Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Med Chem Lett ; 11(5): 754-759, 2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32435381

RESUMEN

Lysine-specific demethylase 1 (LSD1 or KDM1A) is a FAD-dependent enzyme that acts as a transcription corepressor or coactivator by regulating the methylation status of histone H3 lysines K4 and K9, respectively. KDM1A represents an attractive target for cancer therapy. While, in the past, the main medicinal chemistry strategy toward KDM1A inhibition was based on the optimization of ligands that irreversibly bind the FAD cofactor within the enzyme catalytic site, we and others have also identified reversible inhibitors. Herein we reported the discovery of 5-imidazolylthieno[3,2-b]pyrroles, a new series of KDM1A inhibitors endowed with picomolar inhibitory potency, active in cells and efficacious after oral administration in murine leukemia models.

2.
ChemMedChem ; 15(7): 643-658, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32003940

RESUMEN

LSD1 is a lysine demethylase highly involved in initiation and development of cancer. To design highly effective covalent inhibitors, a strategy is to fill its large catalytic cleft by designing tranylcypromine (TCP) analogs decorated with long, hindered substituents. We prepared three series of TCP analogs, carrying aroyl- and arylacetylamino (1 a-h), Z-amino acylamino (2 a-o), or double-substituted benzamide (3 a-n) residues at the C4 or C3 position of the phenyl ring. Further fragments obtained by chemical manipulation applied on the TCP scaffold (compounds 4 a-i) were also prepared. When tested against LSD1, most of 1 and 3 exhibited IC50 values in the low nanomolar range, with 1 e and 3 a,d,f,g being also the most selective respect to monoamine oxidases. In MV4-11 AML and NB4 APL cells compounds 3 were the most potent, displaying up to sub-micromolar cell growth inhibition against both cell lines (3 a) or against NB4 cells (3 c). The most potent compounds in cellular assays were also able to induce the expression of LSD1 target genes, such as GFI-1b, ITGAM, and KCTD12, as functional read-out for LSD1 inhibition. Mouse and human intrinsic clearance data highlighted the high metabolic stability of compounds 3 a, 3 d and 3 g. Further studies will be performed on the new compounds 3 a and 3 c to assess their anticancer potential in different cancer contexts.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Histona Demetilasas/antagonistas & inhibidores , Tranilcipromina/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Relación Estructura-Actividad , Tranilcipromina/síntesis química , Tranilcipromina/química
3.
Eur J Med Chem ; 152: 283-297, 2018 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-29730191

RESUMEN

We designed new 3-arylthio- and 3-aroyl-1H-indole derivatives 3-22 bearing a heterocyclic ring at position 5, 6 or 7 of the indole nucleus. The 6- and 7-heterocyclyl-1H-indoles showed potent inhibition of tubulin polymerization, binding of colchicine to tubulin and growth of MCF-7 cancer cells. Compounds 13 and 19 inhibited a panel of cancer cells and the NCI/ADR-RES multidrug resistant cell line at low nanomolar concentrations. Compound 13 at 50 nM induced 77% G2/M in HeLa cells, and at 20 nM caused 50% stable arrest of mitosis. As an inhibitor of HepG2 cells (IC50 = 20 nM), 13 was 4-fold superior to 19. Compound 13 was a potent inhibitor of the human U87MG glioblastoma cells at nanomolar concentrations, being nearly one order of magnitude superior to previously reported arylthioindoles. The present results highlight 13 as a robust scaffold for the design of new anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Indoles/farmacología , Moduladores de Tubulina/farmacología , Tubulina (Proteína)/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/química , Estructura Molecular , Polimerizacion/efectos de los fármacos , Relación Estructura-Actividad , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/química , Células Tumorales Cultivadas
4.
ACS Med Chem Lett ; 8(5): 521-526, 2017 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-28523104

RESUMEN

We designed 3-aroyl-1,4-diarylpyrrole (ARDAP) derivatives as potential anticancer agents having different substituents at the 1- or 4-phenyl ring. ARDAP compounds exhibited potent inhibition of tubulin polymerization, binding of colchicine to tubulin, and cancer cell growth. ARDAP derivative 10 inhibited the proliferation of BCR/ABL-expressing KU812 and LAMA84 cells from chronic myeloid leukemia (CML) patients in blast crisis and of hematopoietic cells ectopically expressing the imatinib mesylate (IM)-sensitive KBM5-WT or its IM-resistant KBM5-T315I mutation. Compound 10 minimally affected the proliferation of normal blood cells, indicating that it may be a promising agent to overcome broad tyrosine kinase inhibitor resistance in relapsed/refractory CML patients. Compound 10 significantly decreased CML proliferation by inducing G2/M phase arrest and apoptosis via a mitochondria-dependent pathway. ARDAP 10 augmented the cytotoxic effects of IM in human CML cells. Compound 10 represents a robust lead compound to develop tubulin inhibitors with potential as novel treatments for CML.

5.
J Med Chem ; 60(5): 1673-1692, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28186755

RESUMEN

Lysine specific demethylase 1 KDM1A (LSD1) regulates histone methylation and it is increasingly recognized as a potential therapeutic target in oncology. We report on a high-throughput screening campaign performed on KDM1A/CoREST, using a time-resolved fluorescence resonance energy transfer (TR-FRET) technology, to identify reversible inhibitors. The screening led to 115 hits for which we determined biochemical IC50, thus identifying four chemical series. After data analysis, we have prioritized the chemical series of N-phenyl-4H-thieno[3, 2-b]pyrrole-5-carboxamide for which we obtained X-ray structures of the most potent hit (compound 19, IC50 = 2.9 µM) in complex with the enzyme. Initial expansion of this chemical class, both modifying core structure and decorating benzamide moiety, was directed toward the definition of the moieties responsible for the interaction with the enzyme. Preliminary optimization led to compound 90, which inhibited the enzyme with a submicromolar IC50 (0.162 µM), capable of inhibiting the target in cells.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Histona Demetilasas/antagonistas & inhibidores , Pirroles/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Diseño de Fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Espectroscopía de Protones por Resonancia Magnética , Pirroles/química , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad
6.
J Med Chem ; 60(5): 1693-1715, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28186757

RESUMEN

The balance of methylation levels at histone H3 lysine 4 (H3K4) is regulated by KDM1A (LSD1). KDM1A is overexpressed in several tumor types, thus representing an emerging target for the development of novel cancer therapeutics. We have previously described ( Part 1, DOI 10.1021.acs.jmedchem.6b01018 ) the identification of thieno[3,2-b]pyrrole-5-carboxamides as novel reversible inhibitors of KDM1A, whose preliminary exploration resulted in compound 2 with biochemical IC50 = 160 nM. We now report the structure-guided optimization of this chemical series based on multiple ligand/KDM1A-CoRest cocrystal structures, which led to several extremely potent inhibitors. In particular, compounds 46, 49, and 50 showed single-digit nanomolar IC50 values for in vitro inhibition of KDM1A, with high selectivity in secondary assays. In THP-1 cells, these compounds transcriptionally affected the expression of genes regulated by KDM1A such as CD14, CD11b, and CD86. Moreover, 49 and 50 showed a remarkable anticlonogenic cell growth effect on MLL-AF9 human leukemia cells.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Lisina/química , Pirroles/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/química , Transferencia Resonante de Energía de Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Histona Demetilasas , Humanos , Concentración 50 Inhibidora , Pirroles/química , Relación Estructura-Actividad
7.
J Med Chem ; 58(15): 5789-807, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26132075

RESUMEN

We designed 39 new 2-phenylindole derivatives as potential anticancer agents bearing the 3,4,5-trimethoxyphenyl moiety with a sulfur, ketone, or methylene bridging group at position 3 of the indole and with halogen or methoxy substituent(s) at positions 4-7. Compounds 33 and 44 strongly inhibited the growth of the P-glycoprotein-overexpressing multi-drug-resistant cell lines NCI/ADR-RES and Messa/Dx5. At 10 nM, 33 and 44 stimulated the cytotoxic activity of NK cells. At 20-50 nM, 33 and 44 arrested >80% of HeLa cells in the G2/M phase of the cell cycle, with stable arrest of mitotic progression. Cell cycle arrest was followed by cell death. Indoles 33, 44, and 81 showed strong inhibition of the SAG-induced Hedgehog signaling activation in NIH3T3 Shh-Light II cells with IC50 values of 19, 72, and 38 nM, respectively. Compounds of this class potently inhibited tubulin polymerization and cancer cell growth, including stimulation of natural killer cell cytotoxic activity and repression of Hedgehog-dependent cancer.


Asunto(s)
Citotoxicidad Inmunológica/efectos de los fármacos , Proteínas Hedgehog/fisiología , Indoles/farmacología , Células Asesinas Naturales/efectos de los fármacos , Mitosis/efectos de los fármacos , Neoplasias/patología , Tubulina (Proteína)/efectos de los fármacos , Animales , División Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Células Asesinas Naturales/inmunología , Ratones , Células 3T3 NIH , Neoplasias/inmunología , Tubulina (Proteína)/química
8.
J Med Chem ; 57(15): 6531-52, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25025991

RESUMEN

We synthesized 3-aroyl-1-arylpyrrole (ARAP) derivatives as potential anticancer agents having different substituents at the pendant 1-phenyl ring. Both the 1-phenyl ring and 3-(3,4,5-trimethoxyphenyl)carbonyl moieties were mandatory to achieve potent inhibition of tubulin polymerization, binding of colchicine to tubulin, and cancer cell growth. ARAP 22 showed strong inhibition of the P-glycoprotein-overexpressing NCI-ADR-RES and Messa/Dx5MDR cell lines. Compounds 22 and 27 suppressed in vitro the Hedgehog signaling pathway, strongly reducing luciferase activity in SAG treated NIH3T3 Shh-Light II cells, and inhibited the growth of medulloblastoma D283 cells at nanomolar concentrations. ARAPs 22 and 27 represent a new potent class of tubulin polymerization and cancer cell growth inhibitors with the potential to inhibit the Hedgehog signaling pathway.


Asunto(s)
Compuestos de Anilina/química , Antineoplásicos/química , Guanidinas/química , Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Pirroles/química , Moduladores de Tubulina/química , Compuestos de Anilina/síntesis química , Compuestos de Anilina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colchicina/química , Ensayos de Selección de Medicamentos Antitumorales , Guanidinas/síntesis química , Guanidinas/farmacología , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Ratones , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Polimerizacion , Unión Proteica , Pirroles/síntesis química , Pirroles/farmacología , Transducción de Señal , Relación Estructura-Actividad , Tubulina (Proteína)/química , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/farmacología
9.
ChemMedChem ; 9(7): 1574-85, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24753447

RESUMEN

Heat-shock protein 90 (Hsp90) is a molecular chaperone involved in the stabilization of key oncogenic signaling proteins, and therefore, inhibition of Hsp90 represents a new strategy in cancer therapy. 2-Amino-7-[4-fluoro-2-(3-pyridyl)phenyl]-4-methyl-7,8-dihydro-6H-quinazolin-5-one oxime is a racemic Hsp90 inhibitor that targets the N-terminal adenosine triphosphatase site. We developed a method to resolve the enantiomers and evaluated their inhibitory activity on Hsp90 and the consequent antitumor effects. The (S) stereoisomer emerged as a potent Hsp90 inhibitor in biochemical and cellular assays. In addition, this enantiomer exhibited high oral bioavailability in mice and excellent antitumor activity in two different human cancer xenograft models.


Asunto(s)
Antineoplásicos/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Oximas/química , Quinazolinonas/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sitios de Unión , Línea Celular Tumoral , Femenino , Células HCT116 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Desnudos , Microsomas/metabolismo , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Oximas/farmacología , Oximas/uso terapéutico , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Estereoisomerismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Eur J Med Chem ; 64: 273-84, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23644210

RESUMEN

Histone Deacetylases (HDACs) have become important targets for the treatment of cancer and other diseases. In previous studies we described the development of novel spirocyclic HDAC inhibitors based on the combination of privileged structures with hydroxamic acid moieties as zinc binding group. Herein, we report further explorations, which resulted in the discovery of a new class of spiro[2H-(1,3)-benzoxazine-2,4'-piperidine] derivatives. Several compounds showed good potency of around 100 nM and less in the HDAC inhibition assays, submicromolar IC50 values when tested against tumour cell lines and a remarkable stability in human and mouse microsomes. Two representative examples exhibited a good pharmacokinetic profile with an oral bioavailability equal or higher than 35% and one of them studied in an HCT116 murine xenograft model showing a robust tumour growth inhibition. In addition, the two benzoxazines were found to have a minor affinity for the hERG potassium channel compared to their corresponding ketone analogues.


Asunto(s)
Antineoplásicos/farmacología , Benzoxazinas/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Compuestos de Espiro/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzoxazinas/síntesis química , Benzoxazinas/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Células HeLa , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Humanos , Células K562 , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Neoplasias Experimentales/patología , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Relación Estructura-Actividad
11.
J Med Chem ; 56(1): 123-49, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23214452

RESUMEN

New arylthioindole derivatives having different cyclic substituents at position 2 of the indole were synthesized as anticancer agents. Several compounds inhibited tubulin polymerization at submicromolar concentration and inhibited cell growth at low nanomolar concentrations. Compounds 18 and 57 were superior to the previously synthesized 5. Compound 18 was exceptionally potent as an inhibitor of cell growth: it showed IC50 = 1.0 nM in MCF-7 cells, and it was uniformly active in the whole panel of cancer cells and superior to colchicine and combretastatin A-4. Compounds 18, 20, 55, and 57 were notably more potent than vinorelbine, vinblastine, and paclitaxel in the NCI/ADR-RES and Messa/Dx5 cell lines, which overexpress P-glycoprotein. Compounds 18 and 57 showed initial vascular disrupting effects in a tumor model of liver rhabdomyosarcomas at 15 mg/kg intravenous dosage. Derivative 18 showed water solubility and higher metabolic stability than 5 in human liver microsomes.


Asunto(s)
Antineoplásicos/síntesis química , Imidazoles/síntesis química , Indoles/síntesis química , Piridinas/síntesis química , Moduladores de Tubulina/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Células CACO-2 , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos del Citocromo P-450 , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Imidazoles/química , Imidazoles/farmacología , Indoles/química , Indoles/farmacología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/tratamiento farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microsomas Hepáticos/metabolismo , Mitosis/efectos de los fármacos , Permeabilidad , Polimerizacion , Piridinas/química , Piridinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Rabdomiosarcoma/irrigación sanguínea , Rabdomiosarcoma/tratamiento farmacológico , Solubilidad , Relación Estructura-Actividad , Tubulina (Proteína)/química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
12.
ChemMedChem ; 7(4): 709-21, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22354629

RESUMEN

A series of spiro[chromane-2,4'-piperidine] derivatives based on a previously published lead benzyl spirocycle 1 and bearing various N-aryl and N-alkylaryl substituents on the piperidine ring were prepared as novel histone deacetylase (HDAC) inhibitors. The compounds were evaluated for their abilities to inhibit nuclear HDACs, their in vitro antiproliferative activities, and in vitro ADME profiles. Based on these activities, 4-fluorobenzyl and 2-phenylethyl spirocycles were selected for further characterization. In vivo pharmacokinetic (PK) studies showed that both compounds exhibit an overall lower clearance rate, an increased half-life, and higher AUCs after intravenous and oral administration than spiropiperidine 1 under the conditions used. The improved PK behavior of these two compounds also correlated with superior in vivo antitumor activity in an HCT-116 xenograft model.


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Piperidinas/química , Administración Oral , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Proteínas Sanguíneas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos del Citocromo P-450 , Evaluación Preclínica de Medicamentos , Semivida , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/farmacocinética , Inyecciones Intravenosas , Tasa de Depuración Metabólica , Ratones , Ratones Desnudos , Estructura Molecular , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Med Chem ; 53(2): 822-39, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20017493

RESUMEN

The histone deacetylases (HDACs) are able to regulate gene expression, and histone deacetylase inhibitors (HDACi) emerged as a new class of agents in the treatment of cancer as well as other human disorders such as neurodegenerative diseases. In the present investigation, we report on the synthesis and biological evaluation of compounds derived from the expansion of a HDAC inhibitor scaffold having N-hydroxy-3-phenyl-2-propenamide and N-hydroxy-3-(pyridin-2-yl)-2-propenamide as core structures and containing a phenyloxopropenyl moiety, either unsubstituted or substituted by a 4-methylpiperazin-1-yl or 4-methylpiperazin-1-ylmethyl group. The compounds were evaluated for their ability to inhibit nuclear HDACs, as well as for their in vitro antiproliferative activity. Moreover, their metabolic stability in microsomes and aqueous solubility were studied and selected compounds were further characterized by in vivo pharmacokinetic experiments. These compounds showed a remarkable stability in vivo, compared to hydroxamic acid HDAC inhibitors that have already entered clinical trials. The representative compound 30b showed in vivo antitumor activity in a human colon carcinoma xenograft model.


Asunto(s)
Acrilamidas/síntesis química , Antineoplásicos/síntesis química , Inhibidores de Histona Desacetilasas/síntesis química , Acrilamidas/farmacología , Antineoplásicos/farmacocinética , Derivados del Benceno , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Estabilidad de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Células HeLa , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Piridinas , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Magn Reson Imaging ; 27(4): 872-80, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18383249

RESUMEN

PURPOSE: To investigate the potential value of MRI for noninvasive assessment of angiogenesis in a murine model exploiting the properties of two contrast agents, gadoteridol (ProHance) and gadocoletic acid trisodium salt (B22956/1). MATERIALS AND METHODS: Biocompatible sponges were implanted in both mice flanks. Stimulated sponges contained human recombinant basic fibroblast growth factor (bFGF) as the angiogenic agent; control sponges contained vehicle. Angiogenesis was evaluated by MRI after injection of extravascular (ProHance) or blood-pool (B22956/1) contrast agents at different times after sponge implantation. Sponges signal intensity enhancement was calculated both as the relative enhancement and the rate of relative enhancement. Results from MRI were validated by classic biochemical (hemoglobin level and protein content) and morphological (histology) assays. RESULTS: The intrinsic different properties of ProHance and B22956/1 in wash-in and wash-out kinetics were useful to detect progressive vascularization and the establishment of a functional vascular network in the implants. Moreover, MRI allowed the appreciation of differences in neovessel colonization between bFGF-treated sponges and controls. Hemoglobin level, protein content, and histology confirmed the sponge vascularization and MRI results. CONCLUSION: Contrast-enhanced MRI is a reliable tool to study vascular characteristics in animal models of angiogenesis. The different kinetic properties of contrast agents can provide evidence of different functional neovascularization aspects and levels.


Asunto(s)
Medios de Contraste , Modelos Animales de Enfermedad , Compuestos Heterocíclicos , Imagen por Resonancia Magnética , Neovascularización Patológica/diagnóstico , Compuestos Organometálicos , Animales , Materiales Biocompatibles , Factor 2 de Crecimiento de Fibroblastos , Gadolinio , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/patología , Tapones Quirúrgicos de Gaza
15.
Invest Radiol ; 40(7): 421-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15973133

RESUMEN

OBJECTIVES: The aim of this study was to compare the efficacy of gadoteridol, B22956/1 (a new protein binding blood pool contrast agent), and (Gd-DTPA)37-albumin in detecting, by dynamic contrast-enhanced magnetic resonance imaging (MRI), the effect in vivo of tamoxifen in an experimental model of breast tumor implanted in rats. MATERIALS AND METHODS: Tumors were induced by subcutaneous injection of 10 mammary adenocarcinoma cells (13762 MAT B III). Treatment with tamoxifen (or vehicle) started on day 4 after implantation. On day 10 after implantation, animals were observed by MRI using B22956/1 (or gadoteridol) and, 24 hours later, using (Gd-DTPA)37-albumin. RESULTS: Dynamic contrast-enhanced magnetic resonance imaging data showed that tamoxifen treatment decreased vascular permeability to B22956/1, whereas no difference was detectable in permeability to gadoteridol or to (Gd-DTPA)37-albumin. No effect on fractional plasma volume was detected with either of contrast agents. CONCLUSIONS: B22956/1 is superior to both small Gd chelates and macromolecular contrast agents in the assessment of the effect of tamoxifen treatment on tumor vasculature.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Antagonistas de Estrógenos/farmacología , Compuestos Heterocíclicos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética/métodos , Compuestos Organometálicos , Tamoxifeno/farmacología , Albúminas , Animales , Neoplasias de la Mama/irrigación sanguínea , Medios de Contraste , Antagonistas de Estrógenos/uso terapéutico , Femenino , Gadolinio , Gadolinio DTPA , Aumento de la Imagen , Neoplasias Mamarias Experimentales , Ratas , Tamoxifeno/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA