverse effects and limited effectiveness in preventing metastasis. Here, we report a novel therapeutic strategy named starvation-immunotherapy, wherein an immune checkpoint inhibitor is combined with an ultra-long-acting L-asparaginase that is a fusion protein comprising L-asparaginase (ASNase) and an elastin-like polypeptide (ELP), termed ASNase-ELP. ASNase-ELP's thermosensitivity enables it to generate an in-situ depot following an intratumoral injection, yielding increased dose tolerance, improved pharmacokinetics, sustained release, optimized biodistribution, and augmented tumor retention compared to free ASNase. As a result, in murine models of oral cancer, melanoma, and cervical cancer, the antitumor efficacy of ASNase-ELP by selectively and sustainably depleting L-asparagine essential for tumor cell survival was substantially superior to that of ASNase or Cisplatin, a first-line anti-solid tumor medicine, without any observable adverse effects. Furthermore, the combination of ASNase-ELP and an immune checkpoint inhibitor was more effective than either therapy alone in impeding melanoma metastasis. Overall, the synergistic strategy of starvation-immunotherapy holds excellent promise in reshaping the therapeutic landscape of refractory metastatic tumors and offering a new alternative for next-generation oncology treatments.


Asunto(s)
Asparaginasa , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Animales , Asparaginasa/uso terapéutico , Asparaginasa/farmacología , Asparaginasa/química , Inmunoterapia/métodos , Femenino , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ratones , Humanos , Línea Celular Tumoral , Sinergismo Farmacológico , Elastina/química , Elastina/metabolismo , Metástasis de la Neoplasia , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Distribución Tisular
4.
Drug Dev Res ; 85(6): e22258, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39253992

RESUMEN

The 2-nitroimidazole based 99mTc-radiopharmaceuticals are widely explored for imaging tumor hypoxia. Radiopharmaceuticals for targeting hypoxia are often lipophilic and therefore, show significant uptake in liver and other vital organs. In this context, lipophilic radiopharmaceuticals with design features enabling faster clearance from liver may be more desirable. A dipicolylamine-NCS bifunctional chelator that could generate a thiourea-bridge up on conjugation to primary amine bearing molecule was used to synthesize a 2-nitroimidazole-dipicolyl amine ligand for radiolabeling with 99mTc(CO)3 core. Corresponding Re(CO)3-analogue was prepared to establish the structure of 2-nitroimidazole-99mTc(CO)3 complex prepared in trace level. The 2-nitroimidazole-99mTc(CO)3 complex showed a hypoxic to normoxic ratio of ~2.5 in CHO cells at 3 h. In vivo, the complex showed accumulation and retention in tumor with high tumor to blood and tumor to muscle ratio. The study demonstrated the utility of metabolizable thiourea-bridge in 2-nitroimidazole-99mTc(CO)3 complex in inducing faster clearance of the radiotracer from liver. The dipicolylamine-NCS bifunctional chelator reported herein can also be used for radiolabeling other class of target specific molecules with 99mTc(CO)3 core.


Asunto(s)
Radiofármacos , Tiourea , Hipoxia Tumoral , Animales , Radiofármacos/farmacocinética , Células CHO , Tiourea/análogos & derivados , Tiourea/farmacocinética , Tiourea/química , Cricetulus , Ratones , Nitroimidazoles/farmacocinética , Nitroimidazoles/química , Compuestos de Organotecnecio/farmacocinética , Compuestos de Organotecnecio/química , Compuestos de Organotecnecio/administración & dosificación , Distribución Tisular , Ácidos Picolínicos/farmacocinética , Ácidos Picolínicos/química , Humanos , Tecnecio
5.
Theranostics ; 14(12): 4701-4712, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239524

RESUMEN

Erythropoietin-producing hepatocellular receptor A2 (EphA2), is a receptor tyrosine kinase involved in cell-cell interactions. It is known to be overexpressed in various tumors and is associated with poor prognosis. EphA2 has been proposed as a target for theranostic applications. Low molecular weight peptide-based scaffolds with low nanomolar affinities have been shown to be ideal in such applications. Bicyclic peptides have emerged as an alternative to traditional peptides for this purpose, offering affinities comparable to antibodies due to their constrained nature, along with high tissue penetration, and improved stability compared to linear counterparts. This study presents the development and comprehensive in vitro and in vivo preclinical evaluation of BCY18469, a novel EphA2-targeting bicyclic peptide-based radiotheranostic agent. Methods: The EphA2-targeting Bicycle® peptide BCY18469 was identified through phage-display and chemically optimized. BCY18469 was radiolabeled with 68Ga, 177Lu and 111In. The physicochemical properties, binding affinity and internalization as well as specificity of the peptide were evaluated in vitro. In vivo PET/MR and SPECT/CT imaging studies were performed using [68Ga]Ga-BCY18469 and [111In]In-BCY18469, respectively, along with biodistribution of [177Lu]Lu-BCY18469 up to 24 h post injection in HT1080- and PC-3-tumor bearing BALB/c nu/nu EphA2-overexpressing xenograft mouse models. Results: The EphA2-targeting bicyclic peptide BCY18469 showed high binding affinity toward human and mouse EphA2 (1.9 and 3.8 nM, respectively). BCY18469 specifically bound and internalized into EphA2-expressing HT1080 cells. Imaging studies showed high tumor enrichment at early time-points (SUV of 1.7 g/mL at 1 h p.i. and 1.2 g/mL at 2 h p.i. in PET/MRI, HT1080 xenograft) with tumor contrast as early as 5 min p.i. and kidney-mediated clearance. Biodistribution studies revealed high early tumor uptake (19.5 ± 3.5 %ID/g at 1 h p.i., HT1080 xenograft) with SPECT/CT imaging further confirming these findings (5.7 ± 1.5 %ID/g at 1 h p.i., PC-3 xenograft). Conclusion: BCY18469 demonstrated high affinity, specific targeting of EphA2, a favorable biodistribution profile, and clearance through renal pathways. These findings underscore the potentially important role of bicyclic peptides in advancing radiotheranostic approaches and encourage additional translational research.


Asunto(s)
Receptor EphA2 , Animales , Receptor EphA2/metabolismo , Humanos , Ratones , Línea Celular Tumoral , Distribución Tisular , Péptidos Cíclicos/farmacocinética , Péptidos Cíclicos/química , Radiofármacos/farmacocinética , Radiofármacos/química , Masculino , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Lutecio/química , Radioisótopos de Indio , Radioisótopos/química , Femenino , Radioisótopos de Galio , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/metabolismo
6.
Theranostics ; 14(13): 5022-5101, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39267777

RESUMEN

The potential of intranasal administered imaging agents to altogether bypass the blood-brain barrier offers a promising non-invasive approach for delivery directly to the brain. This review provides a comprehensive analysis of the advancements and challenges of delivering neuroimaging agents to the brain by way of the intranasal route, focusing on the various imaging modalities and their applications in central nervous system diagnostics and therapeutics. The various imaging modalities provide distinct insights into the pharmacokinetics, biodistribution, and specific interactions of imaging agents within the brain, facilitated by the use of tailored tracers and contrast agents. Methods: A comprehensive literature search spanned PubMed, Scopus, Embase, and Web of Science, covering publications from 1989 to 2024 inclusive. Starting with advancements in tracer development, we going to explore the rationale for integration of imaging techniques, and the critical role novel formulations such as nanoparticles, nano- and micro-emulsions in enhancing imaging agent delivery and visualisation. Results: The review highlights the use of innovative formulations in improving intranasal administration of neuroimaging agents, showcasing their ability to navigate the complex anatomical and physiological barriers of the nose-to-brain pathway. Various imaging techniques, MRI, PET, SPECT, CT, FUS and OI, were evaluated for their effectiveness in tracking these agents. The findings indicate significant improvements in brain targeting efficiency, rapid uptake, and sustained brain presence using innovative formulations. Conclusion: Future directions involve the development of optimised tracers tailored for intranasal administration, the potential of multimodal imaging approaches, and the implications of these advancements for diagnosing and treating neurological disorders.


Asunto(s)
Administración Intranasal , Encéfalo , Humanos , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Animales , Medios de Contraste/administración & dosificación , Medios de Contraste/farmacocinética , Neuroimagen/métodos , Sistemas de Liberación de Medicamentos/métodos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/diagnóstico por imagen , Nanopartículas/química , Nanopartículas/administración & dosificación , Distribución Tisular , Imagen por Resonancia Magnética/métodos
7.
J Nanobiotechnology ; 22(1): 538, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39227962

RESUMEN

The risk of ulcerative colitis (UC) is increasing worldwide with limited success using classical drugs, which has underscored the development of novel agents. Recently, carrier-free molecular assembly has been proven to be an effective drug delivery system, but it has yet to be examined for UC drug development using phytochemicals. Based on traditional Chinese medicine compatibility and potential medicinal uses, a pair of natural compounds, berberine (BBR) and magnolol (MAG), were found to self-assemble into nanostructures in aqueous solutions. Spectral analysis revealed that the assembly mechanisms of BBR and MAG were mediated through charge interactions and π-π stacking. Pharmacokinetic studies and animal imaging showed that BBR-MAG self-assembly (BM) effectively promoted the oral bioavailability and biodistribution of BBR in the colon. BM exhibited superior effects in regulating inflammatory factors, maintaining colon barrier integrity, and regulating gut microbiota in a dextran sulfate sodium salt-induced colitis mouse model. Additionally, no apparent signs of toxicity were observed, suggesting that BM has a favorable safety profile. This study presents a new strategy for UC management and highlights the cooperative effects of combined phytochemicals.


Asunto(s)
Berberina , Compuestos de Bifenilo , Colitis Ulcerosa , Lignanos , Nanoestructuras , Animales , Colitis Ulcerosa/tratamiento farmacológico , Berberina/química , Berberina/farmacología , Berberina/uso terapéutico , Lignanos/química , Lignanos/farmacología , Lignanos/uso terapéutico , Ratones , Compuestos de Bifenilo/química , Nanoestructuras/química , Masculino , Sulfato de Dextran/química , Colon/efectos de los fármacos , Colon/patología , Modelos Animales de Enfermedad , Distribución Tisular , Ratones Endogámicos C57BL , Microbioma Gastrointestinal/efectos de los fármacos , Disponibilidad Biológica
8.
Molecules ; 29(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39274840

RESUMEN

Acidic amino acid peptides have a high affinity for bone. Previously, we demonstrated that radiogallium complex-conjugated oligo-acidic amino acids possess promising properties as bone-seeking radiopharmaceuticals. Here, to elucidate the effect of stereoisomers of Glu in Glu-containing peptides [(Glu)14] on their accumulation in the kidney, the biodistributions of [67Ga]Ga-N,N'-bis-[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid-conjugated (l-Glu)14 ([67Ga]Ga-HBED-CC-(l-Glu)14), [67Ga]Ga-HBED-CC-(d-Glu)14, [67Ga]Ga-HBED-CC-(dl-Glu)14, and [67Ga]Ga-HBED-CC-(d-Glu-l-Glu)7 were compared. Although the accumulation of these compounds in the bone was comparable, their kidney accumulation and retention were strikingly different, with [67Ga]Ga-HBED-CC-(d-Glu-l-Glu)7 exhibiting the lowest level of kidney accumulation among these compounds. Repeated d- and l-peptides may be a useful method for reducing renal accumulation in some cases.


Asunto(s)
Radioisótopos de Galio , Ácido Glutámico , Riñón , Péptidos , Radiofármacos , Péptidos/química , Ácido Glutámico/química , Riñón/metabolismo , Animales , Radiofármacos/química , Radiofármacos/farmacocinética , Radioisótopos de Galio/química , Distribución Tisular , Ratones , Huesos/metabolismo
9.
Molecules ; 29(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39274933

RESUMEN

64Cu is gaining recognition not only for its diagnostic capabilities in nuclear medical imaging but also for its therapeutic and theranostic potential. The simultaneous ß- and Auger emissions of 64Cu can be utilized to induce a therapeutic effect on cancerous lesions. The finding of the exceptional biodistribution characteristics of the radionuclide 64Cu, when administered as basic copper ions, has highlighted its potential therapeutic application in cancer treatment. Preclinical and clinical research on the effectiveness of [64Cu]CuCl2 as a theranostic radiopharmaceutical has commenced only in the past decade. Current clinical studies are increasingly demonstrating the high specificity and uptake of [64Cu]Cu2+ by malignant tissues during early cancer progression, indicating its potential for early cancer diagnosis across various organs. This short review aims to present the latest preclinical studies involving [64Cu]CuCl2, offering valuable insights for researchers planning new in vitro and in vivo studies to explore the theranostic potential of [64Cu]Cu2+.


Asunto(s)
Radioisótopos de Cobre , Cobre , Neoplasias , Radiofármacos , Nanomedicina Teranóstica , Humanos , Radioisótopos de Cobre/química , Cobre/química , Animales , Radiofármacos/química , Radiofármacos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/diagnóstico por imagen , Neoplasias/diagnóstico , Nanomedicina Teranóstica/métodos , Distribución Tisular
10.
Adv Drug Deliv Rev ; 213: 115445, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39222795

RESUMEN

Deformability is one of the critical attributes of nanoparticle (NP) drug carriers, along with size, shape, and surface properties. It affects various aspects of NP biotransport, ranging from circulation and biodistribution to interactions with biological barriers and target cells. Recent studies report additional roles of NP deformability in biotransport processes, including protein corona formation, intracellular trafficking, and organelle distribution. This review focuses on the literature published in the past five years to update our understanding of NP deformability and its effect on NP biotransport. We introduce different methods of modulating and evaluating NP deformability and showcase recent studies that compare a series of NPs in their performance in biotransport events at all levels, highlighting the consensus and disagreement of the findings. It concludes with a perspective on the intricacy of systematic investigation of NP deformability and future opportunities to advance its control toward optimal drug delivery.


Asunto(s)
Nanopartículas , Nanopartículas/química , Humanos , Animales , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Distribución Tisular , Transporte Biológico , Propiedades de Superficie
11.
Arch Environ Contam Toxicol ; 87(2): 114-126, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39097543

RESUMEN

Wolverines are facultative scavengers that feed near the top of terrestrial food chains. We characterized concentrations of mercury and other trace elements in tissues of wolverine from a broad geographic area, representing much of their contemporary distribution in northwestern North America. We obtained tissues from 504 wolverines, from which mercury was measured on muscle (n = 448), kidney (n = 222), liver (n = 148), hair (n = 130), and brain (n = 52). In addition, methylmercury, seven trace elements (arsenic, cadmium, chromium, cobalt, lead, nickel, selenium), and arsenic compounds were measured on a subset of samples. Concentrations of mercury and other trace elements varied between tissues and were generally highest in kidney compared to brain, liver and muscle. Mercury was predominately as methylmercury in brain and muscle, but largely as inorganic mercury in liver and kidney. Mercury concentrations of hair were moderately correlated with those of internal tissues (Pearson r = 0.51-0.75, p ≤ 0.004), making hair a good non-lethal indicator of broad spatial or temporal differences in mercury exposure to wolverine. Arsenobetaine was the dominant arsenic compound identified in tissues, and arsenite, arsenocholine and dimethylarsinic acid were also detected. A preliminary risk assessment suggested the cadmium, lead, mercury, and selenium concentrations in our sample of wolverines were not likely to pose a risk of overt toxicological effects. This study generated a comprehensive dataset on mercury and other trace elements in wolverine, which will support future contaminants study of this northern terrestrial carnivore.


Asunto(s)
Cabello , Mercurio , Animales , Mercurio/análisis , Medición de Riesgo , Distribución Tisular , Cabello/química , Monitoreo del Ambiente , Riñón , Contaminantes Ambientales/análisis , Oligoelementos/análisis , Hígado/química , Mustelidae , Compuestos de Metilmercurio/análisis
12.
Dalton Trans ; 53(35): 14634-14647, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39163366

RESUMEN

Indium-111 (111In) is a diagnostic radiometal that is important in nuclear medicine for single-photon emission computed tomography (SPECT). In order to apply this radiometal, it needs to be stably chelated and conjugated to a targeting vector that delivers it to diseased tissue. Identifying effective chelators that are capable of binding and retaining [111In]In3+in vivo is an important research area. In this study, two 18-membered macrocyclic chelators, py-macrodipa and py2-macrodipa, were investigated for their ability to form stable coordination complexes with In3+ and to be effectively radiolabeled with [111In]In3+. The In3+ complexes of these two chelators were characterized by NMR spectroscopy, X-ray crystallography, and density functional theory calculations. These studies show that both py-macrodipa and py2-macrodipa form 8-coordinate In3+ complexes and attain an asymmetric conformation, consistent with prior studies on this ligand class with small rare earth metal ions. Spectrophotometric titrations were carried out to determine the thermodynamic stability constants (log KML) of [In(py-macrodipa)]+ and [In(py2-macrodipa)]+, which were found to be 18.96(6) and 19.53(5), respectively, where the values in parentheses are the errors of the last significant figures obtained from the standard deviation from three independent replicates. Radiolabeling studies showed that py-macrodipa and py2-macrodipa can quantitatively be radiolabeled with [111In]In3+ at 25 °C within 5 min, even at ligand concentrations as low as 1 µM. The in vitro stability of the radiolabeled complexes was investigated in human serum at 37 °C, revealing that ∼90% of [111In][In(py-macrodipa)]+ and [111In][In(py2-macrodipa)]+ remained intact after 7 days. The biodistribution of these radiolabeled complexes in mice was investigated, showing lower uptake in the kidneys, liver, and blood at the 24 h mark compared to [111In]InCl3. These results demonstrate the potential of py-macrodipa and py2-macrodipa as chelators for [111In]In3+, suggesting their value for SPECT radiopharmaceuticals.


Asunto(s)
Quelantes , Radioisótopos de Indio , Compuestos Macrocíclicos , Quelantes/química , Compuestos Macrocíclicos/química , Animales , Radioisótopos de Indio/química , Ratones , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Teoría Funcional de la Densidad , Distribución Tisular , Modelos Moleculares , Radiofármacos/química , Indio/química , Cristalografía por Rayos X , Estructura Molecular
13.
Sci Rep ; 14(1): 20241, 2024 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-39215130

RESUMEN

Nuclear medicine is an important tool for use in molecular imaging of important biological processes. Methods for intravenous delivery of radiotracers remains a challenge, with tail vein injections demonstrated to be technically difficult and lacking in reproducibility. Other intravenous methods include jugular vein (JV) injection, which requires a more invasive and precise microsurgical technique. Although the retroorbital (RO) sinus drains directly into the JV, and RO injections are minimally invasive and simpler to perform, they remain underutilized, perhaps due to a lack of studies demonstrating their performance. This study provides a comprehensive comparison of dynamic tissue biodistribution of three categories of commonly utilized radiopharmaceuticals between JV and RO injection methods in prostate tumor-bearing mice using PET-CT imaging. Results show that JV and RO injections have equivalent dynamic tissue biodistributions across the three categories of radiopharmaceuticals used: (1) small molecule measuring tumor metabolism (18F-flurodeoxyglucose [FDG]); (2) peptide-based probe measuring angiogenesis (64Cu-NOTA-PEG4-cRGD2); and (3) dextran-based nanocarrier (64Cu-NOTA-D20). Although RO injections present with some limitations such as type of injectate and difficulty for measuring acute, dynamic pharmacokinetics, this study demonstrates that RO injections are a viable, minimally invasive or stressful, and efficient alternative intravenous delivery technique for molecular imaging.


Asunto(s)
Venas Yugulares , Neoplasias de la Próstata , Radiofármacos , Animales , Ratones , Masculino , Venas Yugulares/diagnóstico por imagen , Venas Yugulares/metabolismo , Distribución Tisular , Radiofármacos/farmacocinética , Radiofármacos/administración & dosificación , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Inyecciones Intravenosas , Línea Celular Tumoral
14.
Sci Rep ; 14(1): 19938, 2024 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-39198676

RESUMEN

Radiopharmaceutical therapy using α -emitting 225 Ac is an emerging treatment for patients with advanced metastatic cancers. Measurement of the spatial dose distribution in organs and tumors is needed to inform treatment dose prescription and reduce off-target toxicity, at not only organ but also sub-organ scales. Digital autoradiography with α -sensitive detection devices can measure radioactivity distributions at 20-40 µ m resolution, but anatomical characterization is typically limited to 2D. We collected digital autoradiographs across whole tissues to generate 3D dose volumes and used them to evaluate the simultaneous tumor control and regional kidney dosimetry of a novel therapeutic radiopharmaceutical for prostate cancer, [225Ac]Ac-Macropa-PEG4-YS5, in mice. 22Rv1 xenograft-bearing mice treated with 18.5 kBq of [225Ac]Ac-Macropa-PEG4-YS5 were sacrificed at 24 h and 168 h post-injection for quantitative α -particle digital autoradiography and hematoxylin and eosin staining. Gamma-ray spectroscopy of biodistribution data was used to determine temporal dynamics and 213 Bi redistribution. Tumor control probability and sub-kidney dosimetry were assessed. Heterogeneous 225 Ac spatial distribution was observed in both tumors and kidneys. Tumor control was maintained despite heterogeneity if cold spots coincided with necrotic regions. 225 Ac dose-rate was highest in the cortex and renal vasculature. Extrapolation of tumor control suggested that kidney absorbed dose could be reduced by 41% while maintaining 90% TCP. The 3D dosimetry methods described allow for whole tumor and organ dose measurements following 225 Ac radiopharmaceutical therapy, which correlate to tumor control and toxicity outcomes.


Asunto(s)
Actinio , Autorradiografía , Neoplasias de la Próstata , Radiofármacos , Masculino , Animales , Neoplasias de la Próstata/radioterapia , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Radiofármacos/administración & dosificación , Radiofármacos/farmacocinética , Ratones , Humanos , Línea Celular Tumoral , Autorradiografía/métodos , Distribución Tisular , Radiometría/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Genes (Basel) ; 15(8)2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39202404

RESUMEN

As a founding member of the Src family of kinases, Src has been confirmed to participate in the regulation of immune responses, integrin signaling, and motility. Ducks are usually asymptomatic carriers of RNA viruses such as Newcastle disease virus and avian influenza virus, which can be deadly to chickens. The beneficial role of Src in modulating the immune response remains largely unknown in ducks. Here, we characterized the duck Src and found that it contains a 192-base-pair 5' untranslated region, a 1602-base-pair coding region, and a 2541-base-pair 3' untranslated region, encoding 533 amino acid residues. Additionally, duSrc transcripts were significantly activated in duck tissues infected by Newcastle disease virus compared to controls. The duSrc transcripts were notably widespread in all tissues examined, and the expression level was higher in liver, blood, lung, pancreas, and thymus. Moreover, we found the expression levels of IFN-ß, NF-κB, IRF3, and Src were significantly increased in DEFs after infection with 5'ppp dsRNA, but there was no significant difference before and after treatment in DF1 cells. Furthermore, overexpression of duSrc followed by stimulation with 5'ppp dsRNA led to an elevation of IFN-ß levels. The SH3 and PTKc domains of duSrc contributed to promoting the activity of IFN-ß and NF-κB in DEFs stimulated by 5'ppp dsRNA.


Asunto(s)
Clonación Molecular , Patos , Animales , Patos/genética , Patos/inmunología , Patos/virología , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/genética , Proteínas Aviares/genética , Proteínas Aviares/inmunología , Proteínas Aviares/metabolismo , Enfermedad de Newcastle/inmunología , Enfermedad de Newcastle/virología , Enfermedad de Newcastle/genética , Interferón beta/genética , Interferón beta/inmunología , Interferón beta/metabolismo , Distribución Tisular , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/genética
16.
Redox Biol ; 75: 103294, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39096854

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a serious interstitial lung disease. However, the definitive diagnosis of IPF is impeded by the limited capabilities of current diagnostic methods, which may fail to capture the optimal timing for treatment. The main goal of this study is to determine the feasibility of a nitroreductase (NTR) responsive probe, 18F-NCRP, for early detection and deterioration monitoring of IPF. 18F-NCRP was obtained with high radiochemical purity (>95 %). BLM-injured mice were established by intratracheal instillation with bleomycin (BLM) and characterized through histological analysis. Longitudinal PET/CT imaging, biodistribution study and in vitro autoradiography were performed. The correlations between the uptake of 18F-NCRP and mean lung density (tested by CT), as well as histopathological characteristics were analyzed. In PET imaging study, 18F-NCRP exhibited promising efficacy in monitoring the progression of IPF, which was earlier than CT. The ratio of uptake in BLM-injured lung to control lung increased from 1.4-fold on D15 to 2.2-fold on D22. Biodistribution data showed a significant lung uptake of 18F-NCRP in BLM-injured mice. There was a strong positive correlation between the 18F-NCRP uptake in the BLM-injured lungs and the histopathological characteristics. Given that, 18F-NCRP PET imaging of NTR, a promising biomarker for investigating the underlying pathogenic mechanism of IPF, is attainable as well as desirable, which might lay the foundation for establishing an NTR-targeted imaging evaluation system of IPF.


Asunto(s)
Diagnóstico Precoz , Fibrosis Pulmonar Idiopática , Nitrorreductasas , Tomografía Computarizada por Tomografía de Emisión de Positrones , Animales , Ratones , Nitrorreductasas/metabolismo , Fibrosis Pulmonar Idiopática/diagnóstico por imagen , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/inducido químicamente , Bleomicina , Pulmón/diagnóstico por imagen , Pulmón/patología , Pulmón/metabolismo , Humanos , Modelos Animales de Enfermedad , Distribución Tisular , Masculino , Radioisótopos de Flúor , Radiofármacos
17.
Sci Rep ; 14(1): 17994, 2024 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-39097625

RESUMEN

CD73 is a cell-surface ectoenzyme that hydrolyzes the conversion of extracellular adenosine monophosphate to adenosine, which in turn can promote resistance to immune checkpoint blockade therapy. Immune response may therefore be improved by targeting tumor CD73, and this possibility underlines the need to non-invasively assess tumor CD73 level. In this study, we developed a cysteine site-specific 89Zr-labeled anti-CD73 (89Zr-CD73) IgG immuno-PET technique that can image tumor CD73 expression in living bodies. Anti-CD73 IgG was reduced with tris(2-carboxyethyl)phosphine, underwent sulfohydryl moiety-specific conjugation with deferoxamine-maleimide, and was radiolabeled with 89Zr. CT26 mouse colon cancer cells, CT26/CD73 cells engineered to constitutively overexpress CD73, and 4T1.2 mouse breast cancer cells underwent cell binding assays and western blotting. Balb/c nude mice bearing tumors underwent 89Zr-CD73 IgG PET imaging and biodistribution studies. 89Zr-CD73 IgG showed 20-fold higher binding to overexpressing CT26/CD73 cells compared to low-expressing CT26 cells, and moderate expressing 4T1.2 cells showed uptake that was 38.9 ± 1.51% of CT26/CD73 cells. Uptake was dramatically suppressed by excess unlabeled antibody. CD73 content proportionately increased in CT26 and CT26/CD73 cell mixtures was associated with linear increases in 89Zr-CD73 IgG uptake. 89Zr-CD73 IgG PET/CT displayed clear accumulation in CT26/CD73 tumors with greater uptake compared to CT26 tumors (3.13 ± 1.70%ID/g vs. 1.27 ± 0.31%ID/g at 8 days; P = 0.04). Specificity was further supported by low CT26/CD73 tumor-to-blood ratio of 89Zr-isotype-IgG compared to 89Zr-CD73 IgG (0.48 ± 0.08 vs. 2.68 ± 0.52 at 4 days and 0.53 ± 0.07 vs. 4.81 ± 1.02 at 8 days; both P < 0.001). Immunoblotting and immunohistochemistry confirmed strong CD73 expression in CT26/CD73 tumors and low expression in CT26 tumors. 4T1.2 tumor mice also showed clear 89Zr-CD73 IgG accumulation at 8 days (3.75 ± 0.70%ID/g) with high tumor-to-blood ratio compared to 89Zr-isotype-IgG (4.91 ± 1.74 vs. 1.20 ± 0.28; P < 0.005). 89Zr-CD73 IgG specifically targeted CD73 on high expressing cancer cells in vitro and tumors in vivo. Thus, 89Zr-CD73 IgG immuno-PET may be useful for the non-invasive monitoring of CD73 expression in tumors of living subjects.


Asunto(s)
5'-Nucleotidasa , Neoplasias del Colon , Cisteína , Tomografía de Emisión de Positrones , Circonio , Animales , 5'-Nucleotidasa/metabolismo , Circonio/química , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Ratones , Línea Celular Tumoral , Tomografía de Emisión de Positrones/métodos , Cisteína/metabolismo , Humanos , Radioisótopos , Femenino , Ratones Endogámicos BALB C , Distribución Tisular , Ratones Desnudos , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo
18.
Int J Mol Sci ; 25(15)2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39125775

RESUMEN

Multimodality reporter gene imaging combines the sensitivity, resolution and translational potential of two or more signals. The approach has not been widely adopted by the animal imaging community, mainly because its utility in this area is unproven. We developed a new complementation-based reporter gene system where the large component of split NanoLuc luciferase (LgBiT) presented on the surface of cells (TM-LgBiT) interacts with a radiotracer consisting of the high-affinity complementary HiBiT peptide labeled with a radionuclide. Radiotracer uptake could be imaged in mice using SPECT/CT and bioluminescence within two hours of implanting reporter-gene-expressing cells. Imaging data were validated by ex vivo biodistribution studies. Following the demonstration of complementation between the TM-LgBiT protein and HiBiT radiotracer, we validated the use of the technology in the highly specific in vivo multimodal imaging of cells. These findings highlight the potential of this new approach to facilitate the advancement of cell and gene therapies from bench to clinic.


Asunto(s)
Genes Reporteros , Luciferasas , Animales , Ratones , Luciferasas/metabolismo , Luciferasas/genética , Humanos , Distribución Tisular , Imagen Óptica/métodos , Mediciones Luminiscentes/métodos , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único/métodos , Cintigrafía/métodos , Línea Celular Tumoral
19.
J Nanobiotechnology ; 22(1): 467, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39103899

RESUMEN

BACKGROUND: The elastomechanical properties of nanocarriers have recently been discussed as important for the efficient delivery of various therapeutics. Some data indicate that optimal nanocarriers' elasticity can modulate in vivo nanocarrier stability, interaction with phagocytes, and uptake by target cells. Here, we presented a study to extensively analyze the in vivo behavior of LIP-SS liposomes that were modified by forming the silicone network within the lipid bilayers to improve their elastomechanical properties. We verified liposome pharmacokinetic profiles and biodistribution, including retention in tumors on a mouse model of breast cancer, while biocompatibility was analyzed on healthy mice. RESULTS: We showed that fluorescently labeled LIP-SS and control LIP-CAT liposomes had similar pharmacokinetic profiles, biodistribution, and retention in tumors, indicating that modified elasticity did not improve nanocarrier in vivo performance. Interestingly, biocompatibility studies revealed no changes in blood morphology, liver, spleen, and kidney function but indicated prolonged activation of immune response manifesting in increased concentration of proinflammatory cytokines in sera of animals exposed to all tested liposomes. CONCLUSION: Incorporating the silicone layer into the liposome structure did not change nanocarriers' characteristics in vivo. Further modification of the LIP-SS surface, including decoration with hydrophilic stealth polymers, should be performed to improve their pharmacokinetics and retention in tumors significantly. Activation of the immune response by LIP-SS and LIP-CAT, resulting in elevated inflammatory cytokine production, requires detailed studies to elucidate its mechanism.


Asunto(s)
Elasticidad , Liposomas , Siliconas , Liposomas/química , Animales , Ratones , Femenino , Siliconas/química , Distribución Tisular , Ratones Endogámicos BALB C , Línea Celular Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Membrana Dobles de Lípidos/química , Portadores de Fármacos/química
20.
Nat Commun ; 15(1): 6729, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112493

RESUMEN

Protein therapeutics are essential in treating various diseases, but their inherent biological instability and short circulatory half-lives in vivo pose challenges. Herein, a quantitative one-pot iterative living polymerization technique is reported towards precision control over the molecular structure and monomer sequence of protein-polymer conjugates, aiming to maximize physicochemical properties and biological functions of proteins. Using this quantitative one-pot iterative living polymerization technique, we successfully develop a series of sequence-controlled protein-multiblock polymer conjugates, enhancing their biostability, pharmacokinetics, cellular uptake, and in vivo biodistribution. All-atom molecular dynamics simulations are performed to disclose the definite sequence-function relationship of the bioconjugates, further demonstrating their sequence-encoded cellular uptake behavior and in vivo biodistribution in mice. Overall, this work provides a robust approach for creating precision protein-polymer conjugates with defined sequences and advanced functions as a promising candidate in disease treatment.


Asunto(s)
Simulación de Dinámica Molecular , Polimerizacion , Polímeros , Animales , Ratones , Polímeros/química , Distribución Tisular , Proteínas/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA