Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Prostate ; 82(5): 505-516, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35037287

RESUMEN

INTRODUCTION: Castration-resistant prostate cancer (CRPC) remains dependent on androgen receptor (AR) signalling, which is largely driven by conversion of adrenal androgen precursors lasting after castration. Abiraterone, an inhibitor of the steroidogenic enzyme CYP17A1, has been demonstrated to reduce adrenal androgen synthesis and prolong CRPC patient survival. To study mechanisms of resistance to castration and abiraterone, we created coculture models using human prostate and adrenal tumours. MATERIALS AND METHODS: Castration-naïve and CRPC clones of VCaP were incubated with steroid substrates or cocultured with human adrenal cells (H295R) and treated with abiraterone or the antiandrogen enzalutamide. Male mice bearing VCaP xenografts with and without concurrent H295R xenografts were castrated and treated with placebo or abiraterone. Response was assessed by tumour growth and PSA release. Plasma and tumour steroid levels were assessed by LC/MS-MS. Quantitative polymerase chain reaction determined steroidogenic enzyme, nuclear receptor and AR target gene expression. RESULTS: In vitro, adrenal androgens induced castration-naïve and CRPC cell growth, while precursors steroids for de novo synthesis did not. In a coculture system, abiraterone blocked H295R-induced growth of VCaP cells. In vivo, H295R promoted castration-resistant VCaP growth. Abiraterone only inhibited VCaP growth or PSA production in the presence of H295R. Plasma steroid levels demonstrated CYP17A1 inhibition by abiraterone, whilst CRPC tumour tissue steroid levels showed no evidence of de novo intratumoural androgen production. Castration-resistant and abiraterone-resistant VCaP tumours had increased levels of AR, AR variants and glucocorticoid receptor (GR) resulting in equal AR target gene expression levels compared to noncastrate tumours. CONCLUSIONS: In our model, ligand-dependent AR-regulated regrowth of CRPC was predominantly supported via adrenal androgen precursor production while there was no evidence for intratumoural androgen synthesis. Abiraterone-resistant tumours relied on AR overexpression, expression of ligand-independent AR variants and GR signalling.


Asunto(s)
Andrógenos , Neoplasias de la Próstata Resistentes a la Castración , Andrógenos/metabolismo , Androstenos/farmacología , Androstenos/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Ligandos , Masculino , Ratones , Nitrilos/uso terapéutico , Orquiectomía , Antígeno Prostático Específico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Glucocorticoides
2.
Breast Cancer Res Treat ; 134(1): 117-29, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22228402

RESUMEN

The P-glycoprotein (P-gp) is a 170-kDa protein that acts as an energy dependent, transmembrane efflux pump and is encoded by the MDR1 gene. It has been shown to be responsible for multidrug resistance (MDR) in a defined subpopulation of breast cancer patients and thus represents a molecular target for circumventing MDR in this tumor indication. MDR modulators have been developed and demonstrated high selectivity for P-gp with inhibitory activities in the low nanomolar range. Although some objective responses were achieved in clinical trials, combination therapy with these MDR modulators, such as Ca2+ antagonists caused unacceptable toxicity. Targeting P-gp inhibitors to the tumor site is a mean to increase their therapeutic index, and in this context binding of tailor-made prodrugs to circulating albumin is an established technology to reduce the toxicity and enhance the efficacy of anticancer drugs. In this study, we consequently developed an acid-sensitive albumin-binding prodrug of the P-gp inhibitor zosuquidar (LY335979) in a two-step synthesis using a maleimide hydrazone linker system established in our laboratory that first introduces acetylbenzoic acid at the HO-group of zosuquidar followed by derivatization with 6-maleimidocaproyl hydrazide to form the acid-sensitive hydrazone bond. The maleimide group enables the prodrug to bind rapidly and selectively to the cysteine-34 position of endogenous albumin after intravenous administration. HPLC analysis demonstrated rapid albumin binding of the zosuquidar prodrug as well as the quantitative release of the acetylbenzoic ester derivative of zosuquidar at pH 5.0. Subsequently, its ability to circumvent MDR was tested in two doxorubicin-resistant breast carcinoma cell lines (MCF-7/ADR and MT-3/ADR). The MDR status of these cell lines can be reversed by zosuquidar which was confirmed in a rhodamine 123 assay using fluorescence microscopy and FACS analysis. Furthermore, zosuquidar as well its acid-sensitive albumin conjugate re-sensitized cells to doxorubicin as well as to an albumin-binding prodrug of doxorubicin, i.e., the 6-maleimidocaproyl hydrazone derivative of doxorubicin, achieving IC50 values in the same order of magnitude as the parental cell lines. Thus, a novel formulation of zosuquidar has been developed that could have the potential to improve the toxicity issues and tumor targeting properties of the original compound.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Dibenzocicloheptenos/farmacología , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Hidrazonas/farmacología , Profármacos/farmacología , Quinolinas/farmacología , Línea Celular Tumoral/efectos de los fármacos , Portadores de Fármacos/farmacología , Diseño de Fármacos , Estabilidad de Medicamentos , Sinergismo Farmacológico , Femenino , Colorantes Fluorescentes/metabolismo , Humanos , Hidrólisis , Concentración 50 Inhibidora , Rodamina 123/metabolismo , Albúmina Sérica/farmacología
3.
Nat Methods ; 5(5): 439-45, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18391960

RESUMEN

The complexity of the angiogenic cascade limits cellular approaches to studying angiogenic endothelial cells (ECs). In turn, in vivo assays do not allow the analysis of the distinct cellular behavior of ECs during angiogenesis. Here we show that ECs can be grafted as spheroids into a matrix to give rise to a complex three-dimensional network of human neovessels in mice. The grafted vasculature matures and is connected to the mouse circulation. The assay is highly versatile and facilitates numerous applications including studies of the effects of different cytokines on angiogenesis. Modifications make it possible to study human lymphangiogenic processes in vivo. EC spheroids can also be coimplanted with other cell types for tissue engineering purposes.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales/citología , Neovascularización Fisiológica/fisiología , Esferoides Celulares/citología , Animales , Comunicación Celular , Células Endoteliales/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/farmacología
4.
Anal Biochem ; 406(2): 233-4, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20638357

RESUMEN

Here we describe an optimization of a nonradioactive immunosorbent p38alpha mitogen-activated protein kinase (MAPK) activity assay to determine inhibitory potency of small molecule inhibitors. The assay omits the secondary antibody and, therefore, is shorter, more accurate, and easier to handle (total assay time of 3 h). This direct assay uses a new monoclonal anti-phospho-ATF-2 (Thr69/71) peroxidase-conjugated antibody, increasing specificity and eliminating problems with cross-reactivities of secondary antibodies.


Asunto(s)
Pruebas de Enzimas/métodos , Inmunoadsorbentes/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Anticuerpos Monoclonales/inmunología , Humanos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Fosfotreonina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Radiactividad
5.
Prostate ; 69(11): 1151-63, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19399788

RESUMEN

BACKGROUND: Gemcitabine (Gemc) is an efficient chemotherapeutic drug in various cancer types (e.g., pancreas) but has only limited effects on hormone-refractory prostate cancer (HRPCa). Since HRPCa cells are highly sensitive to even low doses of Gemc in vitro, the lack of clinical effects might be due to rapid degradation of Gemc by deaminases combined with impaired accumulation in tumor tissue and PCa cells. Liposomal formulation (GemLip) is expected to protect the entrapped cytotoxic substance from enzymatic degradation and furthermore augment its accumulation within tumor tissues due to an enhanced permeability of the tumor vessels. METHODS: Anti-tumoral and anti-metastatic activity of GemLip and Gemc were investigated in two luciferase-expressing, human hormone-refractory PC-3 and Du145 HRPCa xenograft models in immunodeficient mice. Tumor growth was monitored by in vivo luminescence imaging (orthotopic) or callipering (subcutaneous). Anti-metastatic effects of treatment were determined by in vitro luciferase assay of the tissues. RESULTS: Tumor growth of subcutaneous Du145 xenografts was significantly inhibited only by GemLip (8 mg/kg: P = 0.014 and 6 mg/kg: P = 0.011) but not by conventional Gemc (360 mg/kg). In contrast, growth of orthotopic PC-3 xenografts was significantly inhibited by both, GemLip (P = 0.041) and Gemc (P = 0.002). The drugs furthermore strongly reduced spleen and liver metastases in this model. CONCLUSIONS: As shown by the very low efficient concentration of GemLip, liposomal entrapment of Gemc greatly enhances its activity. GemLip has, even at very low doses, a significant anti-tumoral and anti-metastatic therapeutic effect in HRPCa xenografts in vivo and was beneficial even when the conventional Gemc failed.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Neoplasias de la Próstata/tratamiento farmacológico , Trasplante Heterólogo , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Relación Dosis-Respuesta a Droga , Humanos , Liposomas , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/secundario , Luciferasas/metabolismo , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Neoplasias del Bazo/prevención & control , Neoplasias del Bazo/secundario , Gemcitabina
6.
J Cell Biol ; 167(6): 1037-50, 2004 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-15611331

RESUMEN

In Xenopus oocytes, the spindle assembly checkpoint (SAC) kinase Bub1 is required for cytostatic factor (CSF)-induced metaphase arrest in meiosis II. To investigate whether matured mouse oocytes are kept in metaphase by a SAC-mediated inhibition of the anaphase-promoting complex/cyclosome (APC/C) complex, we injected a dominant-negative Bub1 mutant (Bub1dn) into mouse oocytes undergoing meiosis in vitro. Passage through meiosis I was accelerated, but even though the SAC was disrupted, injected oocytes still arrested at metaphase II. Bub1dn-injected oocytes released from CSF and treated with nocodazole to disrupt the second meiotic spindle proceeded into interphase, whereas noninjected control oocytes remained arrested at metaphase. Similar results were obtained using dominant-negative forms of Mad2 and BubR1, as well as checkpoint resistant dominant APC/C activating forms of Cdc20. Thus, SAC proteins are required for checkpoint functions in meiosis I and II, but, in contrast to frog eggs, the SAC is not required for establishing or maintaining the CSF arrest in mouse oocytes.


Asunto(s)
Oocitos/citología , Oocitos/metabolismo , Proteínas Proto-Oncogénicas c-mos/fisiología , Huso Acromático/fisiología , Ciclosoma-Complejo Promotor de la Anafase , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Femenino , Proteínas Mad2 , Meiosis , Ratones , Ratones Endogámicos , Mutación , Nocodazol/farmacología , Proteínas Nucleares , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-mos/metabolismo , Huso Acromático/química , Huso Acromático/genética , Estroncio/farmacología , Complejos de Ubiquitina-Proteína Ligasa/antagonistas & inhibidores , Complejos de Ubiquitina-Proteína Ligasa/fisiología
7.
Bioorg Med Chem Lett ; 19(14): 3725-8, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19553109

RESUMEN

In this Letter we report the synthesis and in vitro studies of cleavable polymer-drug conjugates derived from dendritic polyglycerol and maleimide-bearing prodrugs of doxorubicin and methotrexate that are cleaved by cathepsin B. Cleavage properties and cytotoxicity of the new conjugates are presented.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/síntesis química , Glicerol/química , Polímeros/química , Profármacos/administración & dosificación , Secuencia de Aminoácidos , Antineoplásicos/química , Antineoplásicos/farmacología , Catepsina B/metabolismo , Línea Celular Tumoral , Dendrímeros/química , Doxorrubicina/química , Portadores de Fármacos/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Maleimidas/química , Metotrexato/química , Profármacos/química , Profármacos/farmacología
8.
Bioorg Med Chem Lett ; 19(3): 1030-4, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19097889

RESUMEN

In this paper new dendritic core-shell architectures with pH-labile linkers based on hyperbranched polyglycerol cores and biocompatible poly(ethylene glycol) shells were synthesized which encapsulate the anticancer agent doxorubicin and a dye for near-infrared imaging, an indotricarbocyanine. Acid-sensitive properties of the new nanocarriers and in vitro cytotoxicity of the doxorubicin-nanocarrier are presented as well as preliminary data regarding their toxicity and tumor targeting potential in nude mice.


Asunto(s)
Antineoplásicos/síntesis química , Carbocianinas/química , Células Dendríticas/metabolismo , Doxorrubicina/química , Portadores de Fármacos , Nanotecnología/métodos , Animales , Antineoplásicos/farmacología , Materiales Biocompatibles/química , Carbocianinas/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Glicerol/química , Concentración de Iones de Hidrógeno , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Nanopartículas/química , Polímeros/química
9.
Methods Mol Biol ; 1888: 1-20, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30519938

RESUMEN

PREDECT, a European IMI consortium, has assumed the task to generate robust 2D and 3D culture platforms. Protocols established for 2D and 3D monoculture and stromal coculture models of increasing complexity (spheroid, stirred-tank bioreactor, Matrigel- and collagen-embedded cultures) have been established between six laboratories within academia, biotech, and pharma. These models were tested using three tumor cell lines (MCF7, LNCaP, and NCI-H1437), covering three pathologies (breast, prostate, and lung), but should be readily transferable to other model systems. Fluorescent protein tagged cell lines were used for all platforms, allowing for online measurement of growth curves and drug responses to treatments. All methods, from culture setup to phenotypic characterization and gene expression profiling are described in this chapter.The adaptable methodologies and detailed protocols described here should help to include these models more readily to the drug discovery pipeline.


Asunto(s)
Técnicas de Cultivo de Célula , Reactores Biológicos , Línea Celular Tumoral , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Orden Génico , Genes Reporteros , Vectores Genéticos/genética , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía Fluorescente , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Programas Informáticos , Esferoides Celulares , Transducción Genética , Células Tumorales Cultivadas
10.
Int J Cancer ; 122(5): 1145-54, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17973264

RESUMEN

The prostate-specific antigen (PSA) is a serine protease that is over-expressed in prostate carcinoma and represents a molecular target for selectively releasing an anticancer agent from a prodrug formulation. We have recently investigated a macromolecular prodrug strategy for improved cancer chemotherapy based on 2 features: (i) rapid and selective binding of thiol-reactive prodrugs to the cysteine-34 position of endogenous albumin after intravenous administration, and (ii) enzymatic release of the albumin-bound drug at the tumor site (Mansour et al., Cancer Res 2003, 63, 4062-4066). In this work, we describe an albumin-binding prodrug, EMC-Arg-Ser-Ser-Tyr-Tyr--Ser-Arg-DOXO [EMC: epsilon-Maleimidocaproic acid; DOXO = doxorubicin; X = amino acid] that is cleaved by PSA. Because of the incorporation of 2 arginine residues, the prodrug exhibited excellent water-solubility and was rapidly and selectively bound to endogenous albumin. Incubation studies with PSA and tumor homogenates from PSA-positive tumors (LNCaP) demonstrated that the albumin-bound form of the prodrug was efficiently cleaved by PSA at the P(1)-P' (1) scissile bond releasing the doxorubicin dipeptide H-Ser-Arg-DOXO, which was further degraded to doxorubicin as the final cleavage product. In cell culture experiments, the prodrug was approximately 100-fold less active against LNCaP cells than the free drug. In contrast, in a mouse model of human prostate cancer using luciferase transduced LNCaP cells orthotopically implanted in SCID mice, the prodrug showed enhanced antitumor efficacy when compared to doxorubicin. Doxorubicin treatment at a dose of 2 x 4 mg/kg caused significant weight loss and mortality (-25%), and did not result in a significant antitumor response at the end of the experiment. The prodrug at 3 x 12 mg/kg doxorubicin equivalents, however, was well tolerated and induced a significant reduction in tumor size of 62% (+/-25%, **p = 0.003) as well as a decrease of the metastatic burden in the lungs as detected in luciferase assays (-50%, SD +/- 115%, *p = 0.038).


Asunto(s)
Albúminas/uso terapéutico , Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Profármacos/síntesis química , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Albúminas/metabolismo , Animales , Antibióticos Antineoplásicos/metabolismo , Modelos Animales de Enfermedad , Doxorrubicina/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Profármacos/metabolismo , Profármacos/uso terapéutico , Neoplasias de la Próstata/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Data ; 4: 170170, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29160867

RESUMEN

Two-dimensional (2D) culture of cancer cells in vitro does not recapitulate the three-dimensional (3D) architecture, heterogeneity and complexity of human tumors. More representative models are required that better reflect key aspects of tumor biology. These are essential studies of cancer biology and immunology as well as for target validation and drug discovery. The Innovative Medicines Initiative (IMI) consortium PREDECT (www.predect.eu) characterized in vitro models of three solid tumor types with the goal to capture elements of tumor complexity and heterogeneity. 2D culture and 3D mono- and stromal co-cultures of increasing complexity, and precision-cut tumor slice models were established. Robust protocols for the generation of these platforms are described. Tissue microarrays were prepared from all the models, permitting immunohistochemical analysis of individual cells, capturing heterogeneity. 3D cultures were also characterized using image analysis. Detailed step-by-step protocols, exemplary datasets from the 2D, 3D, and slice models, and refined analytical methods were established and are presented.


Asunto(s)
Modelos Biológicos , Neoplasias , Técnicas de Cultivo de Célula , Humanos , Imagenología Tridimensional
13.
PLoS One ; 11(6): e0156942, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27303813

RESUMEN

In oncology, two-dimensional in-vitro culture models are the standard test beds for the discovery and development of cancer treatments, but in the last decades, evidence emerged that such models have low predictive value for clinical efficacy. Therefore they are increasingly complemented by more physiologically relevant 3D models, such as spheroid micro-tumor cultures. If suitable fluorescent labels are applied, confocal 3D image stacks can characterize the structure of such volumetric cultures and, for example, cell proliferation. However, several issues hamper accurate analysis. In particular, signal attenuation within the tissue of the spheroids prevents the acquisition of a complete image for spheroids over 100 micrometers in diameter. And quantitative analysis of large 3D image data sets is challenging, creating a need for methods which can be applied to large-scale experiments and account for impeding factors. We present a robust, computationally inexpensive 2.5D method for the segmentation of spheroid cultures and for counting proliferating cells within them. The spheroids are assumed to be approximately ellipsoid in shape. They are identified from information present in the Maximum Intensity Projection (MIP) and the corresponding height view, also known as Z-buffer. It alerts the user when potential bias-introducing factors cannot be compensated for and includes a compensation for signal attenuation.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Imagenología Tridimensional/métodos , Luz , Microscopía Confocal/métodos , Esferoides Celulares/citología , Algoritmos , Fibroblastos Asociados al Cáncer/citología , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Simulación por Computador , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Modelos Biológicos , Reproducibilidad de los Resultados , Microambiente Tumoral
14.
Cancer Chemother Pharmacol ; 78(2): 405-17, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27349901

RESUMEN

PURPOSE: This phase I study investigated the maximum tolerated dose (MTD), safety, pharmacokinetics, pharmacodynamics, and antitumor activity of the Aurora B kinase inhibitor BI 811283 in patients with advanced solid tumors. METHODS: BI 811283 was administered via 24-h infusion on Days 1 and 15 of a 4-week cycle (schedule A) or Day 1 of a 3-week cycle (schedule B) in a modified 3 + 3 dose-escalation design. Pharmacodynamic assessments included immunohistochemistry for phosphorylated histone H3 (pHH3) on skin biopsies to determine Aurora B kinase inhibition and plasma concentrations of caspase-cleaved CK-18 (apoptosis marker). RESULTS: A total of 121 patients were treated. The MTDs of BI 811283 were 125 mg (schedule A) and 230 mg (schedule B). Dose-limiting toxicities were primarily hematological (febrile neutropenia and grade 4 neutropenia); the most common drug-related adverse effects included neutropenia, fatigue, leukopenia, nausea, alopecia, diarrhea, and decreased appetite. A trend toward a decrease in pHH3 was observed, with increasing BI 811283 doses, indicating target engagement; there was no consistent trend regarding caspase-cleaved CK-18 levels. No objective response was observed although 19 patients in each schedule achieved clinical benefit (stable disease). CONCLUSIONS: BI 811283 demonstrated a generally manageable safety profile and disease stabilization in some patients. TRIAL REGISTRATION: EudraCT No: 2007-000191-17, ClinicalTrials.gov Identifier: NCT00701324.


Asunto(s)
Antineoplásicos/administración & dosificación , Aurora Quinasa B/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunohistoquímica , Queratina-18/sangre , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Resultado del Tratamiento , Adulto Joven
15.
Sci Rep ; 6: 28951, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27364600

RESUMEN

Two-dimensional (2D) cell cultures growing on plastic do not recapitulate the three dimensional (3D) architecture and complexity of human tumors. More representative models are required for drug discovery and validation. Here, 2D culture and 3D mono- and stromal co-culture models of increasing complexity have been established and cross-comparisons made using three standard cell carcinoma lines: MCF7, LNCaP, NCI-H1437. Fluorescence-based growth curves, 3D image analysis, immunohistochemistry and treatment responses showed that end points differed according to cell type, stromal co-culture and culture format. The adaptable methodologies described here should guide the choice of appropriate simple and complex in vitro models.


Asunto(s)
Técnicas de Cocultivo/métodos , Esferoides Celulares/citología , Línea Celular Tumoral , Humanos , Imagenología Tridimensional , Células MCF-7 , Células del Estroma/citología
16.
Biotechnol J ; 9(9): 1115-28, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25174503

RESUMEN

Cancers are complex and heterogeneous pathological "organs" in a dynamic interplay with their host. Models of human cancer in vitro, used in cancer biology and drug discovery, are generally highly reductionist. These cancer models do not incorporate complexity or heterogeneity. This raises the question as to whether the cancer models' biochemical circuitry (not their genome) represents, with sufficient fidelity, a tumor in situ. Around 95% of new anticancer drugs eventually fail in clinical trial, despite robust indications of activity in existing in vitro pre-clinical models. Innovative models are required that better capture tumor biology. An important feature of all tissues, and tumors, is that cells grow in three dimensions. Advances in generating and characterizing simple and complex (with added stromal components) three-dimensional in vitro models (3D models) are reviewed in this article. The application of stirred bioreactors to permit both scale-up/scale-down of these cancer models and, importantly, methods to permit controlled changes in environment (pH, nutrients, and oxygen) are also described. The challenges of generating thin tumor slices, their utility, and potential advantages and disadvantages are discussed. These in vitro/ex vivo models represent a distinct move to capture the realities of tumor biology in situ, but significant characterization work still remains to be done in order to show that their biochemical circuitry accurately reflects that of a tumor.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Técnicas In Vitro/métodos , Neoplasias/patología , Animales , Reactores Biológicos , Biología Celular , Humanos , Modelos Biológicos
17.
J Biomol Screen ; 19(10): 1350-61, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25260782

RESUMEN

Many receptor tyrosine kinases (RTKs) represent bona fide drug targets in oncology. Effective compounds are available, but treatment invariably leads to resistance, often due to RTK mutations. The discovery of second-generation inhibitors requires cellular models of resistant RTKs. An approach using artificial transmembrane domains (TMDs) to activate RTKs was explored for the rapid generation of simple, ligand-independent cellular RTK assays, including resistance mutants. The RTKs epidermal growth factor receptor (EGFR), MET, and KIT were chosen in a proof-of-concept study. Their intracellular domains were inserted into a series of expression vectors encoding artificial TMDs, and they were tested for autophosphorylation activity in transient transfection assays. Active constructs could be identified for MET and EGFR, but not for KIT. Rat1 cell pools were generated expressing the MET or EGFR constructs, and their sensitivity to reference tool compounds was compared to that of MKN-45 or A431 cells. A good correlation between natural and recombinant cells led us to build a panel of clinically relevant MET mutant cell pools, based on the wild-type construct, which were then profiled via MET autophosphorylation and soft agar assays. In summary, a platform was established that allows for the rapid generation of cellular models for RTKs and their resistance mutants.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Terapia Molecular Dirigida , Mutación , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Recombinantes/genética , Transfección
18.
J Pharm Biomed Anal ; 66: 349-51, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22465233

RESUMEN

The p38 mitogen activated protein kinase (MAPK) has emerged as a target for treating inflammatory diseases, like rheumatoid arthritis (RA). Expression of p38δ is induced in rheumatoid arthritis synovial fibroblasts (RASFs) by a cytokine-independent pathway substantially different from other MAPK pathways. To identify inhibitors of p38δ MAPK, we developed a direct ELISA assay based on a previously described p38α assay for monitoring the phosphorylation of ATF-2. This work presents a straightforward assay for evaluating the potency of small-molecule inhibitors. To validate the assay under optimized conditions, we used reference compounds and achieved results comparable to published data.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Proteína Quinasa 13 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Factor de Transcripción Activador 2/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/fisiopatología , Fibroblastos/metabolismo , Fosforilación
19.
Anticancer Res ; 32(7): 2399-406, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22753696

RESUMEN

BACKGROUND/AIM: Sequential treatment with targeted agents is standard of care for patients with metastatic renal cell carcinoma (mRCC). However, clinical data directly comparing treatment outcomes with a mammalian target of rapamycin inhibitor or a vascular endothelial growth factor-targeted agent in the second-line setting are lacking. We evaluated sequential treatment in a syngeneic, orthotopic mouse model of mRCC. MATERIALS AND METHODS: BALB/c mice were orthotopically implanted with murine RCC (RENCA) cells expressing luciferase and randomized to vehicle, sunitinib, sunitinib followed by sorafenib, or sunitinib followed by everolimus. Tumor growth and metastases were assessed by in vivo (whole body) and ex vivo (primary tumor, lung, liver) luciferase activity and necropsies, performed on day 20 or 46 for vehicle and treatment groups, respectively. RESULTS: Sunitinib followed by everolimus was associated with reduced luciferase activity and primary tumor weight and volume compared with sunitinib, and sunitinib followed by sorafenib. CONCLUSION: Sequential therapy with sunitinib followed by everolimus demonstrated significant antitumor and anti-metastatic effects.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Esquema de Medicación , Everolimus , Femenino , Indoles/administración & dosificación , Indoles/efectos adversos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Luciferasas/biosíntesis , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos BALB C , Pirroles/administración & dosificación , Pirroles/efectos adversos , Distribución Aleatoria , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Sirolimus/análogos & derivados , Sunitinib
20.
J Biomol Screen ; 17(3): 339-49, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22002421

RESUMEN

The insulin-like growth factor I receptor (IGF1-R) system has long been implicated in cancer and is a promising target for tumor therapy. Besides in vitro screening assays, the discovery of specific inhibitors against IGF-1R requires relevant cellular models, ideally applicable to both in vitro and in vivo studies. With this aim in mind, the authors generated an inducible cell line using the tetracycline-responsive gene expression system to mimic the effects of therapeutic inhibition of the IGF-1R both in vitro and on established tumors in vivo. Inducible overexpression of IGF-1R in murine embryonic fibroblasts was achieved and resulted in the transformation of the cells as verified by their ability to grow in soft agar and in nude mice. Continuous repression of exogenous IGF-1R expression completely prevented outgrowth of the tumors. Furthermore, induced repression of IGF-1R expression in established tumors resulted in regression of the tumors. Interestingly, however, IGF-1R-independent relapse of tumor growth was observed upon prolonged IGF-1R repression. The IGF-1R cell line generated using this approach was successfully employed to test reference small-molecule inhibitors in vitro and an IGF-1R-specific inhibitory antibody, EM164, in vivo. Besides efficacy as a read-out, phospho-AKT could be identified as a pharmacodynamic biomarker, establishing this cell line as a valuable tool for the preclinical development of IGF-1R inhibitors.


Asunto(s)
Línea Celular Transformada , Doxiciclina/farmacología , Evaluación Preclínica de Medicamentos/métodos , Receptor IGF Tipo 1/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica , Fibroblastos , Ratones , Ratones Desnudos , Receptor IGF Tipo 1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA