Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 13(26): 30434-30457, 2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34161080

RESUMEN

In the face of the abundant production of various types of carbapenemases, the antibacterial efficiency of imipenem, seen as "the last line of defense", is weakening. Following, the incidence of carbapenem-resistant Acinetobacter baumannii (CRAB), which can generate antibiotic-resistant biofilms, is increasing. Based on the superior antimicrobial activity of silver nanoparticles against multifarious bacterial strains compared with common antibiotics, we constructed the IPM@AgNPs-PEG-NOTA nanocomposite (silver nanoparticles were coated with SH-PEG-NOTA as well as loaded by imipenem) whose core was a silver nanoparticle to address the current challenge, and IPM@AgNPs-PEG-NOTA was able to function as a novel smart pH-sensitive nanodrug system. Synergistic bactericidal effects of silver nanoparticles and imipenem as well as drug-resistance reversal via protection of the ß-ring of carbapenem due to AgNPs-PEG-NOTA were observed; thus, this nanocomposite confers multiple advantages for efficient antibacterial activity. Additionally, IPM@AgNPs-PEG-NOTA not only offers immune regulation and accelerates tissue repair to improve therapeutic efficacy in vivo but also can prevent the interaction of pathogens and hosts. Compared with free imipenem or silver nanoparticles, this platform significantly enhanced antibacterial efficiency while increasing reactive oxygen species (ROS) production and membrane damage, as well as affecting cell wall formation and metabolic pathways. According to the results of crystal violet staining, LIVE/DEAD backlight bacterial viability staining, and real-time quantitative polymerase chain reaction (RT-qPCR), this silver nanocomposite downregulated the levels of ompA expression to prevent formation of biofilms. In summary, this research demonstrated that the IPM@AgNPs-PEG-NOTA nanocomposite is a promising antibacterial agent of security, pH sensitivity, and high efficiency in reversing resistance and synergistically combatting carbapenem-resistant A. baumannii. In the future, various embellishments and selected loads for silver nanoparticles will be the focus of research in the domains of medicine and nanotechnology.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/uso terapéutico , Portadores de Fármacos/química , Nanocompuestos/uso terapéutico , Plata/uso terapéutico , Acinetobacter baumannii/fisiología , Animales , Antibacterianos/química , Biopelículas/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Pared Celular/efectos de los fármacos , Liberación de Fármacos , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Imipenem/química , Imipenem/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Nanocompuestos/química , Polietilenglicoles/química , Especies Reactivas de Oxígeno/metabolismo , Plata/química , Resistencia betalactámica/efectos de los fármacos
2.
Int J Mol Sci ; 21(9)2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397443

RESUMEN

Iminosugars are sugar analogues endowed with a high pharmacological potential. The wide range of biological activities exhibited by these glycomimetics associated with their excellent drug profile make them attractive therapeutic candidates for several medical interventions. The ability of iminosugars to act as inhibitors or enhancers of carbohydrate-processing enzymes suggests their potential use as therapeutics for the treatment of cystic fibrosis (CF). Herein we review the most relevant advances in the field, paying attention to both the chemical synthesis of the iminosugars and their biological evaluations, resulting from in vitro and in vivo assays. Starting from the example of the marketed drug NBDNJ (N-butyl deoxynojirimycin), a variety of iminosugars have exhibited the capacity to rescue the trafficking of F508del-CFTR (deletion of F508 residue in the CF transmembrane conductance regulator), either alone or in combination with other correctors. Interesting results have also been obtained when iminosugars were considered as anti-inflammatory agents in CF lung disease. The data herein reported demonstrate that iminosugars hold considerable potential to be applied for both therapeutic purposes.


Asunto(s)
Fibrosis Quística/tratamiento farmacológico , Compuestos Heterocíclicos con 1 Anillo/uso terapéutico , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/uso terapéutico , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Glicósido Hidrolasas/antagonistas & inhibidores , Glicosiltransferasas/antagonistas & inhibidores , Compuestos Heterocíclicos con 1 Anillo/síntesis química , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Iminopiranosas/química , Iminopiranosas/uso terapéutico , Inflamación , Estructura Molecular , Mutación , Eliminación de Secuencia , Tartratos/química , Tartratos/uso terapéutico
3.
Faraday Discuss ; 222(0): 362-383, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32108214

RESUMEN

Bimodal systems for nuclear and optical imaging are currently being intensively investigated due to their comparable detection sensitivity and the complementary information they provide. In this perspective, we have implemented both modalities on biocompatible ultrasmall silicon nanoparticles (Si NPs). Such nanoparticles are particularly interesting since they are highly biocompatible, have covalent surface functionalization and demonstrate very fast body clearance. We prepared monodisperse citrate-stabilized Si NPs (2.4 ± 0.5 nm) with more than 40 accessible terminal amino groups per particle and, for the first time, simultaneously, a near-infrared dye (IR800-CW) and a radiolabel (64Cu-NOTA = 1,4,7-triazacyclononane-1,4,7-triacetic acid) have been covalently linked to the surface of such Si NPs. The obtained nanomaterials have been fully characterized using HR-TEM, XPS, UV-Vis and FT-IR spectroscopy. These dual-labelled particles do not exhibit any cytotoxicity in vitro. In vivo studies employing both positron emission tomography (PET) and optical imaging (OI) techniques revealed rapid renal clearance of dual-labelled Si NPs from mice.


Asunto(s)
Radioisótopos de Cobre/química , Compuestos Heterocíclicos con 1 Anillo/química , Imagen Multimodal/métodos , Nanopartículas/química , Silicio/química , Coloración y Etiquetado/métodos , Animales , Complejos de Coordinación/síntesis química , Complejos de Coordinación/farmacocinética , Femenino , Inyecciones Intravenosas , Masculino , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Imagen Óptica/métodos , Tamaño de la Partícula , Silicio/farmacocinética
4.
Int J Pharm ; 576: 118954, 2020 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-31935470

RESUMEN

The cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway plays an important role in tumor development and formation of metastases. It was earlier reported that cyclodextrin derivatives have a high affinity to form complexes with PGE2. Based on these results radiolabeled cyclodextrins - as new radiopharmaceuticals - may open a new pathway in the in vivo imaging and diagnosis of PGE2 positive tumors. The aims of this study were to synthetize the PGE2 specific 68Ga-labeled NODAGA-randomly methylated beta-cyclodextrin (68Ga-NODAGA-RAMEB) and investigate its tumor-targeting properties. NODAGA-RAMEB was labeled with Gallium-68 (68Ga), and the radiochemical purity (RCP%), partition coefficient (logP values), and in vitro-in vivo stability of 68Ga-NODAGA-RAMEB were determined. After intravenous injection of 68Ga-NODAGA-RAMEB the accumulation in organs and tissues was monitored in vivo by positron emission tomography (PET) and ex vivo by gamma counter in BxPC-3 and PancTu-1 tumor-bearing CB17 SCID mice. The RCP% of the newly synthesized 68Ga-NODAGA-RAMEB was higher than 98%. The molar activity was 15.34 ± 1.93 GBq/µmol. The logP of 68Ga labeled NODAGA-RAMEB was - 3.63 ± 0.04. Biodistribution studies showed high accumulation of 68Ga-NODAGA-RAMEB in PGE2 positive BxPC-3 tumors; approximately 15-20-fold higher radiotracer uptake was observed, than that of the background. 68Ga-labeled RAMEB is a promising radiotracer in PET diagnostics of PGE2 positive tumors.


Asunto(s)
Dinoprostona/metabolismo , Evaluación Preclínica de Medicamentos , Radioisótopos de Galio/administración & dosificación , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Radiofármacos/metabolismo , beta-Ciclodextrinas/administración & dosificación , Acetatos/administración & dosificación , Acetatos/química , Acetatos/metabolismo , Animales , Línea Celular Tumoral , Radioisótopos de Galio/química , Radioisótopos de Galio/metabolismo , Compuestos Heterocíclicos con 1 Anillo/administración & dosificación , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Neoplasias/metabolismo , Tomografía de Emisión de Positrones/métodos , Radiofármacos/administración & dosificación , Radiofármacos/química , Distribución Tisular/fisiología , beta-Ciclodextrinas/química , beta-Ciclodextrinas/metabolismo
5.
Clin Cancer Res ; 26(6): 1318-1326, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31843751

RESUMEN

PURPOSE: Tumor periphery and lymph nodes of tumor-induced lymphangiogenesis often abundantly express VEGFR-3. In our previous study, we identified a 5-amino acid peptide named TMVP1, which binds specifically to VEGFR-3. The objective of this study was to develop a novel 68Ga-labeled TMVP1 for VEGFR-3 PET imaging and to investigate its safety, biodistribution, and tumor-localizing efficacy in xenograft tumor models and a small cohort of patients with recurrent ovarian and cervical cancer. EXPERIMENTAL DESIGN: The DOTA-conjugated TMVP1 peptide was labeled with radionuclide 68Ga. SPR and saturation binding assays were used for the receptor-binding studies. Gynecologic xenograft tumors were employed for small-animal PET imaging and biodistribution of 68Ga-DOTA-TMVP1 in vivo. In the clinical study, 5 healthy volunteers and 8 patients with gynecologic cancer underwent whole-body PET/CT after being injected with 68Ga-DOTA-TMVP1. RESULTS: DOTA-TMVP1 was successfully labeled with 68Ga. LECs showed higher binding capacity with 68Ga-DOTA-TMVP1 than LEC(shVEGFR-3) and human umbilical vein endothelial cells. In mice with subcutaneous C33-A and SKOV-3 xenografts, the tracer was rapidly eliminated through the kidney to the bladder, and the small-animal PET/CT helped to clearly visualize the tumors. In patients with recurrent ovarian cancer and cervical cancer, tracer accumulation well above the background level was demonstrated in most identified sites of disease; especially with recurrent endodermal sinus tumors, the diagnostic value of 68Ga-DOTA-TMVP1 was comparable with that of 18F-FDG PET/CT. CONCLUSIONS: 68Ga-DOTA-TMVP1 is a potential PET tracer for imaging VEGFR-3 with favorable pharmacokinetics.


Asunto(s)
Fluorodesoxiglucosa F18/farmacocinética , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Neoplasias Ováricas/patología , Fragmentos de Péptidos/farmacocinética , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Neoplasias del Cuello Uterino/patología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Fluorodesoxiglucosa F18/química , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/metabolismo , Fragmentos de Péptidos/química , Radiofármacos/química , Radiofármacos/farmacocinética , Distribución Tisular , Neoplasias del Cuello Uterino/diagnóstico por imagen , Neoplasias del Cuello Uterino/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Artículo en Inglés | MEDLINE | ID: mdl-30373792

RESUMEN

Artesunate (AS), a semisynthetic artemisinin approved for malaria therapy, inhibits human cytomegalovirus (HCMV) replication in vitro, but therapeutic success in humans has been variable. We hypothesized that the short in vivo half-life of AS may contribute to the different treatment outcomes. We tested novel synthetic ozonides with longer half-lives against HCMV in vitro and mouse cytomegalovirus (MCMV) in vivo Screening of the activities of four ozonides against a pp28-luciferase-expressing HCMV Towne recombinant identified OZ418 to have the best selectivity; its effective concentration inhibiting viral growth by 50% (EC50) was 9.8 ± 0.2 µM, and cytotoxicity in noninfected human fibroblasts (the concentration inhibiting cell growth by 50% [CC50]) was 128.1 ± 8.0 µM. In plaque reduction assays, OZ418 inhibited HCMV TB40 in a concentration-dependent manner as well as a ganciclovir (GCV)-resistant HCMV isolate. The combination of OZ418 and GCV was synergistic in HCMV inhibition in vitro Virus inhibition by OZ418 occurred at an early stage and was dependent on the cell density at the time of infection. OZ418 treatment reversed HCMV-mediated cell cycle progression and correlated with the reduction of HCMV-induced expression of pRb, E2F1, and cyclin-dependent kinases 1, 2, 4, and 6. In an MCMV model, once-daily oral administration of OZ418 had significantly improved efficacy against MCMV compared to that of twice-daily oral AS. A parallel pharmacokinetic study with a single oral dose of OZ418 or AS showed a prolonged plasma half-life and higher unbound concentrations of OZ418 than unbound concentrations of AS. In summary, ozonides are proposed to be potential therapeutics, alone or in combination with GCV, for HCMV infection in humans.


Asunto(s)
Antivirales/farmacología , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Compuestos Heterocíclicos con 1 Anillo/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Compuestos de Espiro/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/sangre , Antivirales/química , Antivirales/farmacocinética , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Línea Celular , Citomegalovirus/genética , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/virología , Ganciclovir/farmacología , Regulación de la Expresión Génica , Compuestos Heterocíclicos con 1 Anillo/sangre , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Compuestos de Espiro/sangre , Compuestos de Espiro/química , Compuestos de Espiro/farmacocinética
7.
Small ; 14(35): e1801612, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30084540

RESUMEN

The need for better imaging assisted cancer therapy calls for new biocompatible agents with excellent imaging and therapeutic capabilities. This study successfully fabricates albumin-cooperated human serum albumin (HSA)-GGD-ICG nanoparticles (NPs), which are comprised of a magnetic resonance (MR) contrast agent, glycyrrhetinic-acid-modified gadolinium (III)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetate (GGD), and a fluorescence (FL) dye, indocyanine green (ICG), for multimodal MR/FL imaging assisted cancer therapy. These HSA-GGD-ICG NPs with excellent biocompatibility are stable under physiological conditions, and exhibit enhanced T1 contrast capability and improved fluorescence imaging capacity. In vitro experiments reveal an apparent effect of the NPs in killing tumor cells under low laser irradiation, due to the enhanced photothermal conversion efficiency (≈85.1%). Importantly, multimodal MR/FL imaging clearly shows the in vivo behaviors and the efficiency of tumor accumulation of HSA-GGD-ICG NPs, as confirmed by a pharmacokinetic study. With the guidance of multimodal imaging, photothermal therapy is subsequently conducted, which demonstrates again high photothermal conversion capability for eliminating tumors without relapse. Notably, real-time monitoring of tumor ablation for prognosis and therapy evaluation is also achieved by MR imaging. This strategy of constructing nanoplatforms through albumin-mediated methods is both convenient and efficient, which would enlighten the design of multimodal imaging assisted cancer therapy for potential clinical translation.


Asunto(s)
Materiales Biocompatibles/química , Hipertermia Inducida , Imagen por Resonancia Magnética , Nanopartículas/química , Imagen Óptica , Fototerapia , Animales , Compuestos Aza/química , Línea Celular Tumoral , Terapia Combinada , Compuestos Heterocíclicos con 1 Anillo/química , Verde de Indocianina/química , Ratones , Nanopartículas/ultraestructura , Fantasmas de Imagen , Pronóstico , Albúmina Sérica Humana/química , Temperatura
8.
J Nucl Med ; 59(8): 1234-1242, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29674421

RESUMEN

We have developed the 16F12 mouse monoclonal antibody (mAb), which targets the Müllerian-inhibiting substance receptor, type II (MISRII), expressed by ovarian tumors. Here, we assessed in preclinical models the possibility of using radiolabeled 16F12 in a theranostic approach for small-volume ovarian peritoneal carcinomatosis, such as after cytoreductive surgery. Methods: DOTA-, DTPA- or deferoxamine mesylate-conjugated 16F12 mAb was radiolabeled with ß-particle (177Lu) or α-particle (213Bi) emitters for therapeutic use and with 89Zr for PET imaging. On the 13th postxenograft day, mice bearing intraperitoneal MISRII-positive AN3CA endometrial carcinoma cell xenografts were treated by conventional intraperitoneal radioimmunotherapy (IP-RIT) with 10 MBq of 177Lu-16F12 or 12.9 MBq of 213Bi-16F12 or by brief intraperitoneal radioimmunotherapy (BIP-RIT) using 50 MBq of 177Lu-16F12 or 37 MBq of 213Bi-16F12. For BIP-RIT, 30 min after injection of the radiolabeled mAbs, the peritoneal cavity was washed to remove the unbound radioactivity. The biodistribution of 177Lu- and 213Bi-16F12 mAbs was determined and then used for dose assessment. Hematologic toxicity was also monitored. Results: The 16F12 mAb was satisfactorily radiolabeled for both therapy and imaging. IP-RIT with 177Lu-16F12 was slightly more efficient in delaying tumor growth than IP-RIT with 213Bi-16F12. Conversely, 213Bi-16F12 was more efficient than 177Lu-16F12 in BIP-RIT. The biodistribution analysis showed that the tumor-to-blood uptake ratio was significantly higher with BIP-RIT than with IP-RIT for both 213Bi- and 177Lu-16F12. Hematologic toxicity was more pronounced with 177Lu-16F12 than with 213Bi-16F12. SPECT/CT images (after BIP-RIT with 177Lu-16F12) and PET/CT images (after injection of 89Zr-16F12 in the tail vein) showed focal uptake at the tumor site. Conclusion: Radiolabeled 16F12 could represent a new theranostic tool for small-volume ovarian peritoneal carcinomatosis. Specifically, 213Bi-16F12-based BIP-RIT could be proposed to selected patients as an alternative adjuvant treatment immediately after cytoreductive surgery. An anti-MISRII mAb is currently being used in a first-in-human study, thus making radiolabeled anti-MISRII mAbs a realistic theranostic option for the clinic.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/radioterapia , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Animales , Anticuerpos Monoclonales/farmacocinética , Línea Celular Tumoral , Deferoxamina/química , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Marcaje Isotópico , Ratones , Neoplasias Ováricas/metabolismo , Ácido Pentético/química , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radioquímica , Distribución Tisular
9.
Chem Biol Interact ; 267: 80-88, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27038878

RESUMEN

The potential consequences of a major radiological event are not only large-scale external radiation exposure of the population, but also uncontrolled dissemination of, and internal contamination with, radionuclides. When planning an emergency response to radiological and nuclear incidents, one must consider the need for not only post-exposure treatment for contaminated individuals, but also prophylactic measures to protect the workforce facing contaminated areas and patients in the aftermath of such events. In addition to meeting the desired criteria for post-exposure treatments such as safety, ease of administration, and broad-spectrum efficacy against multiple radionuclides and levels of challenge, ideal prophylactic countermeasures must include rapid onset; induce minimal to no performance-decrementing side effects; be compatible with current military Chemical, Biological, Radiological, Nuclear, and Explosive countermeasures; and require minimal logistical burdens. Hydroxypyridinone-based actinide decorporation agents have shown the most promise as decorporation strategies for various radionuclides of concern, including the actinides plutonium and americium. The studies presented here probe the extent of plutonium decorporation efficacy for two chelating agents, 3,4,3-LI(1,2-HOPO) and 5-LIO(Me-3,2-HOPO), from early pre-exposure time points to a delay of up to 7 days in parenteral or oral treatment administration, i.e., well beyond the initial hours of emergency response. Despite delayed treatment after a contamination event, both ligands clearly enhanced plutonium elimination through the investigated 7-day post-treatment period. In addition, a remarkable prophylactic efficacy was revealed for 3,4,3-LI(1,2-HOPO) with treatment as early as 48 h before the plutonium challenge. This work provides new perspectives in the indication and use of experimental actinide decorporation treatments.


Asunto(s)
Quelantes/química , Descontaminación/métodos , Plutonio/química , Piridonas/química , Americio/análisis , Americio/química , Animales , Heces/química , Compuestos Heterocíclicos con 1 Anillo/química , Ratones , Plutonio/análisis , Plutonio/orina , Factores de Tiempo
10.
Bioconjug Chem ; 27(10): 2460-2468, 2016 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-27617674

RESUMEN

Radioimmunotherapy (RIT) delivers radioisotopes to antigen-expressing cells via monoantibodies for the imaging of lesions or medical therapy. The chelates are typically conjugated to the antibody through cysteine or lysine residues, resulting in heterogeneous chelate-to-antibody ratios and various conjugation sites. To overcome this heterogeneity, we have developed an approach for site-specific radiolabeling of antibodies by combination of genetic code expansion and click chemistry. As a proof-of-concept study, model systems including anti-CD20 antibody rituximab, positron-emitting isotope 64Cu, and a newly synthesized bifunctional linker (4-dibenzocyclooctynol-1,4,7,10-tetraazacyclotetradecane-1,4,7,10-tetraacetic acid, DIBO-DOTA) were used. The approach consists of three steps: (1) site-specific incorporation of an azido group-bearing amino acid (NEAK) via the genetic code expansion technique at the defined sites of the antibody as a "chemical handle"; (2) site-specific and quantitative conjugation of bifunctional linkers with the antibodies under a mild condition; and (3) radiolabeling of the chelate-modified antibodies with the appropriate isotope. We used heavy-chain A122NEAK rituximab as proof-of-concept and obtained a homogeneous radioconjugate with precisely two chelates per antibody, incorporated only at the chosen sites. The conjugation did not alter the binding and pharmacokinetics of the rituximab, as indicated by in vitro assays and in vivo PET imaging. We believe our research is a good supplement to the genetic code expansion technique for the development of novel radioimmunoconjugates.


Asunto(s)
Radioisótopos de Cobre/química , Inmunoconjugados/química , Tomografía de Emisión de Positrones/métodos , Radioinmunoterapia/métodos , Rituximab/química , Animales , Benzoxazinas/química , Quelantes/química , Técnicas de Química Sintética , Estabilidad de Medicamentos , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Marcaje Isotópico/métodos , Lutecio/química , Ratones SCID , Radioisótopos/química , Rituximab/genética , Rituximab/farmacocinética
11.
ACS Nano ; 10(1): 1417-24, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26731174

RESUMEN

A pretargeted oncologic positron emission tomography (PET) imaging that leverages the power of supramolecular nanoparticles with in vivo bioorthogonal chemistry was demonstrated for the clinically relevant problem of tumor imaging. The advantages of this approach are that (i) the pharmacokinetics (PKs) of tumor-targeting and imaging agents can be independently altered via chemical alteration to achieve the desired in vivo performance and (ii) the interplay between the two PKs and other controllable variables confers a second layer of control toward improved PET imaging. In brief, we utilized supramolecular chemistry to synthesize tumor-targeting nanoparticles containing transcyclooctene (TCO, a bioorthogonal reactive motif), called TCO⊂SNPs. After the intravenous injection and subsequent concentration of the TCO⊂SNPs in the tumors of living mice, a small molecule containing both the complementary bioorthogonal motif (tetrazine, Tz) and a positron-emitting radioisotope ((64)Cu) was injected to react selectively and irreversibly to TCO. High-contrast PET imaging of the tumor mass was accomplished after the rapid clearance of the unreacted (64)Cu-Tz probe. Our nanoparticle approach encompasses a wider gamut of tumor types due to the use of EPR effects, which is a universal phenomenon for most solid tumors.


Asunto(s)
Ciclooctanos/química , Glioblastoma/diagnóstico por imagen , Glioblastoma/diagnóstico , Compuestos Heterocíclicos con 1 Anillo/química , Nanopartículas/química , Tomografía de Emisión de Positrones/métodos , Animales , Radioisótopos de Cobre/administración & dosificación , Radioisótopos de Cobre/química , Dendrímeros/química , Glioblastoma/patología , Compuestos Heterocíclicos con 1 Anillo/administración & dosificación , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Desnudos , Nanopartículas/ultraestructura , Trasplante de Neoplasias , Permeabilidad , Polietilenos/química , Trasplante Heterólogo
12.
Biomaterials ; 50: 30-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25736493

RESUMEN

Alginate hydrogels are well-characterized, biologically inert materials that are used in many biomedical applications for the delivery of drugs, proteins, and cells. Unfortunately, canonical covalently crosslinked alginate hydrogels are formed using chemical strategies that can be biologically harmful due to their lack of chemoselectivity. In this work we introduce tetrazine and norbornene groups to alginate polymer chains and subsequently form covalently crosslinked click alginate hydrogels capable of encapsulating cells without damaging them. The rapid, bioorthogonal, and specific click reaction is irreversible and allows for easy incorporation of cells with high post-encapsulation viability. The swelling and mechanical properties of the click alginate hydrogel can be tuned via the total polymer concentration and the stoichiometric ratio of the complementary click functional groups. The click alginate hydrogel can be modified after gelation to display cell adhesion peptides for 2D cell culture using thiol-ene chemistry. Furthermore, click alginate hydrogels are minimally inflammatory, maintain structural integrity over several months, and reject cell infiltration when injected subcutaneously in mice. Click alginate hydrogels combine the numerous benefits of alginate hydrogels with powerful bioorthogonal click chemistry for use in tissue engineering applications involving the stable encapsulation or delivery of cells or bioactive molecules.


Asunto(s)
Alginatos/química , Química Clic/métodos , Reactivos de Enlaces Cruzados/química , Compuestos Heterocíclicos con 1 Anillo/química , Norbornanos/química , Alginatos/síntesis química , Alginatos/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Inmovilizadas/efectos de los fármacos , Células Inmovilizadas/metabolismo , Fuerza Compresiva/efectos de los fármacos , Módulo de Elasticidad/efectos de los fármacos , Femenino , Ácido Glucurónico/síntesis química , Ácido Glucurónico/química , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/síntesis química , Ácidos Hexurónicos/química , Ácidos Hexurónicos/farmacología , Hidrogeles/farmacología , Inyecciones , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Oligopéptidos/farmacología
13.
J Pharm Sci ; 104(5): 1832-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25727482

RESUMEN

The hydroxypyridinonate ligand 3,4,3-LI(1,2-HOPO) is currently under development for radionuclide chelation therapy. The preclinical characterization of this highly promising ligand comprised the evaluation of its in vitro properties, including microsomal, plasma, and gastrointestinal fluid stability, cytochrome P450 inhibition, plasma protein binding, and intestinal absorption using the Caco-2 cell line. When mixed with active human liver microsomes, no loss of parent compound was observed after 60 min, indicating compound stability in the presence of liver microsomal P450. At the tested concentrations, 3,4,3-LI(1,2-HOPO) did not significantly influence the activities of any of the cytochromal isoforms screened. Thus, 3,4,3-LI(1,2-HOPO) is unlikely to cause drug-drug interactions by inhibiting the metabolic clearance of coadministered drugs metabolized by these enzymes. Plasma protein-binding assays revealed that the compound is protein-bound in dogs and less extensively in rats and humans. In the plasma stability study, the compound was stable after 1 h at 37°C in mouse, rat, dog, and human plasma samples. Finally, a bidirectional permeability assay demonstrated that 3,4,3-LI(1,2-HOPO) is not permeable across the Caco-2 monolayer, highlighting the need to further evaluate the effects of various compounds with known permeability enhancement properties on the permeability of the ligand in future studies.


Asunto(s)
Elementos de Series Actinoides/metabolismo , Quelantes/química , Quelantes/metabolismo , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/metabolismo , Piridonas/química , Piridonas/metabolismo , Animales , Células CACO-2 , Perros , Estabilidad de Medicamentos , Femenino , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Ratas
14.
Int J Radiat Biol ; 90(11): 1055-61, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24937372

RESUMEN

PURPOSE: To characterize the dose-dependent and sex-related efficacy of the hydroxypyridinonate decorporation agent 3,4,3-LI(1,2-HOPO) at enhancing plutonium elimination when post-exposure treatment is delayed. MATERIALS AND METHODS: Six parenteral dose levels of 3,4,3-LI(1,2-HOPO) from 1-300 µmol/kg were evaluated for decorporating plutonium in female and male Swiss-Webster mice administered a soluble citrate complex of (238)Pu and treated 24 hours later. Necropsies were scheduled at four time-points (2, 4, 8, and 15 days post-contamination) for the female groups and at three time-points (2, 4, and 8 days post-contamination) for the male groups. RESULTS: Elimination enhancement was dose-dependent in the 1-100 µmol/kg dose range at all necropsy time-points, with some significant reductions in full body and tissue content for both female and male animals. The highest dose level resulted in slight toxicity, with a short recovery period, which delayed excretion of the radionuclide. CONCLUSIONS: While differences were noted between the female and male cohorts in efficacy range and recovery times, all groups displayed sustained dose-dependent (238)Pu elimination enhancement after delayed parenteral treatment with 3,4,3-LI(1,2-HOPO), the actinide decorporation agent under development.


Asunto(s)
Terapia por Quelación/métodos , Compuestos Heterocíclicos con 1 Anillo/química , Plutonio/efectos adversos , Piridonas/química , Animales , Carga Corporal (Radioterapia) , Quelantes/química , Relación Dosis-Respuesta a Droga , Femenino , Riñón/efectos de la radiación , Hígado/efectos de la radiación , Masculino , Ratones , Plutonio/química , Piridonas/uso terapéutico , Factores Sexuales , Factores de Tiempo
15.
Nucl Med Biol ; 41(3): 282-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24461626

RESUMEN

INTRODUCTION: The treatment of prostate cancer using a radiotherapeutic (90)Y labeled N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer can be enhanced with localized tumor hyperthermia. An (111)In labeled HPMA copolymer system for single photon emission computerized tomography (SPECT) was developed to observe the biodistribution changes associated with hyperthermia. Efficacy studies were conducted in prostate tumor bearing mice using the (90)Y HPMA copolymer with hyperthermia. METHODS: HPMA copolymers containing 1, 4, 7, 10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) were synthesized by reversible addition-fragmentation transfer (RAFT) copolymerization and subsequently labeled with either (111)In for imaging or (90)Y for efficacy studies. Radiolabel stability was characterized in vitro with mouse serum. Imaging and efficacy studies were conducted in DU145 prostate tumor bearing mice. Imaging was performed using single photon emission computerized tomography (SPECT). Localized mild tumor hyperthermia was achieved by plasmonic photothermal therapy using gold nanorods. RESULTS: HPMA copolymer-DOTA conjugates demonstrated efficient labeling and stability for both radionuclides. Imaging analysis showed a marked increase of radiolabeled copolymer within the hyperthermia treated prostate tumors, with no significant accumulation in non-targeted tissues. The greatest reduction in tumor growth was observed in the hyperthermia treated tumors with (90)Y HPMA copolymer conjugates. Histological analysis confirmed treatment efficacy and safety. CONCLUSION: HPMA copolymer-DOTA conjugates radiolabeled with both the imaging and treatment radioisotopes, when combined with hyperthermia can serve as an image guided approach for efficacious treatment of prostate tumors.


Asunto(s)
Acrilamidas/química , Acrilamidas/uso terapéutico , Oro/química , Oro/uso terapéutico , Hipertermia Inducida , Nanotubos , Neoplasias de la Próstata/radioterapia , Acrilamidas/farmacocinética , Animales , Línea Celular Tumoral , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Masculino , Ratones , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Tomografía Computarizada de Emisión de Fotón Único , Radioisótopos de Itrio/uso terapéutico
16.
Nucl Med Biol ; 40(8): 994-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24074944

RESUMEN

INTRODUCTION: Monitoring the effectiveness of therapy early and accurately continues to be challenging. We hypothesize that determination of Human Epidermal Growth Factor Receptor 2 (HER2) mRNA in malignant breast cancer (BC) cells by positron emission tomography (PET) imaging, before and after treatment, would reflect therapeutic efficacy. METHOD: WT4340, a peptide nucleic acid (PNA) 12-mer complementary to HER2 mRNA was synthesized together with -CSKC, a cyclic peptide, which facilitated internalization of the PNA via IGFR expressed on BC cells, and DOTA that chelated Cu-64. Mice (n = 8) with BT474 ER+/HER2+ human BC received doxorubicin (DOX, 1.5mg/kg) i.p. once a week for six weeks. Mice (n = 8) without DOX served as controls. All mice were PET imaged with F-18-FDG and 48 h later with Cu-64-WT4340. PET imaging were performed before and 72 h after each treatment. Standardized uptake values (SUVs) were determined and percent change calculated. Animal body weight (BW) and tumor volume (TV) were measured. RESULTS: SUVs for Cu-64-WT4340 after DOX treatment declined by 54% ± 17% after the second dose, 41% ± 15% after the fourth dose, and 29% ± 7% after the sixth dose, compared with 42% ± 22%, 31% ± 18%, and 13% ± 9% (p<0.05) for F-18-FDG. In untreated mice, the corresponding percent SUVs for Cu-64-WT4340 were 145% ± 82%, 165% ± 39%, and 212% ± 105% of pretreatment SUV, compared with 108% ± 28%, 151% ± 8%, and 152% ± 35.5%, (p<0.08) for F-18-FDG. TV in mice after second dose was 114.15% ± 61.83%, compared with 144.7% ± 64.4% for control mice. BW of DOX-treated mice was 103.4% ± 7.6% of pretreatment, vs. 100.1% ± 4.3% for control mice. CONCLUSION: Therapeutic efficacy was apparent sooner by molecular PET imaging than by determination of reduction in TV.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Ácidos Nucleicos de Péptidos , Tomografía de Emisión de Positrones , Receptor ErbB-2/genética , Animales , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Radioisótopos de Cobre , Doxorrubicina/uso terapéutico , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Factor I del Crecimiento Similar a la Insulina/química , Ratones , Ácidos Nucleicos de Péptidos/química , Ácidos Nucleicos de Péptidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
17.
Nucl Med Biol ; 40(7): 946-54, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23850487

RESUMEN

AIM: Integrin αvß3 plays a significant role in angiogenesis during tumor growth and metastasis, and is a receptor for the extracellular matrix proteins with the exposed arginine(R)-glycine(G)-aspartic acid(D) tripeptide sequence. The over-expression of integrin αvß3 during tumor growth and metastasis presents an interesting molecular target for both early detection and treatment of rapidly growing solid tumors. Considering the advantages of (177)Lu for targeted radiotherapy and enhanced tumor targeting capability of cyclic RGD peptide dimer, an attempt has been made to optimize the protocol for the preparation of clinical dose of (177)Lu labeled DOTA-E[c(RGDfK)]2 (E=Glutamic acid, f=phenyl alanine, K=lysine) as a potential agent for targeted tumor therapy. METHODS: (177)Lu was produced by thermal neutron bombardment on enriched Lu2O3 (82% in (176)Lu) target at a flux of 1 × 10(14) n/cm(2).s for 21 d. Therapeutic dose of (177)Lu-DOTA-E[c(RGDfK)]2 (7.4GBq) was prepared by adding the aqueous solution of the ligand and (177)LuCl3 to 0.1M NH4OAC buffer containing gentisic acid and incubating the reaction mixture at 90°C for 30 min. The yield and radiochemical purity of the complex was determined by HPLC technique. Parameters, such as, ligand-to-metal ratio, pH of the reaction mixture, incubation time and temperature were varied using tracer quantity of (177)Lu (37 MBq) in order to arrive at the optimized protocol for the preparation of clinical dose. Biological behavior of the radiotracer prepared was studied in C57/BL6 mice bearing melanoma tumors. RESULTS: (177)Lu was produced with a specific activity of 950 ± 50 GBq/mg (25.7 ± 1.4 Ci/mg) and radionuclidic purity of 99.98%. A careful optimization of several parameters showed that (177)Lu-DOTA-E[c(RGDfK)]2 could be prepared with adequately high radiochemical purity using a ligand-to-metal ratio ~2. Based on these studies therapeutic dose of the agent with 7.4 GBq of (177)Lu was formulated in ~63 GBq/µM specific activity with high yield (98.2 ± 0.7%), radiochemical purity and in vitro stability. Biodistribution studies carried out in C57/BL6 mice bearing melanoma tumors revealed specific accumulation of the radiolabeled conjugate in tumor (3.80 ± 0.55% ID/g at 30 min p.i.) with high tumor to blood and tumor to muscle ratios. However, the uptake of the radiotracer in the tumor was found to be reduced to 1.51 ± 0.32 %ID/g at 72 h p.i. CONCLUSIONS: The present work successfully demonstrates the formulation of an optimized protocol for the preparation of (177)Lu labeled DOTA-E[c(RGDfK)]2 for PRRT applications using (177)Lu produced by direct neutron activation in a medium flux research reactor.


Asunto(s)
Dimerización , Lutecio/uso terapéutico , Péptidos Cíclicos/química , Péptidos Cíclicos/uso terapéutico , Dosis de Radiación , Radioquímica/métodos , Radioisótopos/uso terapéutico , Animales , Estabilidad de Medicamentos , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Marcaje Isotópico , Melanoma/radioterapia , Ratones , Octanoles/química , Péptidos Cíclicos/farmacocinética , Trazadores Radiactivos , Agua/química
18.
Inorg Chem ; 52(15): 8461-6, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23837738

RESUMEN

Derivatives of 4-[2-(4-isothiocyanatophenyl)ethynyl]-2,6,-bis{[N,N-bis(carboxymethyl)-amino]methyl}pyridine europium(III) (1) bearing one (6) or two (7) additional iminodiacetate coordinating arms have been synthesized. 6 and 7 were significantly more stable than 1 as evidenced by competition experiments with ethylenediaminetetraacetic acid (EDTA) and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). While the luminescence quantum yield of 1 remained modest, the other two complexes displayed substantial luminescence efficiency. The introduction of a supplementary iminodiacetate arm in 6 brought important improvements to both the stability and the luminescence properties of the Eu complex. In contrast, although 7 is more luminescent than 1, the introduction of a second iminodiacetate coordinating arm brings no further benefit on the photophysical properties. The most promising results were obtained with the nine-dentate chelate 6 and its Eu complex, which was conjugated to biotin and applied within the frame of a bioaffinity immunoassay of human C-reactive protein.


Asunto(s)
Quelantes/química , Europio/química , Colorantes Fluorescentes/química , Inmunoensayo/métodos , Compuestos Organometálicos/química , Proteína C-Reactiva/análisis , Estabilidad de Medicamentos , Ácido Edético/química , Colorantes Fluorescentes/síntesis química , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Compuestos Organometálicos/síntesis química , Factores de Tiempo
19.
J Nucl Med ; 53(12): 1951-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23139086

RESUMEN

UNLABELLED: Terbium offers 4 clinically interesting radioisotopes with complementary physical decay characteristics: (149)Tb, (152)Tb, (155)Tb, and (161)Tb. The identical chemical characteristics of these radioisotopes allow the preparation of radiopharmaceuticals with identical pharmacokinetics useful for PET ((152)Tb) and SPECT diagnosis ((155)Tb) and for α- ((149)Tb) and ß(-)-particle ((161)Tb) therapy. The goal of this proof-of-concept study was to produce all 4 terbium radioisotopes and assess their diagnostic and therapeutic features in vivo when labeled with a folate-based targeting agent. METHODS: (161)Tb was produced by irradiation of (160)Gd targets with neutrons at Paul Scherrer Institute or Institut Laue-Langevin. After neutron capture, the short-lived (161)Gd decays to (161)Tb. (149)Tb, (152)Tb, and (155)Tb were produced by proton-induced spallation of tantalum targets, followed by an online isotope separation process at ISOLDE/CERN. The isotopes were purified by means of cation exchange chromatography. For the in vivo studies, we used the DOTA-folate conjugate cm09, which binds to folate receptor (FR)-positive KB tumor cells. Therapy experiments with (149)Tb-cm09 and (161)Tb-cm09 were performed in KB tumor-bearing nude mice. Diagnostic PET/CT ((152)Tb-cm09) and SPECT/CT ((155)Tb-cm09 and (161)Tb-cm09) studies were performed in the same tumor mouse model. RESULTS: Carrier-free terbium radioisotopes were obtained after purification, with activities ranging from approximately 6 MBq (for (149)Tb) to approximately 15 MBq (for (161)Tb). The radiolabeling of cm09 was achieved in a greater than 96% radiochemical yield for all terbium radioisotopes. Biodistribution studies showed high and specific uptake in FR-positive tumor xenografts (23.8% ± 2.5% at 4 h after injection, 22.0% ± 4.4% at 24 h after injection, and 18.4% ± 1.8% at 48 h after injection). Excellent tumor-to-background ratios at 24 h after injection (tumor to blood, ≈ 15; tumor to liver, ≈ 5.9; and tumor to kidney, ≈ 0.8) allowed the visualization of tumors in mice using PET ((152)Tb-cm09) and SPECT ((155)Tb-cm09 and (161)Tb-cm09). Compared with no therapy, α- ((149)Tb-cm09) and ß(-)-particle therapy ((161)Tb-cm09) resulted in a marked delay in tumor growth or even complete remission (33% for (149)Tb-cm09 and 80% for (161)Tb-cm09) and a significantly increased survival. CONCLUSION: For the first time, to our knowledge, 4 terbium radionuclides have been tested in parallel with tumor-bearing mice using an FR targeting agent. Along with excellent tumor visualization enabled by (152)Tb PET and (155)Tb SPECT, we demonstrated the therapeutic efficacy of the α-emitter (149)Tb and ß(-)-emitter (161)Tb.


Asunto(s)
Ácido Fólico/metabolismo , Tomografía de Emisión de Positrones/métodos , Radioisótopos/uso terapéutico , Terbio/uso terapéutico , Tomografía Computarizada de Emisión de Fotón Único/métodos , Partículas alfa/uso terapéutico , Animales , Partículas beta/uso terapéutico , Femenino , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/química , Ácido Fólico/uso terapéutico , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Células KB , Ratones
20.
Mol Imaging Biol ; 14(3): 366-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21732165

RESUMEN

PURPOSE: Targeting of cholecystokinin receptor expressing malignancies such as medullary thyroid carcinoma is currently limited by low in vivo stability of radioligands. To increase the stability, we have developed and preclinically evaluated two cyclic 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-minigastrin analogs radiolabeled with (111)In and (68)Ga. PROCEDURES: Radiolabeling efficiency, in vitro characterization, cholecystokinin receptor subtype 2 (CCK-2) binding in human tumor tissues, and cell internalization on CCK-2 receptor expressing AR42J cells, as well as biodistribution and small animal imaging in two different mouse xenograft models were studied. RESULTS: High receptor affinity and receptor-mediated uptake of the radioligands in AR42J cells was confirmed in vitro. (111)In-labeled cyclic DOTA-peptides showed a specific tumor uptake of ~1% ID/g in vivo, (68)Ga-labeled analogs of ~3% ID/g. Small animal SPECT imaging resulted to be superior with (111)In-DOTA-cyclo-MG2 in comparison with (111)In-DOTA-cyclo-MG1. CONCLUSIONS: Cyclic DOTA-minigastrin analogs are promising candidates for gastrin receptor scintigraphy and targeted radionuclide therapy.


Asunto(s)
Gastrinas/farmacocinética , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Receptores de Colecistoquinina/metabolismo , Neoplasias de la Tiroides/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Carcinoma Neuroendocrino , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Radioisótopos de Galio , Gastrinas/química , Compuestos Heterocíclicos con 1 Anillo/química , Radioisótopos de Indio , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Neoplasias de la Tiroides/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA